Skip to main content
Oxford University Press - PMC COVID-19 Collection logoLink to Oxford University Press - PMC COVID-19 Collection
. 2019 Jun 14;59(3):211–246. doi: 10.1093/ilar/ily026

Immune Relevant and Immune Deficient Mice: Options and Opportunities in Translational Research

Enrico Radaelli 2,, Sara F Santagostino 3, Rani S Sellers 4, Cory F Brayton 5
PMCID: PMC7114723  PMID: 31197363

Abstract

In 1989 ILAR published a list and description of immunodeficient rodents used in research. Since then, advances in understanding of molecular mechanisms; recognition of genetic, epigenetic microbial, and other influences on immunity; and capabilities in manipulating genomes and microbiomes have increased options and opportunities for selecting mice and designing studies to answer important mechanistic and therapeutic questions. Despite numerous scientific breakthroughs that have benefitted from research in mice, there is debate about the relevance and predictive or translational value of research in mice. Reproducibility of results obtained from mice and other research models also is a well-publicized concern. This review summarizes resources to inform the selection and use of immune relevant mouse strains and stocks, aiming to improve the utility, validity, and reproducibility of research in mice. Immune sufficient genetic variations, immune relevant spontaneous mutations, immunodeficient and autoimmune phenotypes, and selected induced conditions are emphasized.

Keywords: biomedical research, experimental conditions, genetic background, genetic variation, immune system diseases, inbred strains, mice

Introduction

The main advantages of using mice in research include (1) their small size and very prolific nature, (2) the numerous commonalities existing between mice and humans in terms of physiology and pathobiology, (3) the well-characterized genomes and immune responses, and (4) the availability of advanced technologies for genetic and other experimental manipulations.1,2 Despite the advantages, there is ongoing controversy surrounding the reproducibility and translatability of mouse models of disease.35 Given the immune diversity within the human population, a perfect model relevant to all humans may be neither an achievable nor a reasonable expectation. However, it is possible to strive for relevant and reproducible translational models and to expect experimental designs to address specific research questions. Criticisms of mouse models (mouse blaming) are not always justified. Many factors contribute to study outcomes and reproducibility. These include genetic diversity; microbial, husbandry, and other environmental factors; experimental interventions; etc.2 Increasing the awareness of the immunobiological variations among inbred mice and their substrains as well as other factors that may impact immune responses in mice will help improve both the validity and reproducibility of mouse-based research. Attention to these aspects is warranted in experimental design, data interpretations, and reporting of research on immunity, disease, and therapeutic interventions.6,7

This review aims to provide a useful compendium of resources and references for those investigators who seek to familiarize themselves with key concepts of mouse immunology and translate those notions into the experimental setting. The most relevant sources of immune diversity of the laboratory mouse are here emphasized with a focus on immune sufficient genetic variations, immunologically relevant spontaneous mutations, autoimmune phenotypes, and selected induced immune deficiencies.

Mouse Nomenclature

Accurate mouse nomenclature is mission critical to scientific communication.810 Nomenclature “rules” for mice genes, strains, and substrains were recommended by scientists to the scientific community in the 1940s and 1950s. The first committees on standardized genetics nomenclature11 and on standardized strain nomenclature for mice12 included Nobel laureate George Snell. Early publications provided guidelines for gene and strain nomenclature, a list (database) of strains and substrains, and a list (database) of abbreviations for the researchers or institutions maintaining the mice.12 The list of abbreviations became the “laboratory codes” (lab codes) that are currently curated by ILAR (http://dels.nas.edu/global/ilar/lab-codes) and are available to producers and researchers at no charge. The lab code identifies the mouse source and becomes part of its name. The 1963 revision includes a listing of named genes, including histocompatibility alleles for many of the common strains. Subsequent committees updated the guidelines and included lists of inbred strains, substrains, and known genetic variants.1319 These publications are enlightening regarding the history and research use of contemporary mouse strains. They indicate recognition by the scientific community of the research implications of genetic and phenotypic variations, and reflect scientists’ concerns for accurate communication in published research. In 1972, a recommendation was published for standardized nomenclature for outbred stocks of laboratory animals of various species.20 These recommendations gained traction for mice and rats, but far less for other species. Current gene nomenclature “rules” for mice (International Committee on Standardized Genetic Nomenclature for Mice: http://www.informatics.jax.org/mgihome/nomen/strains.shtml), rats (Rat Gene Nomenclature Committee: https://rgd.mcw.edu/nomen/nomen.shtml), and human genes (HUGO Gene Nomenclature Committee: http://www.genenames.org/) are available online. Guidance for mouse strains, genes, alleles/mutations as well as tutorials and assistance can be accessed from Mouse Genome Informatics Nomenclature sites (http://www.informatics.jax.org/mgihome/nomen/gene.shtml). Recommendations for reporting animal research include correct nomenclature because it communicates key research-relevant elements of the strain or substrain history and genetics, genetic modifications, backcrossing or intercrossing, and other information.2123

Inbred Mouse Strains: Immune Relevant Genotypes and Phenotypes

The immune sufficient common inbred mouse strains are genetically well characterized, with genome projects on more than 30 strains.24,25 Divergent susceptibilities of inbred strains to infections, diseases, and tumor rejection were recognized early in strain development. Characterization of these variations has exposed research-relevant Th1 or Th2 biases, diversity in major histocompatibility complex (MHC) haplotypes, natural killer (NK) cell repertoires, hemolytic complement (complement component 5 or C5) activity, and toll-like receptor (TLR) function, among others.7,26,27 Table 1 and Supplementary Table 1 summarize some of the well-characterized immune relevant variations among immune sufficient common inbred mouse strains. Investigations on how penetrance and expressivity of immune phenotypes vary across different genetic backgrounds have enabled the discovery of key strain-related genetic modifiers that specifically enhance or suppress the manifestation of immunological disorders. This genetic source of diversity can be ultimately ascribed to a number of possible genetic alterations/variations including polymorphic alleles, unique quantitative trait loci (QTL) intervals, or specific haplotypes.2836 The influence of the inbred genetic background pervades many if not all the experimental contexts considered in this review.

Table 1.

Selected Immune Relevant Genetic Variations in Common Inbred Mouse Strains

Gene symbol
Mouse strain Ahr Ctse Hc Il2 Il12b Mx1 Mx2 Naip5 Nlrp Nlrp12 Oas1b Sirpa Slamf Slc11a1 Tcrb-v8 Tlr4 TH-bias
A/J b-2 N/A Hc 0 N/A N/A Ø S R N/A Ø N/A N/A R N/A N 2
AKR/J d N/A Hc 0 N/A N/A Ø N/A R N/A Ø N/A N/A R N/A N 1
BALB/c b N N N/A N/A Ø R S N/A Ø L29V 2 S N N 2
CBA b-2 N/A N/A N/A N/A Ø N/A S N/A Ø N/A N/A R N/A N 1
C3H/HeJ b-2 N N N/A N/A Ø N/A S N/A Ø N/A N/A R N/A Lps-d 1
C3H/HeN b-2 N/A N N/A N/A Ø N/A N/A N/A Ø N/A 1 R N/A N N/A
C57BL/6 b-1 Ø N N N Ø R R V Ø N 1 S N N 1
C57BL/10ScCr N/A N/A N N/A N Ø N/A N/A N/A Ø N/A N/A S N/A Lps-del 1
DBA/1J b N/A N N/A N/A Ø N/A N/A N/A Ø N/A N/A S N/A N 1
DBA/2J d N/A Hc 0 N/A N/A Ø N/A R N/A Ø N/A 2 R N/A N 2
FVB/N FVB/NJ N/A N/A Hc 0 N/A N/A Ø N/A S N/A Ø N/A N/A N/A Ø N N/A
MRL/MpJ N/A N/A N m1 N/A Ø N/A N/A N/A Ø N/A 2 N/A N/A N N/A
NOD/ShiLtJ N/A N/A Hc 0 m1 N/A Ø N/A R N/A Ø S 2 R N/A N N/A
NZB d N/A N N/A N/A Ø N/A N/A N/A Ø N/A 2 R N/A N N/A
NZW N/A N/A N N/A N/A Ø N/A N/A N/A Ø N/A 2 S N/A N N/A
NZM2410 N/A N/A N N/A N/A Ø N/A N/A N/A Ø N/A 2 N/A N/A N N/A
SJL/J d N/A N m1 P Ø N/A N/A N/A Ø N/A N/A R Ø N 1
SWR d N/A Hc 0 N/A N/A Ø N/A S N/A Ø N/A N/A R Ø N N/A
129 d N N N/A N/A Ø R S N/A Ø N/A N/A R N N 1

Ahr (aryl hydrocarbon receptor) activates expression of phase I and II metabolizing enzymes (e.g., Cyp450) and is important in cellular growth and differentiation; b1, b2 and b3 alleles are considered metabolically responsive alleles not linked to autoimmunity whereas d alleles are metabolically nonresponsive and associated with autoimmune susceptibility.3740

Ctse (cathepsin E) plays a role in antigen processing for MHC class II.41

Hc (hemolytic complement) plays a role in innate immune responses; Hc0 mice are null for this allele.42,43

Il2 (interleukin 2) is a key immune signaling cytokine; Il2m1 allele has a hypoactive polymorphism in the Il2 gene.44

Il12b (interleukin 12b) polymorphisms (P) have been associated with autoimmune disorders in humans.4547

Mx1 and Mx2 (MX dynamin-like GTPase 1 & 2) play a role in viral resistance; in most inbred mouse strains, these are not expressed.48,49

Naip5 (NLR family, apoptosis inhibitory protein 5) plays a key role in early innate immune responses mediated by the inflammasome; allelic polymorphism determines susceptibility to intracellular bacteria (Naip5Lgn1s = sensitive, Naip5Lgn1r = resistant).5052

Nlrp (nucleotide-binding oligomerization domain-like receptors aka NOD-like receptor proteins) has a key role in pathogen-associated molecular patterns detection.53

Nlrp12 (NACHT, LRR and PYD domains-containing protein 12) has an important role in inflammasome and activation of caspase 1; it also controls neutrophil chemotaxis in response to bacterial invasion.5456

Oas1b (2’-5’ oligoA synthetase family 1b) plays a role in innate immunity to eliminate viral RNA; most inbred mouse strains carry the susceptibility allele that encodes for a nonfunctional protein.57

Sirpa (signal-regulatory protein alpha); in BALB/c mice it has a single polymorphism in the IgV domain (L29V), which enhances binding to human CD47, decreasing macrophage phagocytosis; in NOD mice, the increased affinity for human CD47 is driven by a deletion of 2 amino acids in domain 1.58,59

Slamf [signaling lymphocytic activation molecule (SLAM) family] plays a role in self-tolerance;60 haplotype 2 is associated with autoimmune susceptibility.6163

Slc11a1 [solute carrier family 11 (proton-coupled divalent metal ion transporters), member 1] transporter that regulates iron homeostasis and impacts on the ability to control intracellular pathogens by phagocytes.64

Tcrb-V8 (T cell receptor beta, variable 8) plays a role in auto-immune disease susceptibility; in some strains, this is not expressed and is associated with increased susceptibility to autoimmune disease.6567

Tlr4 (Toll-like receptor 4) has a role in innate immune responses, in particular responses to LPS;6870 the mutant alleles Tlr4Lps-dand Tlr4Lps-del are not functional.

Th-bias; mice have TH-1 and TH-2 biases in their immune responses.71,72

N/A, no data; N, wild type (normal); NOD, nonobese diabetic; Ø, not expressed nonfunctional or hypofunctional gene product; P, polymorphism; R, resistance polymorphism; S, sensitive polymorphism; V, variable.

Immune Relevant Variations Among Substrains

Substrains with quite similar names harbor important genetic (and other) variations that are increasingly recognized.55,7476 C57BL/6N and C57BL/6J substrains diverged in 1951, so acquisition of mutations among colonies inbreeding at different sites is unsurprising. As illustrated in Table 2, some immune relevant genetic variations among C57BL/6 substrains include a Nlrp12 mutation in C57BL/6J mice and a Dock2 mutation in C57BL/6NHsd mice from certain colonies.55,77 The Nlrp12 gene primarily controls neutrophil chemotaxis in response to bacterial invasion. C57BL/6J mice carry a missense, loss of function mutation (Nlrp12C57BL/6J) and are more susceptible to certain bacterial infections compared with other C57BL/6 substrains harboring the wild-type Nlrp12 allele.55,73 More concerning may be when mutations arise within a substrain (of the same name) with colonies maintained at different sites. The Dock2Hsd mutation was revealed when reduced splenic marginal zone B cells and increased numbers of CD8+ T cells were identified in C57BL/6NHsd (and derived mutant mice) relative to other C57BL/6N mice.7779 Subsequently, Envigo tested their mice and reported that this mutation (Dock2Hsd) was present in 6 of their 19 C57BL/6NHsd colonies (http://www.envigo.com/assets/docs/c57-customer-communication-2-final-9jun16.pdf). Many research programs maintain in-house colonies of genetically engineered animals and “wild-type” background strains that warrant genetic quality assurance (QA) testing and breeding strategies to minimize effects of random mutations and genetic drift. (https://www.jax.org/jax-mice-and-services/customer-support/technical-support/breeding-and-husbandry-support/colony-planning; https://www.taconic.com/quality/genetic-integrity/colony-management/).

Table 2.

A Few B6 Substrains and Genetic Variations

B6 Substrain Source Dock2 Nlrp12 Nnt Snca Mmrn1 Crb1
J C57BL/6J Jackson N Ø Ø N N N
C57BL/6J a Charles River N/A N/A Ø N N N
C57BL/6JOlaHsd Hsd/Envigo N/A N/A N Ø Ø N
C57BL/6JRccHsd Hsd/Envigo N/A N/A N N N N
C57BL/6JBomTac Taconic N/A N/A N N N N
C57BL/6JRj Janvier N/A N/A N/A N/A N/A N/A
N C57BL/6ByJ Jackson N N/A N N N Ø
C57BL/6NHsd Hsd/Envigo Some Ø N/A N N N Ø
C57BL/6NRj Janvier N N/A N/A N/A N/A N/A
C57BL/6NCrl Charles River N N/A N N N Ø
C57BL/6NTac Taconic N N/A N N N Ø
C57BL/6NCr NCI N/A N N/A N/A N/A N/A
References 7779 55,73 80,81 8284 8284 85,86

Dock2 = the protein encoded by this gene belongs to the CDM protein family. It is specifically expressed in hematopoietic cells and is predominantly expressed in peripheral blood leukocytes. The protein is involved in remodeling of the actin cytoskeleton required for lymphocyte migration in response to chemokine signaling. It activates members of the Rho family of GTPases, for example RAC1 and RAC2, by acting as a guanine nucleotide exchange factor (GEF) to exchange bound GDP for free GTP.

Nlrp12 = This gene encodes a member of the CATERPILLER family of cytoplasmic proteins. The encoded protein, which contains an N-terminal pyrin domain, a NACHT domain, a NACHT-associated domain, and a C-terminus leucine-rich repeat region, has an important role in inflammasome and activation of caspase 1, it also controls neutrophil chemotaxis in response to bacterial invasion.

Nnt = nicotinamide nucleotide transhydrogenase; this gene encodes an integral protein of the inner mitochondrial membrane. The enzyme couples hydride transfer between NAD(H) and NADP(+) to proton translocation across the inner mitochondrial membrane.

Snca = alpha synuclein; one in a family of structurally related proteins that are prominently expressed in the brain, particularly in areas associated with learning and adaption. The exact function of alpha synuclein is not yet known.

Mmrn1 = multimerin 1; multimerin 1 is a stored platelet and endothelial cell adhesive protein that shows significant conservation. In vitro, multimerin 1 supports platelet adhesion and it also binds to collagen and enhances von Willebrand factor-dependent platelet adhesion to collagen.

Crb1 = retinal degeneration 8; the rd-8 mutation is due to a single base pair mutation in the CRB1 gene. This gene when mutated in humans is linked to macular degeneration and other age-related vision loss. Mice with this mutation are nearly blind by the time they are 8 weeks of age.

N/A, no data; N, wild type (normal); Ø, not expressed, nonfunctional or hypofunctional gene product.

aJ mice distributed by Charles River in EU

Influence of Genetic Background

Influences of background strain(s) warrant consideration when working with spontaneous or genetically engineered mutations. Many genetically engineered mice (GEM) have mixed or undefined genetic backgrounds that can affect research results. When spontaneous or experimentally induced mutations are transferred congenically from the line of origin onto a different (generally inbred) background strain, penetrance and expressivity of the phenotype may be positively or negatively affected by the recipient genome as well as by remnants of the “donor” genome (i.e., chromosomal regions flanking the mutant allele included in the congenic interval).8790

In immunodeficient strains, genetic and phenotypic contributions from background strains have research implications that may not be well known to those who are new to working with these mice. An internet search for commercially available immunodeficient mice bearing the Prkdcscid (scid) or Foxn1nu (nude or nu) mutations returns more than 20 strains of each on inbred or non-inbred backgrounds, some with quite similar names but with immune variations relevant to their genetic backgrounds (and with quite different costs that can influence purchasing decisions).9193 Variation in “leakiness” in scid mice on different genetic backgrounds is a well-known example. Leakiness refers to the tendency of scid mice to produce some functional B and T cells as they age and are increasingly exposed to environmental antigenic stimuli. Under similar experimental conditions, leakiness is greater on the C57BL/6 and BALB/c backgrounds, low on the C3H/HeJ background, and very low on the nonobese diabetic (NOD) background.93 Genetic factors contributing to “less sensitivity” to antigenic stimuli (and therefore less leakiness) include TRL4 deficiency in the C3H/HeJ mouse and impaired MHC-dependent antigen presentation in the NOD/ShiLtJ mouse.94,95 Especially relevant to human xenografts, NOD mice possess a unique signal-regulatory protein alpha (Sirpa) polymorphism with higher affinity for the human CD47 that results in a sustained “don’t-eat-me” signal and improves engraftment of human cells in NOD-scid and NOD-scid-derived mice.58

Autoimmune-susceptible strains develop spontaneous autoimmune disorders such as immune-mediated (Type 1-like) diabetes and systemic lupus erythematosus (SLE)-like conditions. The proclivity to develop experimentally induced autoimmune conditions, such as experimental autoimmune encephalitis (EAE) and collagen-induced arthritis (CIA), is also greatly influenced by the mouse’s genetic background.31,9699 The NOD mouse model for Type 1 diabetes (T1D) (e.g., NOD/ShiLtJ and NOD/MrkTac mice) is characterized by the development of a T cell-mediated immune response to pancreatic islet proteins (including insulin and chromogranin) similar to humans with T1D.100102 Their diabetic phenotype is polygenic with a significant contribution, as in humans, by their MHC polymorphisms.44,103105 NOD mice have a unique MHC class II lacking expression of I-Eα and I-E surface protein, and expressing I-Ag7MHC class II allele that is structurally and functionally similar to the human T1D susceptibility allele, DQ8.106,107 Other contributors to the autoimmune phenotype include a hypoactive variant of their IL-2 gene (Il2m1), Sirpa and Cd93 polymorphisms, lack of C5 (conferred by homozygosity for Hc0), and absence of complement factor H-related protein C (CFHR-C).43,44,105,108110 Genetic and phenotypic variations among the NOD substrains have been identified.111

Spontaneous lupus-like conditions in mice are associated with mutations such as Faslpr and Yaa and are influenced by genetic background.2836 Inbred strains that spontaneously develop lupus-like conditions include MRL/MpJ, BXSB/MpJ, NZB, NZW, NZBWF1 (aka NZB/W), NZM2410, and Palmerston North (PN/nBSwUmabJ).112,63,113,114

MRL/MpJ inbred mice are autoimmune prone and spontaneously develop an autoimmune phenotype as they age. A spontaneous mutation in the Faslpr gene in this strain resulted in the substrain MRL/MpJ-Faslpr, which develops signs of autoimmunity much earlier in life than the parent MRL/MpJ strain.115118 MRL/MpJ-Faslpr mice have a short lifespan (>50% mortality by 6 months old). They develop lymphoproliferative disease, immune complex glomerulonephritis, lupus-like skin disease, arthritis, and vasculitis.115,120123 It has been demonstrated that onset and severity of symptoms associated with the Faslpr mutation is strain dependent. For example, the Faslpr mutation results in a lymphoproliferative disease that on MRL/MpJ background is more severe than on the C57BL/6J background, but less severe than on the C3H/HeJ background.28,29,31,124 In contrast, immune complex pathologies including glomerulonephritis, vasculitis, and arthritis are more severe and initiate earlier with the Faslpr mutation on the MRL/MpJ background than on either the C57BL/6J or the C3H/HeJ background. Predisposition to the development of autoimmune and/or lymphoproliferative lesions in these strains has been mapped to a number of possible other genetic variations.2836 Interestingly, when compared with C57BL/6J and/or C3H/HeJ mice, the MRL/MpJ strain harbors diverse polymorphic alleles, unique QTL, or specific haplotypes that render this background more susceptible to autoimmune manifestations.2832 As an example, low to no expression of CFHR-C in MRL/MpJ may contribute to the immune hyperresponsiveness typical of this strain.109

BXSB/Mp mice are a recombinant inbred (RI) strain originating from a cross between a C57BL/6J female and a SB/Le male, also developed by Murphy125,126 in his work on autoimmune conditions (lab code Mp). They develop a lupus-like disorder that is accelerated in males and is attributed primarily to the Y-associated autoimmune accelerator locus (Yaa) of the SB/Le male founder. Yaa is a 4-mb translocated region from the X chromosome that includes multiple genes, among which Tlr7 seems to be the major contributor to the phenotype.127129

NZB mice develop a variety of autoimmune phenotypes characterized by hypergammaglobulinemia with elevated circulating autoantibodies (including anti-DNA antibodies and anti-thymocyte antibodies), Coombs positive hemolytic anemia, and immune complex glomerulonephritis. NZB mice also manifest a lymphoproliferative disorder involving the B1 subset of B cells. This condition progresses to lymphoma/leukemia, with similarities to human familial chronic lymphocytic leukemia.130134 NZW mice develop autoantibodies and glomerulonephritis, with a female predisposition.135 F1 hybrid offspring of NZB females and NZW males (also referred to as NZB/W) develop a life-limiting autoimmune condition characterized by high levels of antinuclear antibodies, hemolytic anemia, proteinuria, and progressive immune complex glomerulonephritis that is more severe in females.136138 NZB/W autoimmune phenotypes map to multiple susceptibility loci, including Sle, Lbw, and Wbw loci and polymorphisms in Tnf, Nkt2, and Cd93, and are linked to a low to no expression of CFHR-C.110,111,139

NZM2410 mice (New Zealand Mixed strain 2410, e.g., NZM2410/J https://www.jax.org/strain/002676) derive from NZB/W backcrossed to NZW mice then selected for lupus-like nephritis deaths and inbred. They bear the NZW histocompatibility haplotype H2z (Ku, Au, Sz, Dz). Males as well as females develop autoimmune glomerulonephritis at an early age, and this strain has been especially useful in mapping lupus susceptibility loci.138,140142

Important Spontaneous Mutations

Supplementary Table 2 gives a comprehensive overview for most of the well-known murine immune relevant mutations that exhibit Mendelian inheritance. Historically, identification of the genetic basis for spontaneous Mendelian (monogenic) phenotypes was attained via forward genetics approaches to confirm that the heritable trait (phenotype) maps to a specific locus. Additional molecular investigations, including sequencing, are applied to define the mutation further.88,143 An advantage of forward genetics is the relatively unbiased approach that requires no assumptions or hypotheses regarding the molecular basis of the trait or phenotype. An historical and illustrative example in immunology is the characterization of TLR4, first recognized as the main sensor for lipopolysaccharides (LPS) thanks to studies conducted on the spontaneously TLR4 deficient C3H/HeJ mice, and closely related TLR4 sufficient substrains.95 A null mutation Tlr4lps-del mapping to the same site was identified later in the C57BL/10ScCr substrain of the C57BL/10 mouse and is now available as C57BL/10ScNJ.144,68 Similarly, the role of Foxp3 as an essential transcription factor for the development of regulatory T cell (Tregs) was first revealed via the analysis of mice with the spontaneous scurfy mutation (Foxp3sf).145

Hereditary immune deficiencies related to spontaneous recessive scid, Lystbg (bg or beige), and Btkxid (xid) mutations have been valuable in the study of orthologous conditions in humans and other animals.146 The scid and nu (nude) mutations have been especially important for their utility in studying engrafted human tissues in the context of xenotrasplantation experiments.147

Hereditary hyperimmune or autommune conditions related to spontaneous recessive Faslpr (lpr, lymphoproliferation) and Faslgld (gld, generalized lymphoproliferative disease) mutations in an important cell death pathway have also been informative. Mice homozygous for either mutation develop lymphoproliferative and autoimmune phenotypes. The (recessive) lpr mutation at the Fas locus compromises the FAS-mediated apoptosis pathway.115,123,148,149 The (recessive) gld point mutation is in the Fas ligand (Fasl) locus, and homozygosity for this mutation also compromises FAS-mediated apoptosis. The gld mutation arose spontaneously in C3H/HeJ mice, resulting in the C3H/HeJ-Faslgld substrain.150,151

Interactions Among Mutations

Table 3 summarizes genetic and phenotypic characteristics of some of the widely used mice that carry multiple spontaneous immune relevant mutations. Before the advent of modern genetic engineering capabilities, interbreeding to combine multiple hereditary disorders was used to study phenotypic manifestations of gene interactions and to overcome limitations of the single mutation models, particularly in mice used for xenotransplantation experiments.89 As an example, scid-beige mice homozygous for both the Prkdcscid and Lystbg alleles were generated to combine the impaired B and T cell development of the Prkdcscid mouse with the defective NK cell function associated with the Lystbg mutation. These mice are not only severely immunodeficient, but they also lack the “leaky” phenotype of the Prkdcscid animals. The cooperation between the 2 mutations remarkably improves xenotransplantation compared with the single mutation in the Prkdcscid mouse.152,153

Table 3.

Overview of Immunologically Relevant Mouse Models that Combine Multiple Spontaneous Mutations

Allelic combination Background strain/s Phenotype References
  • Fasl gld /Fasl gld

  • Btk xid /Y

C3H/HeJ Btk xid decreases the severity of B cell manifestations associated with Faslgld including hypergammaglobulinemia, generation of anti-DNA autoantibodies and systemic immune-complex disease; no impact on T cell dependent Faslgld phenotype and lymphadenopathy. 89
  • Fas lpr /Fas lpr

  • Btk xid /Y

MRL/MpJ Btk xid decreases the severity of B cell manifestations associated with Faslpr including hypergammaglobulinemia, generation of anti-DNA autoantibodies and systemic immune-complex disease; no impact on T cell dependent Faslpr phenotype and lymphadenopathy. 89,158,159
  • Fas lpr /Fas lpr

  • Foxn1 nu /Foxn1 nu

C57BL/6J Foxn1 nu prevents the development of Faslpr-induced lymphadenopathy, unregulated B cell activation, hypergammaglobulinemia, anti-DNA autoantibodies and systemic immune-complex disease (a similar effect is obtained via neonatal thymectomy confirming the T cell dependency of Faslpr phenotype). 89,154156
  • Fas lpr /Fas lpr

  • Prkdc scid /Prkdc scid

MRL/MpJ; C.B-17 Fas lpr rescues the developmental deficit of thymic T cells associated with Prkdcscid; no effect on the B cell deficit caused by Prkdcscid. 160
  • Fas lpr /Fas lpr

  • X/Yaa

MRL/MpJ; C57BL/6J Yaa causes accelerated onset and increased severity of Faslpr-induced autoimmune condition and lymphadenopathy. 161,162
  • Foxn1 nu /Foxn1 nu

  • Lyst bg /Lyst bg

C57BL/6J; N:NIH(S) Lyst bg contributes defective NK cells to the T cell-deficient background associated with Foxn1nu; reduced NK cell activity does not seem to impact on the engraftment rate and growth of xenotransplanted human tumor cell lines. 89,163
  • Foxn1 nu /Foxn1 nu

  • Btk xid /Y or Btkxid/Btkxid

N:NIH(S) Defective T (Foxn1nu) and B (Btkxid) cell function and/or maturation; spectrum of the immune abnormalities is very similar to the one characterizing Prkdcscid mutants; severe depletion of both B and T cell domains in the spleen and lymph nodes; limited production of immunoglobulins; females showing high incidence of both lymphomas and ovarian granulosa cell tumors. 89,164166
  • Foxn1 nu /Foxn1 nu

  • Lyst bg /Lyst bg

  • Btk xid /Y or Btkxid/Btkxid

N:NIH(S); KSN Defective T (Foxn1nu), NK (Lystbg) and B (Btkxid) cell function and/or maturation; high incidence of multicentric lymphoblastic lymphoma; compared to single Foxn1nu mutants, improved engraftment rate and growth of xenotransplanted human tumor cell lines. 89,167,168
  • Dh/Dh +

  • Foxn1 nu /Foxn1 nu

N:NIH(S) Combined athymia and asplenia; defective T cell maturation and function; reduced B cell number; hypogammaglobulinemia; increased incidence of spontaneous mammary tumors compared to single-mutant founder lines. 89,169
  • Lyst bg /Lyst bg

  • X/Yaa

SB/Le Lyst bg attenuates severity and progression of Yaa-linked autoimmune condition resulting in prolonged survival and lack of immune complex glomerulonephritis; possible role of Lyst in B cell development and activation. 89
  • Btk xid /Y

  • X/Yaa

BXSB Btk xid prolongs survival and decreases the severity of B cell manifestations associated with Yaa including immune complex glomerulonephritis, hypergammaglobulinemia, autoantibody levels and lymphoid hyperplasia. 170
  • Prkdc scid /Prkdc scid

  • Lyst bg-J /Lyst bg-J

C.B-17 Defective T, B (Prkdcscid) and NK (Lystbg) cell function and/or maturation; reduced level of B cell leakiness; possible role of Lyst in B cell development and activation. 152,153
  • Prkdc scid /Prkdc scid

  • Hr hr / Hr hr

SCID Hairless Outbred (Crl:SHO) Impaired B and T cell development (Prkdcscid) associated with diffuse hair loss/alopecia (Hrhr). 171
  • Foxp3 sf /Foxp3 sf

  • Foxn1 nu /Foxn1 nu

129/RI; BALB/c Foxn1 nu prevents the development of Foxp3sf-induced autoimmune disease including anemia, multisystemic immune/inflammatory cell infiltrates, hypergammaglobulinemia, lymphadenopathy and splenomegaly (a similar, but less potent, effect is obtained via neonatal thymectomy confirming the T cell dependency of Foxp3sf phenotype). 172,173
  • Foxn1 nu /Foxn1 nu

  • Map3k14 aly /Map3k14 aly

BALB/cAJcl; C57BL/6J Athymia combined with lack of secondary lymphoid organs including lymph nodes, splenic white pulp, Peyer’s patches and isolated lymphoid organs; severe immunodeficiency with impaired humoral and cell- mediated immune responses; preserved intestinal γδ-IEL subset; confirmation that thymus and secondary lymphoid organs are not an essential requirement for the development of γδ-IEL. 174

IEL, intraepithelial lymphocytes; NK, natural killer.

Combinations of multiple mutations have proved useful in understanding the epistatic interactions among immune relevant genes. Double-mutant mice homozygous for both Faslpr and the Foxn1nu are an example. The congenital T cell deficiency that characterizes the Foxn1nu mutation is sufficient to abolish the autoimmune and lymphoproliferative phenotype associated with the Faslpr allele. This finding was consistent with the significant abrogation of the phenotype achieved by neonatal thymectomy in MRL/MpJ-Faslpr/J mice, and provided early support for the hypotheses regarding the T cell dependence of the Faslpr-associated autoimmune and lymphoproliferative condition.89,154157 Other important immunodeficient models featuring combinations of spontaneous and induced mutations along with specific strain-related immune variations are further discussed in a companion article by Simons and colleagues in the present issue of the ILAR Journal and include the well-known NSG and NOG mice. Both models carry a slightly different targeted mutation of Il2rg combined with the Prkdcscid mutation on different NOD inbred sublines.

Induced Immunodeficiencies

The mouse immune system can be modulated (regulated or disrupted) intentionally (and unintentionally) through experimental interventions such as exposures to irradiation, chemical compounds, microbial organisms (including virus, bacteria, and their toxins), or biological agents as well as through surgical manipulations. Immune suppression by these means has been especially useful in experiments of engrafted tissues or tumors and to study the immune response against specific infections or neoplasms. Examples from the major categories of intended experimental interventions to induce specific perturbations of the mouse immune system are summarized in Table 4.

Table 4.

Induced Immunodeficiencies (Intended Experimental Interventions)

Inducers Possible Effects on the Immune and Other Systems References
Physical: irradiation
γ rays and X rays Suppression of bone marrow resulting in marrow atrophy and pancytopenia. 182,237244
High dose: decreased splenic and thymic weights; loss of cortical thymocytes; decreased splenic CD4+ and CD8+ T cells; decreased circulating CD3+ cells.
Chronic low dose: prolonged life span in mice homozygous for the lymphoproliferation spontaneous mutation (Faslpr); increased CD4+ cells; suppression of IL6 and IL17, and up-regulation of Tregs in CIA mice; suppression of pro-inflammatory cytokines, reduction of CD8+ T cells, and induction of Tregs in murine EAE model.
Other: acute radiation syndrome and death in Prkdcscid mice and Prkdcdxnphmice (both are highly susceptible to ionizing radiations); radiation induced-thymic lymphoma in both male and female mice on a C57BL/6 background and NFS mice; radiation induced-myeloid leukemia in male RF mice (RF/J, RFM) and male CBA mice (CBA/Ca, CBA/Cne, CBA/H); induction of persistent oxidative stress in murine intestinal epithelium with potential for neoplastic transformation by heavy ion radiations; radiation-induced cataract; increased osteoclast activity and bone loss; radiation nephropathy.
α and β particles Release of DAMPs; activation of DCs; systemic and long-lasting T cell-mediated antitumor response in tumor-bearing mice; efficacy of α and β emitter-labeled monoclonal antibodies against fungal infections in mice. 245247
Other: radiation nephropathy.
UVB Immunosuppressed contact hypersensitivity (Xpa deficient mice); inhibited intra-tumor migration of NKs and CD8+ T cells (Xpa deficient mice); depressed delayed hypersensitivity in immunized mice; enhanced contact hypersensitivity and skin graft rejection in mice with dermal Langerin+ DCs. 248254
Narrowband (NB)-UVB: increased intestinal Tregs, and decreased severity of inflammatory lesions in mouse models of allogeneic GVHD.
UVA High dose: increased IFNγ, IL12, and heme oxygenase; inhibited increment of IL10 from UVB exposure. 255258
Medium dose: NO-mediated depletion of epidermal Langerhans cells; impaired development of skin memory CD8+ T cells in a mouse model of contact hypersensitivity.
Chemical agents
Endogenous and exogenous glucocorticoids Direct and receptor-mediated immunosuppression: attenuated DC activity; decreased DC number (apoptosis, tissue redistribution); enhanced inflammation; thymic atrophy (decreased DP thymocytes); dampened T cell activation (interference with TCR signaling); suppressed responses of TH1 and TH17 cells; reduced immunoglobulins. 259265
Other: osteopenia, decrease in bone formation rate and mineral apposition rate in skeletally mature and young mice; osteoporosis in CD-1 mice (mouse model of glucocorticoid-induced osteoporosis); cleft palate in A/J mice.
Cyclophosphamide (CYP; Cytoxan) Direct immunosuppression: depletion of CD8+ resident DCs in murine spleen and lymph nodes, with subsequent decrease in Treg suppressive function; neutropenia; depletion of suppressor or regulatory T cells in diabetic NOD mice. 266273,200
Other: enhanced antitumor efficacy by promoting proliferation/activation of adoptively transferred B and T cells after CYP-induced lymphodepletion in mice; reduced diversity of the fecal microbiota; hemorrhagic cystitis in C57BL/6 and DBA/2 mice; chronic cystitis in DBA/2 (CYP model of bladder pain syndrome); short root lengths and early apical foramen closure during molar root development in ICR mice; suppressed osteoblastogenesis and osteoclastogenesis in C57BL/6 male mice.
5 FU Direct immunosuppression: depletion of MDSCs, and stimulation of TH17 cells, IL17 production by CD4+ T cells, and tumor growth; no altered levels of circulating B, T, and NK cells. 207,208
Tacrolimus (FK506) Receptor-mediated immunosuppression: immunosuppressive effects on CD4+ T cells; marked tumor-promoting effect (topical tacrolimus) with decreased CD4/CD8 ratio; reduced inflammation in models of allergic rhinitis, conjunctivitis and arthritis. 211215
Other: nephrotoxicity.
Cyclosporin A (CsA) Receptor-mediated immunosuppression, reversible inhibition of T cell proliferation and proinflammatory immune reactions; blockage of all the changes resulting from intercellular signaling and cross-talk between DCs to T cells. 209,210
Rapamicin Receptor-mediated immunosuppression:Inhibition of mTOR: suppressed T cell activation, proliferation, and development of FoxP3+ cells; suppression of DC maturation, B cell activation, neutrophil chemotaxis and uptake of antigen by APCs. 274,275
Other: increases lifespan.
Busulfan; Treosulfan Direct immunosuppression:Busulfan: highly myelosuppressive, minimally immunosuppressive; diminished NK cell activity; late-stage (residual) bone marrow injury; stimulation of neuroinflammation through MCP-1. 276280
Treosulfan: high persisting myeloablation in BALB/c mice; more effective depletion of splenic B and T cells.
Physical: Surgical
Thymectomy Thymectomy (post-natal day 2-5): autoimmune hemolytic anemia, thyroiditis, gastritis, oophoritis, orchitis, and prostatitis at puberty due to lack of Tregs. 232
Splenectomy Systemic immune unresponsiveness; absence of tolerance after ocular injections of antigen in F4/80-deficient mice; retardation of tumor growth in melanoma-bearing mice. 281285,236
Biological agents
Anti-thymocyte globulin (ATG) Depletion of naïve T cells; less effective on memory T cells in NOD mice. 283,229
Prevention of autoimmune encephalomyelitis through expansion of myelin antigen-specific Foxp3+ Tregs in a murine EAE model.
β-1,3-Glucan Increased IL2, TNFα, IL17, IFNγ, and lymphocytes in mice treated with aflatoxin B1. 284
CpG oligodeoxynucleotides In murine models of infections: TH1 cytokine expression, activation of DCs, NK, and B cells.
Combined therapy with monoclonal antibodies: increased NK cell activity.
Bacterially derived ADP-ribosylating enterotoxins CT toxin produced by Vibrio cholera: secretion of TH2 cytokines, maturation of DCs, generation of Th2 and regulatory T cells, active suppression of TH1 responses. 230,285,291
LT enterotoxin from E. coli: mixed TH1/TH2 immune response.
Anti-lymphocyte serum (ALS) Long-term abrogation of autoimmunity in overtly diabetic NOD mice. 286
Monoclonal antibody (mAb) therapy Anti-mouse CD20 mAbs: depletion of mature B cells; reduction of CD4+ T cells, but maintainance of the interactions, functions, and migration of DCs and CD4+T cells; unaffected CD8+ T cell reactivity; absent release of inflammatory cytokines with effects on T cells. 287289
Anti-mouse CD4 mAbs: depletion of CD4+ T cells; expansion of CD8+ T cells with an effector phenotype and of tumor-reactive CD8+ T cells; compromised anti-tumor immune memory.
Anti-mouse CD8 mAbs: depletion of CD8+ T cells; decreased infiltration of CD4+ cells, neutrophils, and macrophages; downregulation of IL1β, IL6, TNFα, CXCL1, CCL2 and up-regulation of IL4 in a mouse model of wound healing.

APCs, antigen-presenting cells; CIA, collagen-induced arthritis; CT, Cholera toxin; DAMPs, damage-associated molecular patterns; DCs, dendritic cells; DP, double positive; EAE, experimental autoimmune encephalitis; GVHD, graft-versus-host disease; LT, heat labile toxin; MCP-1, monocyte chemoattractant protein 1; MDSCs, myeloid-derived suppressor cells; NK, natural killer cell; NO, nitric oxide; Tregs, regulatory T cells.

Ionizing and Ultraviolet Radiation

Ionizing radiation is a historically important method to suppress or ablate immunity. The peculiar vulnerability of the hematolymphoid tissue to ionizing radiation results in extensive lymphoid depletion and sustained myeloablation. For this reason, ionizing radiation remains an important immunosuppressive intervention allowing the engraftment of xenotransplants/allotransplants, including, for example, tumors or human haematopoietic stem cells for the generation of mice with humanized immune system.175,176 Sensitivity to irradiation has been linked to the capacity to repair radiation-induced DNA double-strand breaks. Immunodeficient mice harboring the Prkdcscid alleles are particularly radiosensitive due to the scid mutation that affects repair of radiation-induced DNA double-strand breaks.177,178 Susceptibility to irradiation varies among mice, with strains such as the C57BL/6, A/J, and C3H/HeMs being highly resistant and other strains such as BALB/c being highly sensitive.177,179 A hypomorphic Prkdc allele (Prkdcdxnph), identified in BALB/c strains, seems to have an important role in BALB/c susceptibility to ionizing radiation.178,180,181 Some detail on irradiation tolerance, variations, and dosage protocols is available from the sources of mice that are commonly irradiated (https://www.taconic.com/taconic-insights/oncology-immuno-oncology/rodent-irradiation-considerations.html; https://www.jax.org/jax-mice-and-services/find-and-order-jax-mice/most-popular-jax-mice-strains/immunodeficient-mouse-and-xenograft-host-comparisons). In addition to considering strain sensitivity when determining radiation dosage, calibration of the irradiator is also important, as there is considerable decay over time and actual dosage may differ between studies or between irradiators.

Immune-suppressive effects of high-dose γ-irradiation are well known.182 High-dose γ-irradiation differentially affects the diverse populations of mouse lymphocytes with B cells recognized as more radiosensitive than T cells.183 Repeated low-dose gamma irradiation also has profound immunomodulatory effects and is linked to a robust Th2 skewing that may mitigate autoimmune conditions that are dependent on a Th1 response. Suppression of pro-inflammatory cytokine production, reduced CD8+ CTLs, and up-regulation of Tregs also have been demonstrated in certain experimental conditions, including CIA and EAE.184

Overwhelming infections remain an important cause of mortality of irradiated experimental animals and clinical patients. Mice with defective adaptive immunity including nude, scid and NOD scid mice can effectively control common opportunistic agents such as Pseudomonads, until myeloablative effects of irradiation or other interventions eliminate their innate immunity as well.185 Effects of ionizing radiation on other tissues, and on developing or proliferating cells, influence morbidity and mortality of research mice. Radiation impact on developing brain, bone, eyes and teeth as well as on heart, lung, kidney, may complicate interpretation of disease or death related to rejection, GVHD, or other research endpoints.186195

Ultraviolet (UV) radiation effects on local skin immunity are especially relevant to research on photocarcinogenesis or inflammatory skin conditions.196198 Effects vary with dose, duration of exposure and wavelength composition.196198 UV radiation primarily affects adaptive immunity, and has been used to induce and promote skin photocarcinogenesis, and to modulate the immune response in diverse experimental immunoinflammatory conditions of the skin.196198

Chemicals

Experimental use of chemicals also has been and remains an important method to suppress or ablate immunity. Examples including metals, aromatic hydrocarbons and other environmental contaminants, and antimicrobial agents are summarized in Table 4. Alkylating agents that affect chromosomal DNA through formation of phosphodiesters and DNA-DNA crosslinks, are widely used. Cyclophosphamide (CYP), a cytotoxic alkylating agent used in the treatment of neoplastic and autoimmune diseases, is also exploited to induce neutropenia in the context of infectious disease studies.199 Mice with impaired granulocyte production and/or leukocyte function secondary to CYP are more prone to develop systemic disease upon experimental infection with environmental opportunists such as Pseudomonas aeruginosa or Cryptococcus neoformans.200,201 CYP has both immunomodulatory and immunosuppressive effects.202 Immunosuppression in mice appears to result from the induction of apoptosis in activated B and T cells as well as NK cells.203 At low doses, CYP may enhance immune responses to tumor antigens attributed, at least in part, to suppression of Tregs.204 Similarly, the alkylating agent busulfan is used as conditioning regimen to enhance engraftment of xenotrasplanted hematopoietic stem cells.205,206 Other important agents include 5-fluorouracil (5FU), which selectively depletes tumor-associated myeloid-derived suppressor cells (MDSCs) promoting the activation of tumor-specific CD8+ T cells.207,208 Calcineurin inhibitors (CNI), such as tacrolimus and cyclosporine A, directly inhibit Tregs function, by inhibiting pheripheral Tregs generation, and less directly by limiting IL2 production, in preventing transplant rejection and to treat a variety of autoimmune conditions.209215 Glucocorticoids are important clinically and experimentally for their anti-inflammatory and immunosuppressive effects.216

A variety of experimental interventions including hormones, antimicrobials, nanoparticles, etc., have immunomodulatory effects that may not be intended or expected, especially by investigators who are new to using them in mice. For example, estrogens (and synthetic estrogens such as diethylstilbestrol) and androgens have immunosuppressive effects that affect both adaptive and innate immunity.217220 Nanoparticles, usually studied as a drug delivery method or biomedical imaging tool (e.g., metallic nanoparticles), are typically taken up by macrophage/monocyte cells and may act either as immunostimulants or as immunosuppressants and may have additional immune effects related to imaging methods such as MRI or μCT.221 The unique physicochemical characteristics of nanoparticles influence their interactions with host’s immune system and determine the overall immunotoxicologic profile.222,223

Biologics

Biologics with immune modulating properties have been exploited in the experimental context to target specific functions of the mouse immune system and achieve definite preclinical endpoints.

Antibody-mediated depletion of cell lineage-specific immune effector cells has been used to delineate their roles in innate and adaptive immunity, in rejection, GVHD, and other conditions.216,224226 Anti-thymocyte globulin (ATG), is another important immunosuppressive agent that specifically depletes T cells from peripheral blood and lymphoid organs in NOD mice; it is also used in the modulation of graft rejection and autoimmune disorders in mice.227,228 Glucans, CpG oligodeoxynucleotides (CpG ODN) and bacterial enterotoxins have been used as prophylactic or therapeutic interventions to modify immune responses to infections or vaccination, or to counteract effects of immunotoxic agents (see Table 4).229,230

Surgical

Thymectomy or splenectomy are the traditional surgical methods to alter immunity. Thymectomy in neonatal or adult animals has profound effects on T cell development and continues to be an important procedure in studies of T cell ontogeny, tolerance and education. Neonatal thymectomy experiments offered early evidence of the existence of Tregs as these mice develop autoimmune disease shortly after the removal of thymus.231 Thymectomy is also used to investigate the dynamics of extrathymic T cell development.232 However, mice exhibit a relatively high frequency of functional thymic tissue in ectopic locations, especially in close proximity to the thyroid gland (also known as cervical thymus). While ectopic thymi may be small, they can be confounding source of T cells. They are reported to be more common in NOD and BALB/c mice compared to C57BL/6 mice.232,233

Splenectomy has been used to study the role of the spleen in infectious disease, peripheral antigen tolerance, and tumor growth.234 In cancer, some splenectomy studies implicate the spleen in promoting tumor antigen tolerance.234,235 while others demonstrate a role of the spleen in maintaining an effective antitumor immune response and prevention of metastatic disease.236

Induced Autoimmune and Hyperimmune Conditions

Autoimmune diseases arise when there is poor control of self-reactive lymphocytes and cytokine production, or disrupted regulatory T cell and effector T cell balance. While underlying genetic polymorphisms predispose to immune hyperresponsiveness, manifestation of disease often requires additional triggers such as microbial infections, dysbiosis, or tissue damage. Once initiated, cytokines participate in disruptions of immune tolerance by altering the balance between T-effector functions and T-suppressor functions.290292 Strain-related variations in innate and adaptive immunity affect penetrance, onset and severity of disease.7,27,89,293,294 Modifiers such as Slamf-haplotype 2 seem relevant to autoimmunity in MRL/MpJ mice and not so relevant on other backgrounds such as BALB/c.6062 The complexity of autoimmune conditions in mice has many parallels with human and, because of a more granular characterization of strain genetics, may have much to offer to our understanding of the human conditions and interventions for them.295,296 Two examples are discussed here.

Rheumatoid Arthritis

Rheumatoid arthritis (RA) is an immune-mediated destruction of the synovial lining of the joints, with devastating effects on underlying cartilage and bone. Susceptibility to the induction of rheumatoid arthritis-like conditions in mice, using type II collagen-induced arthritis (CIA) or proteoglycan-(aggrecan)-induced arthritis (PGIA), depends on multiple susceptibility alleles and QTL.96,297,298 The disease in mice and in humans is polygenic and complex. MHC H2 subtypes seem to have more impact on CIA than on PGIA susceptibility, and PGIA susceptibility is influenced by multiple genes.96,298,299 Strains expressing the H-2q and the H-2r haplotypes are most susceptible to CIA. DBA/1 (H-2q) are sensitive to CIA but insensitive to PGIA. BALB/c (H-2d) mice are not so susceptible to CIA but are highly susceptible to PGIA.96,297,299 In contrast, DBA/2 (H-2d) are resistant to arthritis induction by either method, implicating roles for strain associated modifier genes.299,300 Non-MHC QTL associated with susceptibility to CIA and/or PGIA localize to regions on mouse chromosomes 2, 3, 7, 15, and 19 that contain multiple candidate genes with known immune functions.299

Multiple Sclerosis

Multiple sclerosis (MS) is an inflammatory demyelinating disorder with a spectrum of disease manifestations. While disease is associated with certain genetic polymorphisms, environmental triggers as well as sex hormones have roles in disease development.290,301 A spontaneous mouse model of MS has not been identified. But various aspects of MS are recapitulated by experimental autoimmune encephalomyelitis (EAE), classically induced “actively” by immunization with immunodominant myelin epitope components in combination with immunostimulants, or induced “passively” by adoptive transfer of preactivated myelin-specific T cells into naïve mice.98,302305

EAE in mice was first reported in 1975, and the SJL/J and C3H/HeJ strains were identified as susceptible strains.98,294,302304,306 SJL/J mice are used to model features of relapsing-remitting MS, and their susceptibility is associated with several polymorphisms, including hyper-responsive IL12 and hypo-active IL2 and IL4.67,306,307 Additionally, C57BL/6, DBA1, and C3H/HeJ strains also are sensitive to induction of EAE.98,294,308

GEM models such as transgenic mice bearing human TCR and T cells targeting myelin-specific antigens (e.g., myelin basic protein) have been informative,309 as has immune-mediated demyelination associated with infections by Theiler’s Mouse Encephalitis Virus, a Picornavirus, in susceptible SJL/J and resistant C57BL/6.310312 Demyelination with certain strains of Mouse Hepatitis Virus (MHV), a coronavirus, has been used to model features of MS in susceptible C57BL/6 and BALB/c mice. This is primarily a virus-mediated cytolytic phenomenon, and SJL/J resistance is attributed to their spontaneous mutation in Ceacam1, whose protein product is an important receptor for neurovirulent MHV strains.313315

Other Immunomodulators and Unintended Experimental Consequences

Environmental Factors

Table 5 summarizes examples of immune effects of common environmental factors including husbandry conditions, microbiota, as well as effects caused by experimental or therapeutic interventions. These examples illustrate why reporting of environmental and husbandry conditions and specifics of experimental or therapeutic interventions is warranted in scientific publications. Microenvironment refers to the immediate physical environment surrounding the animal such as the cage, pen, or stall. Macroenvironment refers to the physical environment of the secondary enclosure (e.g., a room, a barn, or an outdoor habitat).323 A multitude of factors in the microenvironment and macroenvironment can be stressors. Stressors activate the hypothalamic-pituitary-adrenal axis, in turn increasing circulating glucocorticoids. In mice, corticosterone is the primary stress-induced glucocorticoid. Corticosterone elevations (and corticosterone-mediated lymphocytolysis) are expected with stressors such as adverse environmental conditions, shipping, handling, social stresses, noise, vibration, etc.317319 Responses to stressors also vary with mouse strains.320,321

Table 5.

Other Immunomodulators, Including Unintended Immune Consequences of Husbandry and Environmental Factors, Clinical and Experimental Interventions

Immunomodulators Possible Effects on the Immune and Other Systems References
Environmental factors
Housing conditions
Caging Individual ventilated cages (compared to static microisolator caging): decreased bioburden and risk of intercage infection spread; increased cold stress; decreased circulating leukocytes; decreased intracage ammonia levels and correlated nasal pathology. 322325
Bedding Experimentally relevant parameters influenced by the type of bedding: higher intracage ammonia levels with reclaimed wood pulp bedding; corncob bedding associated with decreased efficiency of feed conversion in mice fed a high-fat diet; hepatotoxicity associated with vermiculite and unbleached pulp from pine and eucalyptus; hepatic and mammary carcinogenesis associated with aromatic red cedar bedding; altered estrogen signaling mainly due to BPA residues; corncob bedding associated with increased aggressivity and social stress in females; drastically lower endotoxin levels and bioburden associated with paper bedding. 323,342,352357,366
Single or group housing and social stressors Group housing: negative social events associated with lower lymphocyte proliferation; lower level of antigen-specific IgG; granulocytosis; lymphopenia, higher predisposition to tumor development and progression, huddling associated with amelioration of cold stress. 326330
Individual housing: decreased antibody production; worsened allergic skin reaction; increased cold stress.
Environmental enrichment Reduced stress levels; reduced oxidative stress; enhanced NK antitumor functions; enhanced macrophage chemotaxis and phagocytosis; improved capacity to clear systemic microbial infection; enhanced lymphocyte chemotaxis and proliferation; increased lifespan. 331336
Temperature and humidity Thermoneutral housing temperature (26°–34°C): reduced tumor formation, growth rate and metastasis due to increased CD8+ T cells; reduced myeloid-derived suppressor cells and Tregs. 327,328,337341,473476
Sub-thermoneutral housing temperature (20°–26°C): suppressed immune responses; increased therapeutic resistance of tumor and GVHD severity; suppressed myeloid cells function; alternative activation of macrophages.
Elevated humidity: increased bioburden; high ammonia levels due to expansion in urea-converting microflora.
Environmental noise and vibration Altered tumor resistance; immunosuppression; reduced body weight; reduced fertility. 348351,477
Inappropriate handling; untrained personnel Increased risk of infection associated with inappropriate PPE and insufficient sterilization of equipment; pain, discomfort and stress associated with frequent/improper handling. 316
Altered light-dark cycle Suppressed immune response; decreased splenic T cells; continuous illumination associated with decreased CD8+ and CD4+ cells in thymus and lymph nodes. 343345
Dim lights Elevated nighttime light exposure in male mice associated with worsened inflammation and weight gain under high-fat diet regimen. 478
Diet and water modifications
Caloric restriction Immune effects: reduced H2O2, TNF α, IL6, IL2, IL10, NO, IFNγ; decreased macrophage activation; impaired NK cell function; reduced IgA in small intestine and serum IgG. 363,478483
Other effects: increased lifespan; reduced age-related morbidities.
Protein-energy malnutrition Impaired proliferation CD8+ T cells; modulation of intestinal IgA responses to rotavirus; increased duodenal γδ IELs; increased production of jejunal proinflammatory cytokines in response to bacteria. 484486
Prolonged fasting (48–120 h) Stress response due to activation of hypothalamic-pituitary-adrenal axis; thymic atrophy (apoptosis of cortical DP thymocytes). 487
High-fat diet (in C57BL/6 mice) Suppression of delayed hypersensitivity; altered intestinal microbiota with stimulation of mucosal immunity; altered systemic metabolomes; inflammation of adipose tissue with release of adipokines, cytokines, and chemokines, and propagation of a chronic inflammatory state (inflamobesity). 488490
Chlorella vulgaris supplementation CYP-treated mice: reinstated lymphocyte proliferation and macrophage phagocytic activity; stimulation of IL2, IL12, TNFα, IFNγ, NK cell cytotoxicity; decreased splenic necrosis. 491
Polyunsaturated fatty acids supplementation Dietary DHA and AA associated with improved allergen-induced dermatitis as consequence of increased FoxP3+ T cells, elevated IL10, and decreased TNFα. 492
Water acidification Switch from normal tap water to acidified water associated with severe and long-lasting stress. 343
Nutritional deficiencies
Zinc deficiency Thymic atrophy (loss of DP thymocytes); accelerated lymphopenia with loss of antibody and cell-mediated responses; decreased number of pre-B cells, better survival for pro-T cells and mature DP and CD8+ T cells; increased myeloid lineage in bone marrow. 493496
Vitamin A deficiency Decreased ILC3 and antibacterial responses; compensatory expansion in IL-13-producing ILC2 and increased anti-helminth responses; intestine devoid of CD4+ and CD8+ T cells; lower salivary IgA levels and increased serum IgG response in mouse model of influenza; decreased mucosal antigen-specific IgA responses. 497499
Vitamin D deficiency VDR-deficient mice: increased mature DCs in skin draining lymph nodes; decreased Th1-cell responses and induction of IL10-producing Tregs. 500
Diet and water contaminations
Estrogenic endocrine-disruptors Isoflavones (genistein): thymic atrophy; suppression of delayed hypersensitivity; decreased splenic NK cells; decreased IFNγ in response to bacterial infection. 365,366,501503
Mycotoxins (aflatoxins, deoxynivalenol, zearalenone): elevated IgA and IgE; kidney mesangial IgA deposits; polyclonal activation of IgA secreting cells; IgA autoantibody.
BPA (cages, water bottles): lupus-like syndrome (C57BL/6 mice); allergic airway disease (BALB/c mice).
Halogenated aromatic hydrocarbons (PCDFs;PCDDs) Contaminated food and bedding: inhibited innate and adaptive immune responses; atrophy of lymphoid organs; TCDD targets thymic lymphoblasts. 364,504,505
Metals (As, Cd, Pb, Hg, Se) Complex immune-modulating effects (immunosuppression and immunostimulation). 504,506
As: decreased DCs in mediastinal lymph nodes of influenza A-infected C57BL/6 mice.
Microbial status, pathogens, and biosecurity
MHV MHV-3-infected C57BL/6: impairment of pre-B cells maturation and B cells functions. 507509
A59-infected BALB/c: transient lymphocyte apoptosis in the thymus.
MHV-JHM-infected BALB/cByJ: functionally altered CD4+ and CD8+ T cells, and APCs.
Sendai virus Interference with macrophage and their phagocytic activity, NK cells, and T and B cell function; increased isograft rejection. 507,510513
MNV Lethal infection in mice deficient for STAT1 and IFN receptors; alteration of immune/inflammatory parameters in diverse mouse models including Mdr1a deficient animals infected with Helicobacter bilis interfering with dendritic cell function and cytokine responses; infection of wild-type mice associated with mild intestinal inflammation, splenic red pulp expansion, and white pulp activation. 514516
MuHV-1 Loss of splenic T and B cells; interference with key coordinating role of DCs; functional impairment of macrophages and loss of response to cytokines; altered responses to mitogens, antigens, increased allograft rejection, delayed type hypersensitivity responses, and clearance of other pathogens; formation of anti-cardiac autoantibodies. 440,517520
MuHV-3 Thymic necrosis (specific targeting of CD4+ T cells in newborn mice); autoimmune gastritis in BALB/c and A strain; autoimmune oophoritis and production of antibodies to thyroglobulin. 413,440,521
MPV Suppressed proliferation (spleen, popliteal lymph node), increased proliferation (mesenteric lymph node) in ovalbumin-primed mice; altered alloreactive T cells and abnormal CD8+ T cell rejection of tumors and skin allografts (BALB/c); rejection of syngeneic grafts. 522,523
MVM MVM: oncolytic, cytotoxic, replicative cancer inhibitor; deregulation of the Raf signaling cascade. 415,524
MVMi: depressed myelopoeisis in neonatal BALB/c; depletion of hemopoietic precursors, leukopenia, and compensatory erythropoiesis in adult and neonate SCID mice.
Murine retroviruses Insertional mutagenesis (with reintegration of endogenous retroviruses or transposition of retroelements): immune relevant mutation such as Foxn1nu, Lepob, Faslpr. 439443,448,525529
Endogenous retroviruses in pancreatic islets: contribution to immune-mediated insulitis NOD mice.
LP-BM5-infected C57BL/6 mice: lymphadenopathy, splenomegaly; hypergammaglobulinemia; T and B cell dysfunctions; late appearance of B cell lymphomas; opportunistic infections.
LCMV LCMV disease: all pathological alterations following infection are immune-mediated; prototype for virus-induced T-lymphocyte-mediated immune injury and for immune complex disease; protection from LCMV-induced disease conferred through immunesuppression; noncanonical type I IFN signaling responsible for lethality in LCMV-infected Stat1 deficient mice. 530532
MHV-68 Experimental infections of laboratory mice to study the pathogenesis of human lymphoproliferative disorders associated with EBV. 422,426430
Bacteria Mortality/morbidity (sepsis) in immune deficient mice: Pseudomonas aeruginosa, Klebsiella spp., E coli; potentially any bacteria in severely immunocompromised mice. 375,378,381,440,451,533
Abscesses: Staphylococci, Pasteurella pneumotropica.
Skin disease/morbidity: Corynebacterium bovis, Staphylococci.
Mycoplasma arginini: suppurative arthritis in Prkdcscid mice inoculated with contaminated cell lines.
Fungi Pneumocystis murina: respiratory disease and mortality in immunodeficient mice. 378,381,534536
Candida spp.: recent reports associated with immune deficiency/suppression and or use of antimicrobials.
Biosecurity in immunodeficient mice High risk of Pneumocystis carinii infection in T cell-deficient mice including Foxn1nu, Prkdcscid mice and immune impaired GEMs; immunodeficient traits in mutant mice masked by the immune/inflammatory response associated with chronic γ-herpesvirus infection; MNV infection in Atg16l1-deficient mice associated with Paneth cell abnormalities; murine papillomavirus associated with proliferative lesions at the mucocutaneous junctions of Foxn1nu mice; mousepox recrudescence following immunosuppression and transmission to naïve mice. 375,400,537540
Biosecurity: contaminated biologicals Rodent pathogens (latent infections): contaminated serum with mousepox. 378,410,434,451,452,541
Human pathogens: contaminated human cell lines (humanized mice and patient derived xenografts mice).
Mycoplasma arginini: suppurative arthritis in Prkdcscid mice (contaminated cell lines).
Modulation of the microbiome SFB associated with the development of IL17 and IL22-producing CD4+ T cells (TH17 cells) in the intestinal lamina propria of germ-free mice. 386,387,405,406
Tritrichomonas muris: associated with elevated TH1 response in the cecum of naive WT mice and accelerated colitis in Rag1-deficient mice after T cell transfer.
Drugs administered for clinical or experimental purposes
Tamoxifen-inducible Cre/loxP system (Cre-ERT2) Estrogen-dependent and -independent tamoxifen immunomodulatory effect; shift from a TH1- to a TH2-mediated immune response. 458,459
Tetracycline/doxycycline-inducible Tet-Off/Tet-On system Doxycycline-dependent modulation of immune and inflammatory functions including allotransplant rejection, response to LPS, neutrophil chemotaxis; tetracycline/doxycycline-induced dysbiosis. 461,462,472
Nitrosamines, nitrates, nitrites (mutagens, carcinogens) DMN: suppression of both humoral and cell-mediated immunity. 542544
ENU: lymphoma (AKR/J, C58/J, C57BL/6J, NOD/LtJ); myeloid malignancies (SWR/J, DBA/2J); thymic lymphoma with/without K-ras mutations.
TMP-SMX TMP-SMX alone: no effect on hematopoiesis or immune cell functions. 545
TMP-SMX synergized with zidovudine: anemia, thrombocytopenia, lymphopenia, and neutropenia, decreased splenic macrophages, suppressed AC-dependent T cell responses.
Ivermectin Immunomodulation of T-helper cells; decreased recruitment of immune cells and cytokines in a model of asthma; unintended activation of tamoxifen-regulated Cre fusion protein in T cells. 460,546,547
Estrogens (for engraftment of estrogen-dependent tumors) Increased splenic neutrophils (estrogen-treated C57BL/6 mice); enhanced IFNγ expression; thymic atrophy (DERKO mice); myelosuppression (decreased pluripotent hematopoietic stem cells). 501,548552
Synthetic estrogens (DES): altered thymic T cell differentiation through interference with positive and negative selection processes in prenatally exposed mice; functionally defective NK cells and increased tumor susceptibility in neonatally exposed female mice.
Other: increased trabecular bone mineral density, fat reduction and increased uterine weight (DERKO mice); fibro-osseous lesions (bone marrow replacement by fibrovascular stroma (KK/HlJ and NZW/LacJ female mice).
Androgens (for engraftment of androgen-dependent tumors) Androgen stimulation: thymic involution resulting from decreased colonization of bone-marrow-derived stem cells; loss of thymic epithelial cells; thymocyte apoptosis; inhibition of CD4+ T cell differentiation through upregulation of phosphate Ptpn1; erythroid hyperplasia. 553556
Castration: enhanced CD8+ T cell vaccine response to prostate-specific antigens.
Streptozotocin Early lymphopenia in both blood and spleen; relative increased Tregs in spleen, peripheral blood, and lymph nodes; delayed islet and skin allograft rejection. 557
NPs Suppression of systemic humoral immunity (multi wall carbon nanotubes); inhibition of T cell-mediated immunity (iron oxide NPs, fuellerene 60); myelosuppression (Sb2O3, Co, ZnO, TiO2 NPs); allergic reactions (Ag NPs); anti-inflammatory activity and inhibition of cellular responses induced by IL1B (citrate-coated gold NPs). 558563
Other experimental interventions
Cre/loxP Activation of STING antiviral response by endonuclease activity of Cre recombinase. 457
CRISPR-Cas9 Adaptive immune response against Cas9. 458,459
Tetracycline/doxycycline-inducible Tet-Off/Tet-On system Apoptotic response in activated lymphocytes resulting from DNA binding by tTA/rtTA. 464
Classical reporter molecules Increase in the CTL response against transplanted eGFP-expressing leukemia cells in BALB/c mice; IFNγ response to the dominant CTL epitope of Luc, with consequent restricted growth and metastatic activity of the reporter-labelled tumor cells in a mouse model of mammary adenocarcinoma; antigen specific activation of T cells to the reporter gene β-galactosidase, with loss of transgene expression. 465470,564,565

AA, arachidonic acid; AC, accessory cell; BPA, Bisphenol A; CTL, cytotoxic T lymphocyte; CYP, cyclophospharmide; DCs, dendritic cells; DERKO, double ER knockout mice; DES, diethylstilbestrol; DP, double positive; DHA, docosahexaenoic acid; DMN, dimethylnitrosamine; EBV, Epstein-Barr virus; eGFP, enhanced green fluorescent protein; ENU, N-ethyl-N-nitrosourea; GVHD, graft-versus-host disease; IBD, inflammatory bowel disease; IELs, intra-epithelial lymphocytes; ILC3, type 3 innate lymphoid cells; ILC2, type 2 innate lymphoid cells; LCMV, lymphocytic choriomeningitis virus; Luc, luciferase; MHV, mouse hepatitis virus; MHV-68, murine gammaherpesvirus 68; MNV, murine norovirus; MNM, minute virus of mice; MPV, mouse parvovirus; MuHV-1, murid herpesvirus 1 (mouse cytomegalovirus); MuHV-3, murid herpesvirus 3 (mouse thymic virus); NKs, natural killer cells; NPs, nanoparticles; PPE, personal protective equipment; rtTA, reverse tetracycline-controlled transactivator protein; SFB, segmented filamentous bacteria; TCDD, 2,3,7,8-tetrachlorodibenzodioxin; TMP-SMZ, trimethoprim/sulfamethoxazole; Tregs, regulatory T cells; tTA, tetracycline-controlled transactivator protein; VDR, vitamin D receptor.

Caging

Common contemporary caging options are open top, static microsiolators (filter top cages), and individually ventilated caging. Suspended wire caging is less common today but may be scientifically justified to prevent coprophagy and ingestion of drugs or metabolites in feces. Individually ventilated caging is increasingly available with advantages in terms of barrier protection of the animals, lower bioburden, and cage changing frequency and with concerns in terms of microenvironment temperature, humidity, wind, and dust. Temperature, vibration, and microbial burden (discussed further below) are among the variables with expected immune effects.322325

Housing density

Co-housing or group housing of mice is practical and economical with compatible animals that do not fight and kill each other before study endpoints. Single housing can be required, especially for male mice to survive to study endpoints. Co-housing vs single housing effects on stress and immunity vary with strain, sex, and other conditions.326329

Enrichment

Enrichment for shelter, nesting, and gnawing have variable effects that are often associated with strain, sex, and other conditions. In general, provision of nesting material helps to reduce the level of stress and influences positively several immune parameters including NK cell antitumor functions.331336

Temperature humidity

Current temperature recommendations for mouse housing of 22–26°C are below the mouse thermoneutral zone of 30–32°C. The “mild” cold stress caused by standard sub-thermoneutral housing temperatures affects immune responses, tumor growth, and other experimental outcomes. Huddling and nest building are methods of behavioral thermoregulation used by mice under cold stress. Recommended relative humidity is 55% ± 10%. Humidity levels vary with type of caging, season, and geographic location. Higher humidity is associated with increased levels of ammonia and bioburden with severe impairment of respiratory mucosal immune response and increased risk of opportunistic infections, respectively.327,328,337342

Illumination (Light)

Circadian and light effects on immunity are recognized in many species, including humans and mice. Albino animals have higher light sensitivity, and a number of common mouse strains are blind with retinal degeneration but still exhibit responses to light and light cycles.343345 Dysregulation of circadian rhythmicity in mice induces a generalized proinflammatory macrophage activation and exacerbates diet-induced systemic insulin resistance and glucose intolerance. A balanced circadian rhythm is also critical to maintain immune homeostasis via the immunoregulatory activity of the neurohormone melatonin.346,347

Noise vibration

While a number of common mouse strains are deaf or become deaf with age, hearing mice perceive and respond to sounds outside of human ranges. Noise and vibration are shown to cause stress, induce corticosterone, and negatively affect reproduction.348351

Bedding

While contemporary commercial contact bedding materials tend to be far more standardized with more quality control and freedom from contaminants than previously, contaminants with potential effects on research outcomes can still occur in bedding material. Dust, ammonia levels, fungal spores, phytoestrogens, and endotoxins in bedding also have implications for diverse research. Regional variation among bedding material has implications for various research areas, including immunology, with corncob bedding more available in the United States than in the European Union and other sites, and with hardwoods, cellulose, or paper being other common options. The relative palatability of or preference for a bedding over the intended diet may affect consumption of the diet.323,352357

Diet

Contemporary commercial research diets also are far more standardized with more quality control than previously, and nutritional deficiencies are unlikely on contemporary commercial diets. Nutritional requirements for mice, including adequate levels of nutrients,358 minerals,359 and vitamins,360 exist as do guidelines for contaminants in laboratory rodent diets.361,362 Possible contaminants with immunomodulatory effects include industrial chemicals (e.g., PCBs, PCDDs, and PCDFs), pesticides (e.g., DDT), metals, nitrosamines, endocrine-disrupting compounds, and mycotoxins. However, contaminants are identified in contemporary diets and are a concern for biomedical research and regulatory toxicology.363,364 Endocrine-disrupting phytoestrogen-rich ingredients, especially soy and alfalfa, as primary protein sources are expected in natural ingredient (aka grain-based or cereal-based) diets. Phytoestrogens are recognized to have influences on rodent reproduction, immunity, cardiovascular, neoplastic, and other conditions.365,366 Animal byproducts, bone meal, and fish meal are used in many natural ingredient diets and are a source of nitrites and nitrosamines.363,367

Poor reporting of research-relevant diet factors such as differences between purified and natural ingredient diets have attracted attention and concern recently.10,358,368 Research diets are frequently provided ad libitum to rodents on shorter term studies. Diet restriction in long-term studies usually improves survival and reduces neoplastic, kidney, inflammatory, and other lesions.369371

Water

Contemporary water sources and delivery methods frequently include reverse osmosis, filtration, hyperchlorination, acidification, or some combination of these, delivered by water bottles, glass, or various plastics, tinted or untinted, and/or automated watering systems.

Acidification became a common practice for research rodents to control opportunistic bacteria (especially P. aeuroginosa) causing morbidity mortality in immune-deficient rodents that were further immunosuppressed by irradiation that further compromised or eliminated their innate immunity. Water treatments including administered drugs can affect water consumption and have immune or other effects that warrant reporting in publications.343,372374

Husbandry and Biosecurity

Special husbandry needs of immunodeficient mice are largely related to protection from agents that may cause morbidity and mortality. Such agents may be harbored by clinically “healthy” immune sufficient mice, or possibly by human handlers, and may be transferred by common equipment and other fomites. Proximity to immune sufficient mice or to any mouse cohort with different microbial status warrants special procedures and policies for sanitation and sterilization of caging, feed, water and other materials, sequence of animal handling, and microbial surveillance. GEM models may also manifest unexpected immunodeficiencies.375 Immunomodulatory effects by common agents (Table 5) demand that immune relevant research must pay greater attention to microbial exclusion lists and definition of the specific pathogen free (SPF) status in the vivarium as well as in reporting. Use of the term SPF requires specification of the excluded agents.316,376378

Some of the most concerning opportunistic agents in contemporary immunodeficient mice, such as Staphylococcus xylosus, Corynebacterium bovis, and Pneumocystis murina, are fairly common and usually subclinical in immune sufficient mice.379384 (see also Table 5)

Microbiota and Microbiome

Autochthonous (commensal and symbiotic) microbiota

Systemic and mucosal immunity in mice are influenced by the intestinal flora (microbiota).27,375,385 The intestinal microbiota are important to effective mucosal immunity and to immune responses beyond the gut. As an example, segmented filamentous bacteria (SFB) have been identified as an important antigenic stimulus in inducing Th17 responses, and murine Th17 responses are blunted in mice that lack SFB.395 Also SFB are shown to influence neuroinflammation in EAE models, diabetes susceptibility in NOD mice, and development of autoimmune arthritis in some models.387390 SFB normally colonize the distal small intestine of infant mice and decline with the maturation of the mucosal barrier and local IgA levels.391 In mice with deficient adaptive immunity or Ig production, or mice specifically deficient in IgA, SFB persist with expanded distribution throughout the small intestine.392,393 SFB are difficult to propagate in vitro and have not been included in the standardized communities of intestinal microbiota (e.g., Altered Schaedler flora) specifically maintained in some sources of laboratory mice to uniform the influence of microbiota on the experimental conditions. In this context, SFB are not expected in immune deficient mice from certain commercial vendors that maintain the mice in isolators with defined or highly restricted flora.394,395

Strain-associated and vendor-dependent differences in the gut microflora of laboratory mice have been identified and are implicated in variability in research results (see Table 5).378,385,396400 Flora with more Bacteroides spp. and Parabacteroides spp. such as Parabacteroides distasonis may mitigate DSS-induced colitis.401 Mice of similar strains but from sources with more simplified or restricted microbiota, lacking SFB, have quite different dendritic cell profiles and Th17 responses.402 In several immune relevant GEM including IL10, T cell receptor alpha, and IL2 knockout mice, intestinal inflammation also is substantially influenced by intestinal microbiome.403,404 Enteric protists are common in mice (but usually excluded from commercial sources) and also have been shown to influence Th17 and Th1 responses as well.405,406 The microbiota or autochthonous microflora of research animals are increasingly recognized as highly research relevant. The restricted microflora of naïve mice from reputable commercial sources have been presented as a research concern, but their well-characterized microbiota also represent an opportunity for this area of immune relevant research.407408

Allochthonous (noncommensal) agents

Morbidity, mortality, and other adverse or confounding effects of infectious agents on research have led to great effort and expense toward microbial definition and exclusion by commercial sources of mice and for quarantine and surveillance by research programs to protect animals and research from infections.410 Immune deficient mice are notoriously susceptible to disease and death from pathogens and opportunists. The same agents in immune sufficient mice may result in subclinical infections or a spectrum of disease phenotypes that are influenced by genetic background, age, sex, and other factors. But any agent detected by an immune system can be expected to elicit an immune response, or “immunomodulate.” Table 5 summarizes examples of microbial effects on immunity and particular concerns for morbidity and mortality in immune deficient mice.411,412

Viruses with selective tropisms for immune cells include some of the murine parvoviruses, herpesviruses, and retroviruses. Many of the parvoviruses infecting mice are lymphocytotropic, altering both CD4+ and CD8+ T cell-mediated responses during acute infection.378,413,414 Although long-term immune effects may not be identified with natural infections by some parvoviruses, significant immunomodulation is well documented with infection by others (Table 5).378 Parvoviruses replicate in actively dividing cells and are studied as oncolytic agents in combined anti-cancer therapies.415 Several mouse parvoviruses were identified originally as contaminants in biological materials such as tumor cell lines. They remain among the most common agents identified in research mice, pet store and feral mice, and biological materials. Despite the usual absence of clinical signs in parovirus-infected mice, these agents should be especially concerning in immune relevant and cancer studies.409,416420

Although mouse herpesviruses are not expected in contemporary research colonies, mice are host to several lymphocytotropic herpesviruses that are reported in pet store and feral mice.421,422 Mouse thymic virus infection in newborn mice causes thymic necrosis, with selective targeting of T cells, and transient immunosuppression.413 This agent or a close relative was recently classified under the genus Roseolovirus similar to human roseoloviruses.423,424 Murine cytomegalovirus is used to model human cytomegalovirus infection and targets hematolymphoid tissues and salivary glands. Disease manifestations vary with the genetic background.425 Occult (seronegative) murine cytomegalovirus infection has been shown to affect responses to allografts.426

Murine gammaherpesvirus 68, a natural pathogen of bank voles, is related to human gamma herpesviruses Epstein-Barr virus (EBV) and Kaposi sarcoma-associated herpesvirus and is used to study the pathogenesis of gammaherpesviruses in experimentally infected mice. However, Mus musculus ssp. are not the natural host, and horizontal transmission between laboratory mice is not expected.422,426430 EBV is a human B-lymphotropic gamma herpesvirus that infects more than 90% of the human population. Human infections are subclinical (latent) when effectively controlled or can result in infectious mononucleosis or malignancies such as Burkitt’s lymphoma, nasopharyngeal carcinoma, Hodgkin’s lymphoma, and post-transplant lymphoproliferative disorders. Immunodeficient and humanized mice have been informative preclinical tools for studying the pathogenesis of some of the conditions associated with EBV.431433 EBV-induced post-transplant lymphoproliferative disorders are also increasingly recognized to complicate research with human patient derived xenografts in severely immunodeficient mice434436 and may be amenable to suppression of human lymphocyte proliferation in the donor tissue.437,438

Exogenous retroviruses and active endogenous retroviruses have lymphocyte tropisms and roles in immune modulation and lymphoproliferative conditions as well as in mammary carcinogenesis, sarcoma development, and lymphomagenesis. Exogenous horizontally transmitted retroviruses have been eliminated from commercially available mice but are identified in wild mice. Insertional mutagenesis with reintegration of endogenous retroviruses or transposition of retroelements has resulted in spontaneous mutations including some immune relevant ones such as Foxn1nu, Lepob, and Faslpr.439441 Mice infected with LP-BM5 (defective) murine leukemia virus develop murine acquired immunodeficiency syndrome and have been widely used as a preclinical model to study the pathogenesis of human retroviral infections (Table 5).442,443 Lifelong expression of viral proteins encoded by endogenous retroviruses/retroelements may be responsible for most of the spontaneous immune-mediated conditions observed in some inbred strains during aging, including glomerulonephritis and polyarteritis.440,444 Strain-specific variations in the composition and activity of endogenous retroviruses/retroelements and immune response against retroviral antigens also play a role in the susceptibility of specific mouse backgrounds to experimental autoimmune conditions including SLE and T1D.445448

While the parvoviruses, herpesviruses, and exogenous retroviruses have been eliminated from commercial sources of contemporary laboratory mice because of disease or other confounding effects on research, recent interest in the “normal” immunity of wild or pet store mice may render these agents, as well as historically important mouse disease problems and zoonotic concerns, more relevant.449,450

Biological Materials

Biological materials, including transplantable tumors, cell lines, serum, embryos, and gametes, can harbor a diversity of mouse viruses (parvoviruses, ectromelia virus, MHV, lactose dehydrogenase elevating virus, and retroviruses), human viruses, and bacteria, notoriously the Mycoplasmas.420,451,452 They therefore represent a substantial concern as a source of pathogens and microbial confounders, especially in studies that involve immunodeficient rodents. Reporting recommendations plead for QA of cell lines: genetic QA (authentication to confirm the identity of the cell lines), and microbial QA (to assure freedom from pathogens).453456

Unintended Consequences of Genetic Engineering Strategies

Genetic engineering strategies have immune effects that may have unintended or unexpected consequences for diverse research areas.

Cre/loxP-based DNA recombination technology is used for conditional (tissue-specific) gene targeting. The endonuclease activity of Cre recombinase, including the “illegitimate” targeting of the numerous pseudo-loxP sites across the mouse genome, results in the strong induction of an antiviral response. This is due to the recruitment of the specific cytosolic DNA sensor stimulator of interferon genes (STING), concurrent with Cre-dependent DNA damage and the accumulation of cytoplasmic DNA fragments. Given the primary role of STING in the activation of antiviral immune pathways (including type-I IFN), Cre expression can impact multiple immune parameters in Cre/loxP-based mouse models. Appropriate Cre-only controls may help in distinguishing signal from noise.457

The tamoxifen-inducible Cre/loxP system (Cre-ERT2) allows site- and time-specific gene targeting in the mouse. Tamoxifen has immune relevant effects, as well as toxic and genotoxic effects. The estrogen-dependent and -independent effects of tamoxifen have been demonstrated to promote a shift from a Th1- to a Th2-mediated immune responses. Such effects can especially impact allergy and autoimmune models involving activation of Th1-mediated immunity (e.g., EAE and some SLE models).458,459 Recently, oral ivermectin treatment has been specifically linked to the unintended activation of Cre-ERT2 system in T cells.460

Tetracycline-controlled transcriptional activation (Tet-Off/Tet-On) systems allow site-specific, reversible, and dose-dependent control of gene expression in mice. Doxycycline (a tetracycline derivative) is administered or withdrawn to regulate target gene expression. Doxycycline in mice interferes with and modulates immune and inflammatory responses relevant to allotransplant rejection, response to LPS, and neutrophil chemotaxis, among others.461463 Recent works have also unveiled the effect of doxycycline on murine gut microbiota and how the resulting dysbiosis might affect the immune response in diverse experimental settings.461463 DNA binding by tetracycline/doxycycline-controlled Tet-transactivator (tTA) and its reverse is apparently sufficient to induce apoptosis in activated lymphocytes. These findings indicate that a major experimental bias exists in the use of the Tet-On/Off system for lymphocyte targeting as the approach may (1) limit the extent of the adaptive immune reaction and (2) favor the outgrowth of apoptosis-resistant subpopulations of lymphoid cells.464

Expression of fluorescent or enzymatic reporters driven by gene-specific regulatory elements is used to study in vivo or ex vivo activity and distribution of specific molecular targets or mutant alleles in GEM models. However, an increasing number of studies show that reporters can be highly immunogenic. Indeed, response of the mouse immune system against classical reporter molecules (including enhanced green fluorescent protein, luciferase, and β-galactosidase) has been demonstrated. The inherent immunogenicity of reporter gene’s products depends on different factors including the mouse’s background strain as well as level of expression and tissue distribution/accumulation. It is therefore extremely important to consider carefully any potential variable associated with the use of genetic reporter systems for immunological studies in mice.465470

Even the most recent and sophisticated strategies for genome editing, including the revolutionary CRISPR-Cas9 system, have demonstrated experimental caveats influencing the immune system. In addition to the potential immunogenicity of viral vectors in viral delivery systems, human and mice have demonstrated preexisting adaptive immunity to Cas9 homologues expressed by common bacteria such as Staphylococcus aureus and Streptococcus pyogenes. The inherent immunogenicity of Cas9 is a concern not only for the preclinical application of the CRISPR-Cas9 system, but also for its potential clinical use as gene therapy strategy.471,472

Future Directions in Mouse Immunology

Human Surrogate/“Avatar” Approaches

Options to take advantage of humanized mice and other animals to study human derived immune elements in nonhuman surrogates are reviewed elsewhere. These present a diversity of opportunities for better understanding of human disease conditions as well as a number challenges that also may be informative if approached critically and scientifically.431,566,567 For a comprehensive overview on this topic, readers are encouraged to consult the contribution from Simons et al. in the present issue of the ILAR Journal.

Genetic Approaches

Options to take advantage of the spectrum of mouse genetic and immune diversity include factorial study design and Collaborative Cross (CC)-derived RI strains and Diversity Outbred (DO) mice. In a factorial study design, significance can be achieved with relatively small “n” from several strains selected for informative differences in immune relevant genotypes and phenotypes.568,569 Recognizing that an inbred strain represents an intentionally limited fraction of the spectrum of genetic variability of laboratory mice not designed or suited to model immunological endpoints at a population scale,570,571 the CC-derived RI strains represent the genetic variability across the 7 major families of mice and offer fairly new options for dissecting genetic and molecular mechanisms of immunity and disease.572,573 The CC is a mouse reference population with high allelic diversity constructed by a breeding strategy that systematically outcrosses 8 founder strains, followed by inbreeding to obtain new RI strains. Five of the 8 founder strains are “classical” laboratory strains including 129S1/SvImJ, A/J, C57BL/6J, NOD/ShiLtJ, and NZO/HlLtJ. Three founder strains are “wild-derived”: CAST/EiJ, PWK/PhJ, and WSB/EiJ. Currently available CC RI lines are distributed through consortia (e.g., http://csbio.unc.edu/CCstatus/index.py) and public repositories (e.g., https://www.jax.org/strain/027296). Since their inception, partially inbred CC mice have been characterized and compared for the identification of deviant immune traits or phenotypes. They have provided opportunities to study the evolution of complex genetic interactions.573 The application of immunogenomics and immunonogenetics techniques on CC mice has identified QTLs, polymorphic regions, and candidate genes that control mouse immunodiversity572 and have contributed to our understanding of susceptibilities to SARS coronavirus, West Nile virus, and Aspergillus fumigatus.574577 DO mice (https://www.jax.org/strain/009376) were developed by random outcross matings of 160 CC RI lines, and the breeding strategy of continued random matings is designed to maximize their genetic diversity.578581 The genetic heterogeneity of DO mice far exceeds that of genetically undefined mice, termed “outbred,” that derive from the Swiss branch of the mouse family tree (e.g., CD-1, CFW, ICR, ND4, NMRI, SW) originating from Clara Lynch’s original 9 albino mice brought to the United States from Switzerland in 1926. The genetic heterogeneity and heterozygosity among these mice is more limited and varies with their source.582,583 While the literature is still fairly limited on CC RI strains and the derived DO mice, these represent translational research tools that take advantage of mouse genetic variability to identify disease mechanisms, select novel drug targets, and discover associated biomarkers.

Microbial Approaches

There is recent interest in the use of genetically and microbially “wild-like” mice as a more human like or human relevant strategy.3,4,6,450,573,584 The studies make relevant and useful points about the naïve immune systems of “clean” C57BL/6 mice recently received from microbially restricted commercial sources. However, many mice bred in house in research institutions are not quite so naïve or microbially restricted.378,585587 Undefined or incompletely defined microbiota of pet store or feral mice raise concerns for infection related morbidity, mortality, and unpredictable experimental confounds as well as biosafety concerns related to zoonotic agents. Advances in gnotobiotics and microbiota characterization offer opportunities for defined and strategic approaches that will deliver important insights to immune modulation by autochthonous and allochthonous microflora.385,409,449,450

Conclusions

Mice have had important roles in advancing the field of immunology and fostering the development of new diagnostic and therapeutic avenues. Recognition of intrinsic and extrinsic contributors to immune phenotypes is crucial for the selection of more relevant and reproducible mouse models and generation of robust translational data. Known contributors can be intentionally used or intentionally avoided in the experimental system. Accurate reporting of animals and study conditions is mission critical to communicating biomedical research. Well-designed and reported research in mice has much to offer to our understanding of immunity and important diseases of humans and other species.

Supplementary Material

ily026_Suppl_Table-1_ER_06-05-2019
ily026_Suppl-Table-2_Revision_ER_06-06-2019

References

  • 1. Bryda EC. The mighty mouse: the impact of rodents on advances in biomedical research. Mo Med. 2013;110(3):207 http://www.ncbi.nlm.nih.gov/pubmed/23829104. [PMC free article] [PubMed] [Google Scholar]
  • 2. Justice MJ, Dhillon P. Using the mouse to model human disease: increasing validity and reproducibility. Dis Model Mech. 2016;9(2):101–103. 10.1242/dmm.024547. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3. Abolins S, King EC, Lazarou L, et al. . The comparative immunology of wild and laboratory mice, mus musculus domesticus. Nat Commun. 2017;8:14811 10.1038/ncomms14811. https://www.nature.com/articles/ncomms14811. Accessed Jan 5, 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4. Cauwels A, Vandendriessche B, Brouckaert P. Of mice, men, and inflammation. Proc Natl Acad Sci USA. 2013;110(34):E3150 10.1073/pnas.1308333110. http://www.pnas.org/content/110/34/E3150.extract. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5. Perlman RL. Mouse models of human disease: an evolutionary perspective. Evol Med Public Health. 2016;2016(1):170 http://www.ncbi.nlm.nih.gov/pubmed/27121451. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6. Mestas J, Hughes CCW. Of mice and not men: differences between mouse and human immunology. J Immunol. 2004;172(5):2731–2738. [DOI] [PubMed] [Google Scholar]
  • 7. Sellers RS. Translating mouse models. Toxicol Pathol. 2017;45(1):134–145. 10.1177/0192623316675767. [DOI] [PubMed] [Google Scholar]
  • 8. Sundberg JP, Schofield PN. Commentary: mouse genetic nomenclature. Standardization of strain, gene, and protein symbols. Vet Pathol. 2010;47(6):1100–1104. 10.1177/0300985810374837. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9. Fontaine DA, Davis DB. Attention to background strain is essential for metabolic research: C57BL/6 and the international knockout mouse consortium. Diabetes. 2016;65(1):25–33. 10.2337/db15-0982. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10. Omary MB, Cohen DE, El-Omar EM, et al. . Not all mice are the same: standardization of animal research data presentation. Gastroenterology. 2016;150(7):1503–1504. 10.1053/j.gastro.2016.03.034. [DOI] [PubMed] [Google Scholar]
  • 11. Dunn LC, Grüneberg H, Snell GD. Report of the committee on mouse genetics nomenclature. J Hered. 1940;31(12):505–506. 10.1093/oxfordjournals.jhered.a104827. https://academic.oup.com/jhered/article/31/12/505/889124. Accessed May 8, 2018. [DOI] [Google Scholar]
  • 12. Carter TC, Dunn LC, Falconer DS, Grüneberg H, Heston WE, Snell GD. Standardized nomenclature for inbred strains of mice. Cancer Res. 1952;12(8):602–613. [PubMed] [Google Scholar]
  • 13. Green MC, Grueneberg H, Hertwig P, et al. . A revision of the standardized genetic nomenclature for mice. J Hered. 1963;54:159–162. [PubMed] [Google Scholar]
  • 14. Staats J. Standardized nomenclature for inbred strains of mice: third listing. Cancer Res. 1964;24:147–168. [PubMed] [Google Scholar]
  • 15. Staats J. Standardized nomenclature for inbred strains of mice: fourth listing. Cancer Res. 1968;28(3):391–420. [PubMed] [Google Scholar]
  • 16. Staats J. Standardized nomenclature for inbred strains of mice: fifth listing. Cancer Res. 1972;32(8):1609–1646. [PubMed] [Google Scholar]
  • 17. Staats J. Standardized nomenclature for inbred strains of mice: sixth listing. Cancer Res. 1976;36(12):4333–4377. [PubMed] [Google Scholar]
  • 18. Staats J. Standardized nomenclature for inbred strains of mice: seventh listing for the international committee on standardized genetic nomenclature for mice. Cancer Res. 1980;40(7):2083–2128. [PubMed] [Google Scholar]
  • 19. Staats J. Standardized nomenclature for inbred strains of mice: eighth listing. Cancer Res. 1985;45(3):945–977. [PubMed] [Google Scholar]
  • 20. Festing M, Kondo K, Loosli R, Poiley SM, Spiegel A. International standardized nomenclature for outbred stocks of laboratory animals. Z Versuchstierkd. 1972;14(4):215–224. [PubMed] [Google Scholar]
  • 21. [NRC] National Research Council Guidance for the description of animal research in scientific publications. Washington (DC): National Academies Press (US); 2011. http://www.ncbi.nlm.nih.gov/books/NBK84205/. Accessed May 10, 2018. [PubMed] [Google Scholar]
  • 22. Everitt JI. The future of preclinical animal models in pharmaceutical discovery and development: a need to bring in cerebro to the in vivo discussions. Toxicol Pathol. 2015;43(1):70–77. 10.1177/0192623314555162. [DOI] [PubMed] [Google Scholar]
  • 23. Kilkenny C, Browne WJ, Cuthill IC, Emerson M, Altman DG. Improving bioscience research reporting: the ARRIVE guidelines for reporting animal research. PLoS Biol. 2010;8(6):e1000412 10.1371/journal.pbio.1000412. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24. Doran AG, Wong K, Flint J, Adams DJ, Hunter KW, Keane TM. Deep genome sequencing and variation analysis of 13 inbred mouse strains defines candidate phenotypic alleles, private variation and homozygous truncating mutations. Genome Biol. 2016;17(1):167 10.1186/s13059-016-1024-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25. Adams DJ, Doran AG, Lilue J, Keane TM. The mouse genomes project: a repository of inbred laboratory mouse strain genomes. Mamm Genome. 2015;26(9–10):403–412. 10.1007/s00335-015-9579-6. [DOI] [PubMed] [Google Scholar]
  • 26. Rahim MMA, Tu MM, Mahmoud AB, et al. . Ly49 receptors: innate and adaptive immune paradigms. Front Immunol. 2014;5:145 10.3389/fimmu.2014.00145. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27. Sellers RS, Clifford CB, Treuting PM, Brayton C. Immunological variation between inbred laboratory mouse strains: points to consider in phenotyping genetically immunomodified mice. Vet Pathol. 2012;49(1):32–43. 10.1177/0300985811429314. https://www.ncbi.nlm.nih.gov/pubmed/22135019. Accessed May 3, 2018. [DOI] [PubMed] [Google Scholar]
  • 28. Liu J, Karypis G, Hippen KL, et al. . Genomic view of systemic autoimmunity in MRLlpr mice. Genes Immun. 2006;7(2):156–168. 10.1038/sj.gene.6364286. http://www.ncbi.nlm.nih.gov/pubmed/16508641. [DOI] [PubMed] [Google Scholar]
  • 29. Nose M, Nishihara M, Kamogawa J, Terada M, Nakatsuru S. Genetic basis of autoimmune disease in MRL/lpr mice: dissection of the complex pathological manifestations and their susceptibility loci. Rev Immunogenet. 2000;2(1):154–164. [PubMed] [Google Scholar]
  • 30. Yamada A, Miyazaki T, Lu L, et al. . Genetic basis of tissue specificity of vasculitis in MRL/lpr mice. Arthritis Rheum. 2003;48(5):1445–1451. 10.1002/art.10952. [DOI] [PubMed] [Google Scholar]
  • 31. Vidal S, Kono DH, Theofilopoulos AN. Loci predisposing to autoimmunity in MRL-fas lpr and C57BL/6-faslpr mice. J Clin Invest. 1998;101(3):696–702. 10.1172/JCI1817. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. Takahashi S, Fossati L, Iwamoto M, et al. . Imbalance towards Th1 predominance is associated with acceleration of lupus-like autoimmune syndrome in MRL mice. J Clin Invest. 1996;97(7):1597–1604. 10.1172/JCI118584. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33. Xu M, Hou R, Sato-Hayashizaki A, et al. . Cd72(c) is a modifier gene that regulates fas(lpr)-induced autoimmune disease. J Immunol. 2013;190(11):5436–5445. 10.4049/jimmunol.1203576. [DOI] [PubMed] [Google Scholar]
  • 34. Hogarth MB, Slingsby JH, Allen PJ, et al. . Multiple lupus susceptibility loci map to chromosome 1 in BXSB mice. J Immunol. 1998;161(6):2753–2761. [PubMed] [Google Scholar]
  • 35. Kimura J, Ichii O, Nakamura T, Horino T, Otsuka S, Kon Y. BXSB-type genome causes murine autoimmune glomerulonephritis: pathological correlation between telomeric region of chromosome 1 and yaa. Genes Immun. 2014;15(3):182–189. 10.1038/gene.2014.4. [DOI] [PubMed] [Google Scholar]
  • 36. Choi Y, Simon-Stoos K, Puck JM. Hypo-active variant of IL-2 and associated decreased T cell activation contribute to impaired apoptosis in autoimmune prone MRL mice. Eur J Immunol. 2002;32(3):677–685. . [DOI] [PubMed] [Google Scholar]
  • 37. Poland A, Glover E. Characterization and strain distribution pattern of the murine ah receptor specified by the ahd and ahb-3 alleles. Mol Pharmacol. 1990;38(3):306–312. [PubMed] [Google Scholar]
  • 38. De Souza VR, Cabrera WK, Galvan A, et al. . Aryl hydrocarbon receptor polymorphism modulates DMBA-induced inflammation and carcinogenesis in phenotypically selected mice. Int J Cancer. 2009;124(6):1478–1482. [DOI] [PubMed] [Google Scholar]
  • 39. Esser C, Rannug A, Stockinger B. The aryl hydrocarbon receptor in immunity. Trends Immunol. 2009;30(9):447–454. [DOI] [PubMed] [Google Scholar]
  • 40. Esser C. The immune phenotype of AhR null mouse mutants: not a simple mirror of xenobiotic receptor over-activation. Biochem Pharmacol. 2009;77:597–607. http://www.sciencedirect.com/science/article/pii/S0006295208007193. [DOI] [PubMed] [Google Scholar]
  • 41. Tulone C, Tsang J, Prokopowicz Z, Grosvenor N, Chain B.. Natural cathepsin E deficiency in the immune system of C57BL/6J mice. Immunogenetics. 2007;59(12):927–935. [DOI] [PubMed] [Google Scholar]
  • 42. Wetsel RA, Fleischer DT, Haviland DL. Deficiency of the murine fifth complement component (C5). A 2-base pair gene deletion in a 5’-exon. J Biol Chem. 1990;265(5):2435–2440. [PubMed] [Google Scholar]
  • 43. Skerka C, Chen Q, Fremeaux-Bacchi V, Roumenina LT. Complement factor H related proteins (CFHRs). Mol Immunol. 2013;56:170–180. http://www.sciencedirect.com/science/article/pii/S0161589013004185. [DOI] [PubMed] [Google Scholar]
  • 44. Encinas JA, Wicker LS, Peterson LB, et al. . QTL influencing autoimmune diabetes and encephalomyelitis map to a 0.15-cM region containing Il2. Nat Genet. 1999;21(2):158–160. [DOI] [PubMed] [Google Scholar]
  • 45. Javan MR, Shahraki S, Safa A, Zamani MR, Salmaninejad A, Aslani S. An interleukin 12 B single nucleotide polymorphism increases IL-12p40 production and is associated with increased disease susceptibility in patients with relapsing-remitting multiple sclerosis. Neurol Res. 2017;39(5):435–441. [DOI] [PubMed] [Google Scholar]
  • 46. Wen X, Chen S, Li P, et al. . Single nucleotide polymorphisms of IL12B are associated with takayasu arteritis in Chinese Han population. Rheumatol Int. 2017;37(4):547–555. [DOI] [PubMed] [Google Scholar]
  • 47. Zwiers A, Fuss IJ, Seegers D, et al. . A polymorphism in the coding region of Il12b promotes IL-12p70 and IL-23 heterodimer formation. J Immunol. 2011;186(6):3572–3580. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48. Staeheli P, Grob R, Meier E, Sutcliffe JG, Haller O. Influenza virus-susceptible mice carry mx genes with a large deletion or a nonsense mutation. Mol Cell Biol. 1988;8:4518–4523. http://mcb.asm.org/content/8/10/4518.abstract. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49. Verhelst J, Spitaels J, Nurnberger C, et al. . Functional comparison of Mx1 from two different mouse species reveals the involvement of loop L4 in the antiviral activity against influenza A viruses. J Virol. 2015;89(21):10879–10890. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50. Lightfield KL, Persson J, Brubaker SW, et al. . Critical function for Naip5 in inflammasome activation by a conserved carboxy-terminal domain of flagellin. Nat Immunol. 2008;9(10):1171–1178. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51. Tenthorey JL, Haloupek N, Lopez-Blanco JR, et al. . The structural basis of flagellin detection by NAIP5: a strategy to limit pathogen immune evasion. Science. 2017;358(6365):888–893. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52. Fortier A, Min-Oo G, Forbes J, Lam-Yuk-Tseung S, Gros P. Single gene effects in mouse models of host: pathogen interactions. J Leukoc Biol. 2005;77(6):868–877. [DOI] [PubMed] [Google Scholar]
  • 53. Moayeri M, Crown D, Newman ZL, et al. . Inflammasome sensor Nlrp1b-dependent resistance to anthrax is mediated by caspase-1, IL-1 signaling and neutrophil recruitment. PLoS Pathog. 2010;6(2):e1001222 10.1371/journal.ppat.1001222. http://www.ncbi.nlm.nih.gov/pubmed/21170303. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54. Tschopp J, Martinon F, Burns K. NALPs: a novel protein family involved in inflammation. Nat Rev Mol Cell Biol. 2003;4(2):95–104. [DOI] [PubMed] [Google Scholar]
  • 55. Ulland TK, Jain N, Hornick EE, et al. . Nlrp12 mutation causes C57BL/6J strain-specific defect in neutrophil recruitment. Nat Commun. 2016;7:13180 10.1038/ncomms13180. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56. Chen L, Wilson JE, Koenigsknecht MJ, et al. . NLRP12 attenuates colon inflammation by maintaining colonic microbial diversity and promoting protective commensal bacterial growth. Nat Immunol. 2017;18(5):541–551. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57. Courtney SC, Di H, Stockman BM, Liu H, Scherbik SV, Brinton MA. Identification of novel host cell binding partners of Oas1b, the protein conferring resistance to flavivirus-induced disease in mice. J Virol. 2012;86(15):7953–7963. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58. Kwong LS, Brown MH, Barclay AN, Hatherley D. Signal-regulatory protein alpha from the NOD mouse binds human CD47 with an exceptionally high affinity—implications for engraftment of human cells. Immunology. 2014;143(1):61–67. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59. Iwamoto C, Takenaka K, Urata S, et al. . The BALB/c-specific polymorphic SIRPA enhances its affinity for human CD47, inhibiting phagocytosis against human cells to promote xenogeneic engraftment. Exp Hematol. 2014;42(3):171.e1 10.1016/j.exphem.2013.11.005. [DOI] [PubMed] [Google Scholar]
  • 60. Detre C, Keszei M, Romero X, Tsokos GC, Terhorst C. SLAM family receptors and the SLAM-associated protein (SAP) modulate T cell functions. Semin Immunopathol. 2010;32:157–171. http://www.ncbi.nlm.nih.gov/pmc/articles/PMC2868096/. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61. Keszei M, Latchman YE, Vanguri VK, et al. . Auto-antibody production and glomerulonephritis in congenic Slamf1-/- and Slamf2-/- B6.129] but not in Slamf1-/- and Slamf2-/- BALB/c.129] mice. Int Immunol. 2011;23(2):149–158. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62. Koh AE, Njoroge SW, Feliu M, et al. . The SLAM family member CD48 (Slamf2) protects lupus-prone mice from autoimmune nephritis. J Autoimmun. 2011;37:48–57. http://www.ncbi.nlm.nih.gov/pubmed/21561736. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63. Crampton SP, Morawski PA, Bolland S. Linking susceptibility genes and pathogenesis mechanisms using mouse models of systemic lupus erythematosus. Dis Model Mech. 2014;7(9):1033–1046. 10.1242/dmm.016451. http://www.ncbi.nlm.nih.gov/pmc/articles/PMC4142724/. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64. Stober CB, Brode S, White JK, Popoff JF, Blackwell JM. Slc11a1, formerly Nramp1, is expressed in dendritic cells and influences major histocompatibility complex class II expression and antigen-presenting cell function. Infect Immun. 2007;75(10):5059–5067. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65. Osman GE, Hannibal MC, Anderson JP, Lasky SR, Ladiges WC, Hood L. FVB/N (H2(q)) mouse is resistant to arthritis induction and exhibits a genomic deletion of T-cell receptor V beta gene segments. Immunogenetics. 1999;49(10):851–859. [DOI] [PubMed] [Google Scholar]
  • 66. Osman GE, Hannibal MC, Anderson JP, et al. . T-cell receptor vbeta deletion and valpha polymorphism are responsible for the resistance of SWR mouse to arthritis induction. Immunogenetics. 1999;49(9):764–772. [DOI] [PubMed] [Google Scholar]
  • 67. Behlke MA, Chou HS, Huppi K, Loh DY. Murine T-cell receptor mutants with deletions of beta-chain variable region genes. Proc Natl Acad Sci USA. 1986;83(3):767–771. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68. Poltorak A, He X, Smirnova I, et al. . Defective LPS signaling in C3H/HeJ and C57BL/10ScCr mice: mutations in Tlr4 gene. Science. 1998;282(5396):2085–2088. [DOI] [PubMed] [Google Scholar]
  • 69. Poltorak A, Smirnova I, He X, et al. . Genetic and physical mapping of the lps locus: identification of the toll-4 receptor as a candidate gene in the critical region. Blood Cells Mol Dis. 1998;24(3):340–355. [DOI] [PubMed] [Google Scholar]
  • 70. Liaunardy-Jopeace A, Gay NJ. Molecular and cellular regulation of toll-like receptor-4 activity induced by lipopolysaccharide ligands. Front Immunol. 2014;5:473. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71. De Vooght V, Vanoirbeek JA, Luyts K, Haenen S, Nemery B, Hoet PH. Choice of mouse strain influences the outcome in a mouse model of chemical-induced asthma. PLoS One. 2010;5:e12581 http://www.ncbi.nlm.nih.gov/pubmed/20830207. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72. Watanabe H, Numata K, Ito T, Takagi K, Matsukawa A. Innate immune response in Th1- and Th2-dominant mouse strains. Shock. 2004;22(5):460–466. [DOI] [PubMed] [Google Scholar]
  • 73. Simon MM, Greenaway S, White JK, et al. . A comparative phenotypic and genomic analysis of C57BL/6J and C57BL/6N mouse strains. Genome Biol. 2013;14(7):R82 10.1186/gb-2013-14-7-r82. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74. Zurita E, Chagoyen M, Cantero M, et al. . Genetic polymorphisms among C57BL/6 mouse inbred strains. Transgenic Res. 2011;20(3):481–489. 10.1007/s11248-010-9403-8. [DOI] [PubMed] [Google Scholar]
  • 75. Mekada K, Abe K, Murakami A, et al. . Genetic differences among C57BL/6 substrains. Exp Anim. 2009;58(2):141–149. [DOI] [PubMed] [Google Scholar]
  • 76. Mekada K, Hirose M, Murakami A, Yoshiki A. Development of SNP markers for C57BL/6N-derived mouse inbred strains. Exp Anim. 2015;64(1):91–100. 10.1538/expanim.14-0061. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77. Mahajan VS, Demissie E, Mattoo H, et al. . Striking immune phenotypes in gene-targeted mice are driven by a copy-number variant originating from a commercially available C57BL/6 strain. Cell Rep. 2016;15(9):1901–1909. 10.1016/j.celrep.2016.04.080. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78. Hedlund M, Tangvoranuntakul P, Takematsu H, et al. . N-glycolylneuraminic acid deficiency in mice: implications for human biology and evolution. Mol Cell Biol. 2007;27(12):4340–4346. 10.1128/MCB.00379-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79. Purtha WE, Swiecki M, Colonna M, Diamond MS, Bhattacharya D. Spontaneous mutation of the Dock2 gene in Irf5-/- mice complicates interpretation of type I interferon production and antibody responses. Proc Natl Acad Sci USA. 2012;109(15):898 10.1073/pnas.1118155109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80. Freeman HC, Hugill A, Dear NT, Ashcroft FM, Cox RD. Deletion of nicotinamide nucleotide transhydrogenase: a new quantitive trait locus accounting for glucose intolerance in C57BL/6J mice. Diabetes. 2006;55(7):2153–2156. 10.2337/db06-0358. [DOI] [PubMed] [Google Scholar]
  • 81. Ronchi JA, Figueira TR, Ravagnani FG, Oliveira HCF, Vercesi AE, Castilho RF. A spontaneous mutation in the nicotinamide nucleotide transhydrogenase gene of C57BL/6J mice results in mitochondrial redox abnormalities. Free Radic Biol Med. 2013;63:446–456. 10.1016/j.freeradbiomed.2013.05.049. [DOI] [PubMed] [Google Scholar]
  • 82. Liron T, Raphael B, Hiram-Bab S, Bab IA, Gabet Y. Bone loss in C57BL/6J-OlaHsd mice, a substrain of C57BL/6J carrying mutated alpha-synuclein and multimerin-1 genes. J Cell Physiol. 2018;233(1):371–377. 10.1002/jcp.25895. [DOI] [PubMed] [Google Scholar]
  • 83. Specht CG, Schoepfer R. Deletion of the alpha-synuclein locus in a subpopulation of C57BL/6J inbred mice. BMC Neurosci. 2001;2(1):11 10.1186/1471-2202-2-11. http://www.ncbi.nlm.nih.gov/pubmed/11591219. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84. López‐Jiménez A, Walter NAR, Giné E, et al. . A spontaneous deletion of α‐Synuclein is associated with an increase in CB1 mRNA transcript and receptor expression in the hippocampus and amygdala: effects on alcohol consumption. Synapse. 2013;67(6):280–289. 10.1002/syn.21639. https://onlinelibrary.wiley.com/doi/abs/10.1002/syn.21639. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86. Mattapallil MJ, Wawrousek EF, Chan C, et al. . The Rd8 mutation of the Crb1 gene is present in vendor lines of C57BL/6N mice and embryonic stem cells, and confounds ocular induced mutant phenotypes. Invest Ophthalmol Vis Sci. 2012;53(6):2921–2927. 10.1167/iovs.12-9662. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86. Mehalow AK, Kameya S, Smith RS, et al. . CRB1 is essential for external limiting membrane integrity and photoreceptor morphogenesis in the mammalian retina. Hum Mol Genet. 2003;12(17):2179–2189. 10.1093/hmg/ddg232. [DOI] [PubMed] [Google Scholar]
  • 87. Davisson MT, Bergstrom DE, Reinholdt LG, Donahue LR. Discovery genetics—the history and future of spontaneous mutation research. Curr Protoc Mouse Biol. 2012;2:103–118. 10.1002/9780470942390.mo110200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88. Davisson MT. Discovery genetics: serendipity in basic research. ILAR J. 2005;46(4):338–345. [DOI] [PubMed] [Google Scholar]
  • 89. [NRC] National Research Council Immunodeficient rodents: a guide to their immunobiology, husbandry, and use. Washington (DC): National Academies Press (US); 1989. http://www.ncbi.nlm.nih.gov/books/NBK218200/. Accessed Nov 16, 2017. [PubMed] [Google Scholar]
  • 90. Montagutelli X. Effect of the genetic background on the phenotype of mouse mutations. J Am Soc Nephrol. 2000;11(Suppl 16):101. [PubMed] [Google Scholar]
  • 91. Zeineldin M, Jensen D, Paranjape SR, et al. . Human cancer xenografts in outbred nude mice can be confounded by polymorphisms in a modifier of tumorigenesis. Genetics. 2014;197(4):1365–1376. 10.1534/genetics.114.166587. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92. Nonoyama S, Ochs HD. Immune deficiency in SCID mice. Int Rev Immunol. 1996;13(4):289–300. [DOI] [PubMed] [Google Scholar]
  • 93. Nonoyama S, Smith FO, Bernstein ID, Ochs HD. Strain-dependent leakiness of mice with severe combined immune deficiency. J Immunol. 1993;150(9):3817–3824. [PubMed] [Google Scholar]
  • 84. Hoshino K, Takeuchi O, Kawai T, et al. . Cutting edge: Toll-like receptor 4 (TLR4)-deficient mice are hyporesponsive to lipopolysaccharide: evidence for TLR4 as the lps gene product. J Immunol. 1999;162(7):3749–3752. [PubMed] [Google Scholar]
  • 95. Shultz LD, Schweitzer PA, Christianson SW, et al. . Multiple defects in innate and adaptive immunologic function in NOD/LtSz-scid mice. J Immunol. 1995;154(1):180–191. [PubMed] [Google Scholar]
  • 96. Caplazi P, Baca M, Barck K, et al. . Mouse models of rheumatoid arthritis. Vet Pathol. 2015;52:819–826. http://www.ncbi.nlm.nih.gov/pubmed/26063174. [DOI] [PubMed] [Google Scholar]
  • 97. Pearson JA, Wong FS, Wen L. The importance of the non obese diabetic (NOD) mouse model in autoimmune diabetes. J Autoimmun. 2016;66:76–88. 10.1016/j.jaut.2015.08.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98. Constantinescu CS, Farooqi N, O’Brien K, Gran B. Experimental autoimmune encephalomyelitis (EAE) as a model for multiple sclerosis (MS). Br J Pharmacol. 2011;164:1079–1106. http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3229753/. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99. Rottman JB, Willis CR. Mouse models of systemic lupus erythematosus reveal a complex pathogenesis. Vet Pathol. 2010;47(4):664–676. 10.1177/0300985810370005. http://journals.sagepub.com/doi/full/10.1177/0300985810370005. [DOI] [PubMed] [Google Scholar]
  • 100. Krishnamurthy B, Dudek NL, McKenzie MD, et al. . Responses against islet antigens in NOD mice are prevented by tolerance to proinsulin but not IGRP. J Clin Invest. 2006;116(12):3258–3265. 10.1172/JCI29602. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101. Stadinski BD, Delong T, Reisdorph N, et al. . Chromogranin A is an autoantigen in type 1 diabetes. Nat Immunol. 2010;11(3):225–231. 10.1038/ni.1844. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102. Unanue ER, Ferris ST, Carrero JA. The role of islet antigen presenting cells and the presentation of insulin in the initiation of autoimmune diabetes in the NOD mouse. Immunol Rev. 2016;272(1):183–201. 10.1111/imr.12430. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103. Noble JA, Erlich HA. Genetics of type 1 diabetes. Cold Spring Harb Perspect Med. 2012;2:a007732 http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3253030/. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104. Dwyer CJ, Bayer AL, Fotino C, et al. . Altered homeostasis and development of regulatory T cell subsets represent an IL-2R-dependent risk for diabetes in NOD mice. Sci Signal. 2017;10(510). https://www.ncbi.nlm.nih.gov/pubmed/29259102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105. Wong AS, Mortin-Toth S, Sung M, et al. . Polymorphism in the innate immune receptor SIRPalpha controls CD47 binding and autoimmunity in the nonobese diabetic mouse. J Immunol. 2014;193(10):4833–4844. [DOI] [PubMed] [Google Scholar]
  • 106. Suri A, Walters JJ, Gross ML, Unanue ER. Natural peptides selected by diabetogenic DQ8 and murine I-A(g7) molecules show common sequence specificity. J Clin Invest. 2005;115:2268–2276. http://www.ncbi.nlm.nih.gov/pmc/articles/PMC1180544/. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107. Hattori M, Buse JB, Jackson RA, et al. . The NOD mouse: recessive diabetogenic gene in the major histocompatibility complex. Science. 1986;231:733–735. http://science.sciencemag.org/content/sci/231/4739/733.full.pdf. [DOI] [PubMed] [Google Scholar]
  • 108. Dai H, Friday AJ, Abou-Daya KI, et al. . Donor SIRPα polymorphism modulates the innate immune response to allogeneic grafts. Sci Immunol. 2017;2(12). doi: 10.1126/sciimmunol.aam6202. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109. Mehta G, Ferreira VP, Pickering MC, Skerka C, Zipfel PF, Banda NK. New insights into disease-specific absence of complement factor H related protein C in mouse models of spontaneous autoimmune diseases. Mol Immunol. 2014;62:235–248. http://www.ncbi.nlm.nih.gov/pmc/articles/PMC4157073/. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110. Zekavat G, Mozaffari R, Arias VJ, et al. . A novel CD93 polymorphism in non-obese diabetic (NOD) and NZB/W F1 mice is linked to a CD4+ iNKT cell deficient state. Immunogenetics. 2010;62:397–407. http://www.ncbi.nlm.nih.gov/pmc/articles/PMC2875467/. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111. Simecek P, Churchill GA, Yang H, et al. . Genetic analysis of substrain divergence in non-obese diabetic (NOD) mice. G3 (Bethesda). 2015;5:771–775. http://www.ncbi.nlm.nih.gov/pmc/articles/PMC4426365/. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112. Celhar T, Fairhurst A. Toll-like receptors in systemic lupus erythematosus: potential for personalized treatment. Front Pharmacol. 2014;5:265 10.3389/fphar.2014.00265. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113. Perry D, Sang A, Yin Y, Zheng YY, Morel L. Murine models of systemic lupus erythematosus. J Biomed Biotechnol. 2011;2011:271694. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114. Celhar T, Fairhurst A. Modelling clinical systemic lupus erythematosus: similarities, differences and success stories. Rheumatology (Oxford). 2017;56(suppl_1):i88–i99. 10.1093/rheumatology/kew400. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115. Andrews BS, Eisenberg RA, Theofilopoulos AN, et al. . Spontaneous murine lupus-like syndromes. clinical and immunopathological manifestations in several strains. J Exp Med. 1978;148(5):1198–1215. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116. Santiago-Raber M, Laporte C, Reininger L, Izui S. Genetic basis of murine lupus. Autoimmun Rev. 2004;3(1):33–39. 10.1016/S1568-9972(03)00062-4. [DOI] [PubMed] [Google Scholar]
  • 117. Górnikiewicz B, Ronowicz A, Madanecki P, Sachadyn P. Genome-wide DNA methylation profiling of the regenerative MRL/MpJ mouse and two normal strains. Epigenomics. 2017;9(8):1105–1122. 10.2217/epi-2017-0009. [DOI] [PubMed] [Google Scholar]
  • 118. Heber-Katz E. Oxygen, metabolism, and regeneration: lessons from mice. Trends Mol Med. 2017;23(11):1024–1036. 10.1016/j.molmed.2017.08.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119. McBrearty BA, Clark LD, Zhang XM, Blankenhorn EP, Heber-Katz E. Genetic analysis of a mammalian wound-healing trait. Proc Natl Acad Sci USA. 1998;95(20):11792–11797. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120. Nose M, Nishimura M, Ito MR, Toh J, Shibata T, Sugisaki T. Arteritis in a novel congenic strain of mice derived from MRL/lpr lupus mice: genetic dissociation from glomerulonephritis and limited autoantibody production. Am J Pathol. 1996;149(5):1763–1769. [PMC free article] [PubMed] [Google Scholar]
  • 121. Ghoreishi M, Dutz JP. Murine models of cutaneous involvement in lupus erythematosus. Autoimmun Rev. 2009;8(6):484–487. 10.1016/j.autrev.2009.02.028. [DOI] [PubMed] [Google Scholar]
  • 122. Nose M, Komori H, Miyazaki T, Mori S. Genomics of vasculitis: lessons from mouse models. Ann Vasc Dis. 2013;6(1):16–21. 10.3400/avd.oa.12.00096. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123. Nose M, Nishihara M, Fujii H. Genetic basis of the complex pathological manifestations of collagen disease: lessons from MRL/lpr and related mouse models. Int Rev Immunol. 2000;19(4–5):473–498. [DOI] [PubMed] [Google Scholar]
  • 124. Morse HC, Roths JB, Davidson WF, Langdon WY, Fredrickson TN, Hartley JW. Abnormalities induced by the mutant gene, lpr. patterns of disease and expression of murine leukemia viruses in SJL/J mice homozygous and heterozygous for lpr. J Exp Med. 1985;161:602–616. http://www.ncbi.nlm.nih.gov/pubmed/2982991. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Lane PW, Murphy ED. Susceptibility to spontaneous pneumonitis in an inbred strain of beige and satin mice. Genetics. 1972;72(3):451–460. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Murphy ED, Rohs JB. A Y chromosome associated factor in strain BXSB producing accelerated autoimmunity and lymphoproliferation. Arthritis Rheum. 1979;22(11):1188–1194. [DOI] [PubMed] [Google Scholar]
  • 127. Fairhurst AM, Hwang SH, Wang A, et al. . Yaa autoimmune phenotypes are conferred by overexpression of TLR7. Eur J Immunol. 2008;38(7):1971–1978. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128. Santiago-Raber ML, Kikuchi S, Borel P, et al. . Evidence for genes in addition to Tlr7 in the yaa translocation linked with acceleration of systemic lupus erythematosus. J Immunol. 2008;181(2):1556–1562. [DOI] [PubMed] [Google Scholar]
  • 129. Jain S, Ward JM, Shin D, et al. . Associations of autoimmunity, immunodeficiency, lymphomagenesis, and gut microbiota in mice with knockins for a pathogenic autoantibody. Am J Pathol. 2017;187(9):2020–2033. 10.1016/j.ajpath.2017.05.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130. East J, de Sousa MA, Parrott DM. Immunopathology of New Zealand black (NZB) mice. Transplantation. 1965;3(6):711–729. [DOI] [PubMed] [Google Scholar]
  • 131. East J, Branca M. Autoimmune reactions and malignant changes in germ-free New Zealand black mice. Clin Exp Immunol. 1969;4(6):621–635. [PMC free article] [PubMed] [Google Scholar]
  • 132. Beck-Engeser GB, Ahrends T, Knittel G, et al. . Infectivity and insertional mutagenesis of endogenous retrovirus in autoimmune NZB and B/W mice. J Gen Virol. 2015;96(11):3396–3410. 10.1099/jgv.0.000271. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133. Salerno E, Yuan Y, Scaglione BJ, et al. . The New Zealand black mouse as a model for the development and progression of chronic lymphocytic leukemia. Cytometry B Clin Cytom. 2010;78(Suppl 1):98 10.1002/cyto.b.20544. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134. Scatizzi JC, Haraldsson MK, Pollard KM, Theofilopoulos AN, Kono DH. The Lbw2 locus promotes autoimmune hemolytic anemia. J Immunol. 2012;188(7):3307–3314. 10.4049/jimmunol.1103561. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135. Kelley VE, Winkelstein A. Age- and sex-related glomerulonephritis in New Zealand white mice. Clin Immunol Immunopathol. 1980;16(2):142–150. [DOI] [PubMed] [Google Scholar]
  • 136. Mellors RC. Autoimmune and immunoproliferative diseases of NZB/bl mice and hybrids. Int Rev Exp Pathol. 1966;5:217–252. [PubMed] [Google Scholar]
  • 137. Dubois EL, Horowitz RE, Demopoulos HB, Teplitz R. NZB/NZW mice as a model of systemic lupus erythematosus. JAMA. 1966;195(4):285–289. [PubMed] [Google Scholar]
  • 138. Morel L, Wakeland EK. Susceptibility to lupus nephritis in the NZB/W model system. Curr Opin Immunol. 1998;10(6):718–725. [DOI] [PubMed] [Google Scholar]
  • 139. Rahman ZSM, Tin S, Buenaventura PL, et al. . A novel susceptibility locus on chromosome 2 in the (New Zealand black x New Zealand white)F1 hybrid mouse model of systemic lupus erythematosus. J Immunol. 2002;168(6):3042–3049. [DOI] [PubMed] [Google Scholar]
  • 140. Rudofsky UH, Lawrence DA. New Zealand mixed mice: a genetic systemic lupus erythematosus model for assessing environmental effects. Environ Health Perspect. 1999;107(Suppl 5):713–721. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141. Morel L. Mapping lupus susceptibility genes in the NZM2410 mouse model. Adv Immunol. 2012;115:113–139. [DOI] [PubMed] [Google Scholar]
  • 142. Waters ST, Fu SM, Gaskin F, et al. . NZM2328: a new mouse model of systemic lupus erythematosus with unique genetic susceptibility loci. Clin Immunol. 2001;100:372–383. http://www.sciencedirect.com/science/article/pii/S152166160195079X. Accessed Dec 1, 2018. [DOI] [PubMed] [Google Scholar]
  • 143. Moresco EMY, Li X, Beutler B. Going forward with genetics. Am J Pathol. 2013;182(5):1462–1473. 10.1016/j.ajpath.2013.02.002. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3644711/. Accessed Dec 1, 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144. Coutinho A, Meo T. Genetic basis for unresponsiveness to lipopolysaccharide in C57BL/10Cr mice. Immunogenetics. 1978;7(1):17–24. 10.1007/BF01843983. [DOI] [PubMed] [Google Scholar]
  • 145. Sakaguchi S, Wing K, Miyara M. Regulatory T cells—a brief history and perspective. Eur J Immunol. 2007;37(Suppl 1):S116–S123 10.1002/eji.200737593. http://www.ncbi.nlm.nih.gov/pubmed/17972355. Accessed Dec 1, 2018. [DOI] [PubMed] [Google Scholar]
  • 146. McClellan J, Macasocal R, Hare T, Horn M. Characterization of immunodeficient mouse models. FASEB J. 2017;31(1_suppl):807.14 10.1096/fasebj.31.1_supplement.807.14. https://www.fasebj.org/doi/abs/10.1096/fasebj.31.1_supplement.807.14. Accessed Jun 7, 2018. [DOI] [Google Scholar]
  • 147. Joliat MJ, Shultz LD. The molecular bases of spontaneous immunological mutations in the mouse and their homologous human diseases. Clin Immunol. 2001;101(2):113–129. 10.1006/clim.2001.5120. [DOI] [PubMed] [Google Scholar]
  • 148. Izui S, Kelley VE, Masuda K, Yoshida H, Roths JB, Murphy ED. Induction of various autoantibodies by mutant gene lpr in several strains of mice. J Immunol. 1984;133(1):227–233. [PubMed] [Google Scholar]
  • 149. Nose M, Terada M, Nishihara M, et al. . Genome analysis of collagen disease in MRL/lpr mice: polygenic inheritance resulting in the complex pathological manifestations. Int J Cardiol. 2000;75(Suppl 1):S61. [DOI] [PubMed] [Google Scholar]
  • 150. Takahashi T, Tanaka M, Brannan CI, et al. . Generalized lymphoproliferative disease in mice, caused by a point mutation in the fas ligand. Cell. 1994;76(6):969–976. [DOI] [PubMed] [Google Scholar]
  • 151. Roths JB, Murphy ED, Eicher EM. A new mutation, gld, that produces lymphoproliferation and autoimmunity in C3H/HeJ mice. J Exp Med. 1984;159(1):1–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152. Mosier DE, Stell KL, Gulizia RJ, Torbett BE, Gilmore GL. Homozygous scid/scid;beige/beige mice have low levels of spontaneous or neonatal T cell-induced B cell generation. J Exp Med. 1993;177(1):191–194. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153. Shibata S, Asano T, Ogura A, et al. . SCID-bg mice as xenograft recipients. Lab Anim. 1997;31(2):163–168. 10.1258/002367797780600107. [DOI] [PubMed] [Google Scholar]
  • 154. Mosbach-Ozmen L, Fonteneau P, Loor F. The C57BL/6 nu/nu lpr/lpr mouse. I. Expression of the ‘lpr phenotype’ in the C57BL/6 genetic background. Thymus. 1985;7(4):221–232. [PubMed] [Google Scholar]
  • 155. Mosbach-Ozmen L, Fonteneau P, Loor F. The C57BL/6 nu/nu lpr/lpr mouse. II. Pedigree and preliminary characteristics. Thymus. 1985;7(4):233–245. [PubMed] [Google Scholar]
  • 156. Trembleau S, Pflumio F, Kuntz L, Jachez B, Loor F. Immunoglobulin isotypes of C57BL/6 nu/nu, lpr/lpr mice. lack of direct intrinsic effect of the lpr gene on B cell hyperactivity. Autoimmunity. 1991;9(2):165–170. [DOI] [PubMed] [Google Scholar]
  • 157. Steinberg AD, Roths JB, Murphy ED, Steinberg RT, Raveche ES. Effects of thymectomy or androgen administration upon the autoimmune disease of MRL/mp-lpr/lpr mice. J Immunol. 1980;125(2):871–873. [PubMed] [Google Scholar]
  • 158. Pisetsky DS, Caster SA, Piper M, Scott DW, Steinberg AD. Cloning of B cells from autoimmune MRL-lpr/lpr and MRL.xid mice. Cell Immunol. 1984;84(1):32–40. [DOI] [PubMed] [Google Scholar]
  • 159. Steinberg EB, Santoro TJ, Chused TM, Smathers PA, Steinberg AD. Studies of congenic MRL-ipr/ipr.xid mice. J Immunol. 1983;131(6):2789–2795. [PubMed] [Google Scholar]
  • 160. Yasutomo K, Maeda K, Hisaeda H, Good RA, Kuroda Y, Himeno K. The fas-deficient SCID mouse exhibits the development of T cells in the thymus. J Immunol. 1997;158(10):4729–4733. [PubMed] [Google Scholar]
  • 161. Pisetsky DS, Klatt C, Dawson D, Roths JB. The influence of yaa on anti-DNA responses of B6-lpr mice. Clin Immunol Immunopathol. 1985;37(3):369–376. [DOI] [PubMed] [Google Scholar]
  • 162. Suzuka H, Yoshifusa H, Nakamura Y, Miyawaki S, Shibata Y. Morphological analysis of autoimmune disease in MRL-lpr,yaa male mice with rapidly progressive systemic lupus erythematosus. Autoimmunity. 1993;14(4):275–282. [DOI] [PubMed] [Google Scholar]
  • 163. Pflumio F, Fonteneau P, Gavériaux C, Cammisuli S, Loor F. The C57BL/6 nude, beige mouse: a model of combined T cell and NK effector cell immunodeficiency. Cell Immunol. 1989;120(1):218–229. [DOI] [PubMed] [Google Scholar]
  • 164. Sadoff DA, Giddens WE, DiGiacomo RF, Vogel AM. Neoplasms in NIH type II athymic (nude) mice. Lab Anim Sci. 1988;38(4):407–412. [PubMed] [Google Scholar]
  • 165. Azar HA, Hansen CT, Costa J. N:NIH(S)-nu/nu mice with combined immunodeficiency: a new model for human tumor heterotransplantation. J Natl Cancer Inst. 1980;65(2):421–430. [PubMed] [Google Scholar]
  • 166. Sainte-Marie G, Peng FS. Lymph nodes of the N:NIH(S)II-nu/nu mouse. Lab Invest. 1985;52(6):631–637. [PubMed] [Google Scholar]
  • 167. Waggie KS, Wu-Owens J, Hollifield V, Hansen CT. Lymphoblastic lymphoma in a colony of N:NIH(S)-bg-nu-xid mice. Lab Anim Sci. 1992;42(4):375–377. [PubMed] [Google Scholar]
  • 168. Ishigaki Y, Yasuda K, Hashimoto N, Nikaido H, Nikaido O, Hayakawa JI. Enhanced human tumor cell transplantability in a new congenic immunodeficient mouse; KSN-BNX. Folia Microbiol (Praha). 1998;43(5):493–494. [DOI] [PubMed] [Google Scholar]
  • 169. Lozzio BB, Lopez DM, Coulson P, Lair SV. High incidence of mammary tumors in mice with inherited asplenia carriers for the nude gene. Cancer Res. 1979;39(5):1529–1533. [PubMed] [Google Scholar]
  • 170. Bender AT, Pereira A, Fu K, et al. . Btk inhibition treats TLR7/IFN driven murine lupus. Clin Immunol. 2016;164:65–77. 10.1016/j.clim.2016.01.012. http://www.sciencedirect.com/science/article/pii/S1521661616300134. Accessed Jan 23, 2018. [DOI] [PubMed] [Google Scholar]
  • 171. Smee DF, Dagley A, Downs B, Hagloch J, Tarbet EB. Enhanced efficacy of cidofovir combined with vaccinia immune globulin in treating progressive cutaneous vaccinia virus infections in immunosuppressed hairless mice. Antimicrob Agents Chemother. 2015;59(1):520–526. https://www.ncbi.nlm.nih.gov/pubmed/25385098. Accessed Dec 1, 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172. Godfrey VL, Wilkinson JE, Rinchik EM, Russell LB. Fatal lymphoreticular disease in the scurfy (sf) mouse requires T cells that mature in a sf thymic environment: potential model for thymic education. Proc Natl Acad Sci USA. 1991;88:5528–5532. 10.1073/pnas.88.13.5528. http://www.pnas.org/content/88/13/5528. Accessed Jan 24, 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173. Sundberg JP. Handbook of mouse mutations with skin and hair abnormalities: animal models and biomedical tools. Boca Raton, FL: CRC Press; 1994. [Google Scholar]
  • 174. Nonaka S, Naito T, Chen H, et al. . Intestinal gamma delta T cells develop in mice lacking thymus, all lymph nodes, Peyer’s patches, and isolated lymphoid follicles. J Immunol. 2005;174(4):1906–1912. [DOI] [PubMed] [Google Scholar]
  • 175. Griesemer A, Yamada K, Sykes M. Xenotransplantation: immunological hurdles and progress toward tolerance. Immunol Rev. 2014;258(1):241–258. 10.1111/imr.12152. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176. Pearson T, Greiner DL, Shultz LD. Creation of “humanized” mice to study human immunity. Curr Protoc Immunol. 2008;Chapter 15:Unit 15.21. 10.1002/0471142735.im1521s81. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177. Iwakawa M, Noda S, Ohta T, et al. . Different radiation susceptibility among five strains of mice detected by a skin reaction. J Radiat Res. 2003;44(1):7–13. [DOI] [PubMed] [Google Scholar]
  • 178. Mori N, Matsumoto Y, Okumoto M, Suzuki N, Yamate J. Variations in prkdc encoding the catalytic subunit of DNA-dependent protein kinase (DNA-PKcs) and susceptibility to radiation-induced apoptosis and lymphomagenesis. Oncogene. 2001;20(28):3609–3619. 10.1038/sj.onc.1204497. [DOI] [PubMed] [Google Scholar]
  • 179. Mukherjee S, Sainis KB, Deobagkar DD. F1 hybrids of BALB/c and C57BL/6 mouse strains respond differently to low-dose ionizing radiation exposure. J Genet. 2014;93(3):667–682. [DOI] [PubMed] [Google Scholar]
  • 180. Degg NL, Weil MM, Edwards A, et al. . Adenoma multiplicity in irradiated apc(min) mice is modified by chromosome 16 segments from BALB/c. Cancer Res. 2003;63(10):2361–2363. [PubMed] [Google Scholar]
  • 181. Yu Y, Okayasu R, Weil MM, et al. . Elevated breast cancer risk in irradiated BALB/c mice associates with unique functional polymorphism of the prkdc (DNA-dependent protein kinase catalytic subunit) gene. Cancer Res. 2001;61(5):1820–1824. [PubMed] [Google Scholar]
  • 182. Lombardini ED, Pacheco-Thompson ME, Melanson MA. Chapter 44 radiation and other physical agents—dimensions In: Haschek WM, Rousseaux CG, Wallig MA, Bolon B, Ochoa R, eds. Haschek and Rousseaux’s handbook of toxicologic pathology third edition. London: Academic Press; 2013:1421–1503. https://app.dimensions.ai/details/publication/pub.1010167368. Accessed May 14, 2018. [Google Scholar]
  • 183. Hernández-Godoy J, Silvestre DP, Hernández BB. Immediate and short-, mid- and long-term effects of in vivo ionizing radiation exposure in BALB/c mice: i. activation of lymphocytes and subpopulations. In Vivo. 2010;24(5):719–726. [PubMed] [Google Scholar]
  • 184. Manda K, Glasow A, Paape D, Hildebrandt G. Effects of ionizing radiation on the immune system with special emphasis on the interaction of dendritic and T cells. Front Oncol. 2012;2:102 10.3389/fonc.2012.00102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 185. Wensinck F, Van Bekkum DW, Renaud H. The prevention of Pseudomonas aeruginosa infections in irradiated mice and rats. Radiat Res. 1957;7(5):491–499. [PubMed] [Google Scholar]
  • 186. Chiang CS, McBride WH, Withers HR. Myelin-associated changes in mouse brain following irradiation. Radiother Oncol. 1993;27(3):229–236. [DOI] [PubMed] [Google Scholar]
  • 187. Larsen SR, Kingham JA, Hayward MD, Rasko JEJ. Damage to incisors after nonmyeloablative total body irradiation may complicate NOD/SCID models of hemopoietic stem cell transplantation. Comp Med. 2006;56(3):209–214. [PubMed] [Google Scholar]
  • 188. Schwarte S, Bremer M, Fruehauf J, Sorge Y, Skubich S, Hoffmann MW. Radiation protocols determine acute graft-versus-host disease incidence after allogeneic bone marrow transplantation in murine models. Int J Radiat Biol. 2007;83(9):625–636. 10.1080/09553000701534572. https://www.ncbi.nlm.nih.gov/pubmed/17654104. Accessed Dec 1, 2018. [DOI] [PubMed] [Google Scholar]
  • 189. Borges HL, Chao C, Xu Y, Linden R, Wang JYJ. Radiation-induced apoptosis in developing mouse retina exhibits dose-dependent requirement for ATM phosphorylation ofp53. Cell Death Differ. 2004;11(5):494–502. 10.1038/sj.cdd.4401366. [DOI] [PubMed] [Google Scholar]
  • 190. Down JD, Tarbell NJ, Warhol M, Mauch P. Dose-limiting complications from upper half body irradiation in C3H mice. Int J Radiat Oncol Biol Phys. 1988;14(3):483–489. [DOI] [PubMed] [Google Scholar]
  • 191. Gartner LP, Hiatt JL, Provenza DV. Effects of ionizing radiation on incisor development of the prenatal mouse. Acta Anat (Basel). 1977;98(4):367–375. [DOI] [PubMed] [Google Scholar]
  • 192. Jaggi JS, Seshan SV, McDevitt MR, LaPerle K, Sgouros G, Scheinberg DA. Renal tubulointerstitial changes after internal irradiation with alpha-particle-emitting actinium daughters. J Am Soc Nephrol. 2005;16(9):2677–2689. 10.1681/ASN.2004110945. [DOI] [PubMed] [Google Scholar]
  • 193. Medak H, Weinreb M, Sicher H, Weinmann JP, Schour I. The effect of single doses of irradiation upon the tissues of the upper rat incisor. J Dent Res. 1952;31(4):559–574. 10.1177/00220345520310040601. [DOI] [PubMed] [Google Scholar]
  • 194. Pearson AE, Phelps TA. Radiation effects on mouse incisor teeth following whole-body doses of up to 16 gray. Int J Radiat Biol Relat Stud Phys Chem Med. 1981;39(4):409–417. [DOI] [PubMed] [Google Scholar]
  • 195. Schmidt SL, Vitral RW, Linden R. Effects of prenatal ionizing irradiation on the development of the ganglion cell layer of the mouse retina. Int J Dev Neurosci. 2001;19(4):469–473. [DOI] [PubMed] [Google Scholar]
  • 196. Byrne SN, Spinks N, Halliday GM. Ultraviolet a irradiation of C57BL/6 mice suppresses systemic contact hypersensitivity or enhances secondary immunity depending on dose. J Invest Dermatol. 2002;119(4):858–864. 10.1046/j.1523-1747.2002.00261.x. [DOI] [PubMed] [Google Scholar]
  • 197. Schwarz T. The dark and the sunny sides of UVR-induced immunosuppression: photoimmunology revisited. J Invest Dermatol. 2010;130(1):49–54. 10.1038/jid.2009.217. [DOI] [PubMed] [Google Scholar]
  • 198. González Maglio DH, Paz ML, Leoni J. Sunlight effects on immune system: is there something else in addition to UV-induced immunosuppression? Biomed Res Int. 2016;2016:1–10. 10.1155/2016/1934518. https://www.ncbi.nlm.nih.gov/pubmed/28070504. Accessed Dec 1, 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 199. Zuluaga AF, Salazar BE, Rodriguez CA, Zapata AX, Agudelo M, Vesga O. Neutropenia induced in outbred mice by a simplified low-dose cyclophosphamide regimen: characterization and applicability to diverse experimental models of infectious diseases. BMC Infect Dis. 2006;6:55 10.1186/1471-2334-6-55. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 200. Brownstein DG. Pathogenesis of bacteremia due to pseudomonas aeruginosa in cyclophosphamide-treated mice and potentiation of virulence of endogenous streptococci. J Infect Dis. 1978;137(6):795–801. [DOI] [PubMed] [Google Scholar]
  • 201. De Bernardis F, Palliola E, Lorenzini R, Antonucci G. Evaluation of the experimental pathogenicity of some cryptococcus species in normal and cyclophosphamide-immunodepressed mice. Microbiol Immunol. 1987;31(5):449–460. [DOI] [PubMed] [Google Scholar]
  • 202. Sistigu A, Viaud S, Chaput N, Bracci L, Proietti E, Zitvogel L. Immunomodulatory effects of cyclophosphamide and implementations for vaccine design. Semin Immunopathol. 2011;33(4):369–383. 10.1007/s00281-011-0245-0. [DOI] [PubMed] [Google Scholar]
  • 203. Peng KW, Myers R, Greenslade A, et al. . Using clinically approved cyclophosphamide regimens to control the humoral immune response to oncolytic viruses. Gene Ther. 2013;20(3):255–261. 10.1038/gt.2012.31. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 204. Heylmann D, Bauer M, Becker H, et al. . Human CD4+CD25+ regulatory T cells are sensitive to low dose cyclophosphamide: implications for the immune response. PLoS One. 2013;8(12):e83384 10.1371/journal.pone.0083384. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 205. Wilkinson FL, Sergijenko A, Langford-Smith KJ, Malinowska M, Wynn RF, Bigger BW. Busulfan conditioning enhances engraftment of hematopoietic donor-derived cells in the brain compared with irradiation. Mol Ther. 2013;21(4):868–876. 10.1038/mt.2013.29. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 206. Robert-Richard E, Ged C, Ortet J, et al. . Human cell engraftment after busulfan or irradiation conditioning of NOD/SCID mice. Haematologica. 2006;91(10):1384. [PubMed] [Google Scholar]
  • 207. Ghiringhelli F, Bruchard M, Apetoh L. Immune effects of 5-fluorouracil: ambivalence matters. Oncoimmunology. 2013;2(3):e23139 10.4161/onci.23139. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 208. Vincent J, Mignot G, Chalmin F, et al. . 5-Fluorouracil selectively kills tumor-associated myeloid-derived suppressor cells resulting in enhanced T cell-dependent antitumor immunity. Cancer Res. 2010;70(8):3052–3061. 10.1158/0008-5472.CAN-09-3690. [DOI] [PubMed] [Google Scholar]
  • 209. Halloran PF, Helms LM, Kung L, Noujaim J. The temporal profile of calcineurin inhibition by cyclosporine in vivo. Transplantation. 1999;68(9):1356–1361. [DOI] [PubMed] [Google Scholar]
  • 210. Matsue H, Yang C, Matsue K, Edelbaum D, Mummert M, Takashima A. Contrasting impacts of immunosuppressive agents (rapamycin, FK506, cyclosporin A, and dexamethasone) on bidirectional dendritic cell-T cell interaction during antigen presentation. J Immunol. 2002;169(7):3555–3564. [DOI] [PubMed] [Google Scholar]
  • 211. Barequet IS, Platner E, Sade K, et al. . Topical tacrolimus for the management of acute allergic conjunctivitis in a mouse model. Graefes Arch Clin Exp Ophthalmol. 2013;251(7):1717–1721. 10.1007/s00417-013-2333-4. [DOI] [PubMed] [Google Scholar]
  • 212. Niwa Y, Terashima T, Sumi H. Topical application of the immunosuppressant tacrolimus accelerates carcinogenesis in mouse skin. Br J Dermatol. 2003;149(5):960–967. [DOI] [PubMed] [Google Scholar]
  • 213. Obayashi K, Tomonari M, Yoshimatsu H, et al. . Dosing time-dependency of the arthritis-inhibiting effect of tacrolimus in mice. J Pharmacol Sci. 2011;116(3):264–273. [DOI] [PubMed] [Google Scholar]
  • 214. Shin J, Park HR, Kim SW, et al. . The effect of topical FK506 (tacrolimus) in a mouse model of allergic rhinitis. Am J Rhinol Allergy. 2012;26(2):71 10.2500/ajra.2012.26.3743. [DOI] [PubMed] [Google Scholar]
  • 215. Vandenbussche C, Van der Hauwaert C, Dewaeles E, et al. . Tacrolimus-induced nephrotoxicity in mice is associated with microRNA deregulation. Arch Toxicol. 2018;92(4):1539–1550. 10.1007/s00204-018-2158-3. [DOI] [PubMed] [Google Scholar]
  • 216. Furukawa A, Wisel SA, Tang Q. Impact of immune-modulatory drugs on regulatory T cell. Transplantation. 2016;100(11):2288–2300. 10.1097/TP.0000000000001379. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 217. Bonkhoff H. Estrogen receptor signaling in prostate cancer: implications for carcinogenesis and tumor progression. Prostate. 2018;78(1):2–10. 10.1002/pros.23446. [DOI] [PubMed] [Google Scholar]
  • 218. Nelles JL, Hu W, Prins GS. Estrogen action and prostate cancer. Expert Rev Endocrinol Metab. 2011;6(3):437–451. 10.1586/eem.11.20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 219. Song W, Soni V, Soni S, Khera M. Testosterone inhibits the growth of prostate cancer xenografts in nude mice. BMC Cancer. 2017;17(1):635 10.1186/s12885-017-3569-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 220. Turo R, Smolski M, Esler R, et al. . Diethylstilboestrol for the treatment of prostate cancer: past, present and future. Scand J Urol. 2014;48(1):4–14. 10.3109/21681805.2013.861508. [DOI] [PubMed] [Google Scholar]
  • 221. Ngobili TA, Daniele MA. Nanoparticles and direct immunosuppression. Exp Biol Med. 2016;241(10):1064–1073. 10.1177/1535370216650053. http://journals.sagepub.com/doi/full/10.1177/1535370216650053. Accessed Dec 1, 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 222. Luo Y, Chang LW, Lin P. Metal-based nanoparticles and the immune system: activation, inflammation, and potential applications. Biomed Res Int. 2015;2015:143720 http://www.ncbi.nlm.nih.gov/pubmed/26125021. Accessed Dec 1, 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 223. Zolnik BS, González-Fernández A, Sadrieh N, Dobrovolskaia MA. Nanoparticles and the immune system. Endocrinology. 2010;151(2):458–465. 10.1210/en.2009-1082. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 224. Daley JM, Thomay AA, Connolly MD, Reichner JS, Albina JE. Use of Ly6G-specific monoclonal antibody to deplete neutrophils in mice. J Leukoc Biol. 2008;83(1):64–70. 10.1189/jlb.0407247. [DOI] [PubMed] [Google Scholar]
  • 225. Kruisbeek AM. In vivo depletion of CD4- and CD8-specific T cells. Curr Protoc Immunol. 2001;Chapter 4: Unit4.1. 10.1002/0471142735.im0401s01. [DOI] [PubMed] [Google Scholar]
  • 226. Montalvao F, Garcia Z, Celli S, et al. . The mechanism of anti-CD20-mediated B cell depletion revealed by intravital imaging. J Clin Invest. 2013;123(12):5098–5103. 10.1172/JCI70972. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 227. Vergani A, D’Addio F, Jurewicz M, et al. . A novel clinically relevant strategy to abrogate autoimmunity and regulate alloimmunity in NOD mice. Diabetes. 2010;59(9):2253–2264. 10.2337/db09-1264. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 228. Xia C, Chernatynskaya AV, Wasserfall CH, et al. . Anti-thymocyte globulin (ATG) differentially depletes naïve and memory T cells and permits memory-type regulatory T cells in nonobese diabetic mice. BMC Immunol. 2012;13:70 10.1186/1471-2172-13-70. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 229. Jahrsdörfer B, Weiner GJ. CpG oligodeoxynucleotides as immunotherapy in cancer. Update Cancer Ther. 2008;3(1):27–32. 10.1016/j.uct.2007.11.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 230. Liu W, Yang X, Wang N, et al. . Multiple immunosuppressive effects of CpG-c41 on intracellular TLR-mediated inflammation. Mediators Inflamm. 2017;2017:6541729 10.1155/2017/6541729. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 231. Asano M, Toda M, Sakaguchi N, Sakaguchi S. Autoimmune disease as a consequence of developmental abnormality of a T cell subpopulation. J Exp Med. 1996;184(2):387–396. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 232. Dooley J, Erickson M, Gillard GO, Farr AG. Cervical thymus in the mouse. J Immunol. 2006;176(11):6484–6490. [DOI] [PubMed] [Google Scholar]
  • 233. Many MC, Drexhage HA, Denef JF. High frequency of thymic ectopy in thyroids from autoimmune prone nonobese diabetic female mice. Lab Invest. 1993;69(3):364–367. [PubMed] [Google Scholar]
  • 234. Bronte V, Pittet MJ. The spleen in local and systemic regulation of immunity. Immunity. 2013;39(5):806–818. 10.1016/j.immuni.2013.10.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 235. Lin H, Faunce DE, Stacey M, et al. . The macrophage F4/80 receptor is required for the induction of antigen-specific efferent regulatory T cells in peripheral tolerance. J Exp Med. 2005;201(10):1615–1625. 10.1084/jem.20042307. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 236. Higashijima J, Shimada M, Chikakiyo M, et al. . Effect of splenectomy on antitumor immune system in mice. Anticancer Res. 2009;29(1):385–393. [PubMed] [Google Scholar]
  • 237. Datta K, Hyduke DR, Suman S, Moon B, Johnson MD, Fornace AJ. Exposure to ionizing radiation induced persistent gene expression changes in mouse mammary gland. Radiat Oncol. 2012;7:205 10.1186/1748-717x-7-205. https://www.ncbi.nlm.nih.gov/pubmed/23216862. Accessed May 28, 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 238. Nakatsukasa H, Tsukimoto M, Tokunaga A, Kojima S. Repeated gamma irradiation attenuates collagen-induced arthritis via up-regulation of regulatory T cells but not by damaging lymphocytes directly. Radiat Res. 2010;174(3):313–324. 10.1667/RR2121.1. [DOI] [PubMed] [Google Scholar]
  • 239. Pecaut MJ, Nelson GA, Gridley DS. Dose and dose rate effects of whole-body gamma-irradiation: i. lymphocytes and lymphoid organs. In Vivo. 2001;15(3):195–208. [PubMed] [Google Scholar]
  • 240. Rivina L, Davoren MJ, Schiestl RH. Mouse models for radiation-induced cancers. Mutagenesis. 2016;31(5):491–509. 10.1093/mutage/gew019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 241. Tsukimoto M, Nakatsukasa H, Sugawara K, Yamashita K, Kojima S. Repeated 0.5-gy gamma irradiation attenuates experimental autoimmune encephalomyelitis with up-regulation of regulatory T cells and suppression of IL17 production. Radiat Res. 2008;170(4):429–436. [DOI] [PubMed] [Google Scholar]
  • 242. Willey JS, Lloyd SAJ, Nelson GA, Bateman TA. Ionizing radiation and bone loss: space exploration and clinical therapy applications. Clin Rev Bone Miner Metab. 2011;9(1):54–62. 10.1007/s12018-011-9092-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 243. Worgul BV, Smilenov L, Brenner DJ, Junk A, Zhou W, Hall EJ. Atm heterozygous mice are more sensitive to radiation-induced cataracts than are their wild-type counterparts. Proc Natl Acad Sci USA. 2002;99(15):9836–9839. 10.1073/pnas.162349699. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 244. Bellés M, Gonzalo S, Serra N, et al. . Environmental exposure to low-doses of ionizing radiation. Effects on early nephrotoxicity in mice. Environ Res. 2017;156:291–296. 10.1016/j.envres.2017.03.034. https://www.ncbi.nlm.nih.gov/pubmed/28371757. Accessed May 28, 2018. [DOI] [PubMed] [Google Scholar]
  • 245. Bryan RA, Jiang Z, Howell RC, et al. . Radioimmunotherapy is more effective than antifungal treatment in experimental cryptococcal infection. J Infect Dis. 2010;202(4):633 10.1086/654813. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC2904428/. Accessed Jun 1, 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 246. Gorin J, Guilloux Y, Morgenstern A, Chérel M, Davodeau F, Gaschet J. Using α radiation to boost cancer immunity? Oncoimmunology. 2014;3(9):e954925 10.4161/21624011.2014.954925. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4292714/. Accessed Jun 1, 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 247. Jaggi JS, Seshan SV, McDevitt MR, Sgouros G, Hyjek E, Scheinberg DA. Mitigation of radiation nephropathy after internal α-particle irradiation of kidneys. Int J Radiat Oncol Biol Phys. 2006;64(5):1503–1512. 10.1016/j.ijrobp.2005.11.036. https://www.redjournal.org/article/S0360-3016(05)02985-8/fulltext. Accessed Jun 1, 2018. [DOI] [PubMed] [Google Scholar]
  • 248. Denkins Y, Fidler IJ, Kripke ML. Exposure of mice to UV-B radiation suppresses delayed hypersensitivity to candida albicans. Photochem Photobiol. 1989;49(5):615–619. [DOI] [PubMed] [Google Scholar]
  • 249. Jeevan A, Kripke ML. Effect of a single exposure to ultraviolet radiation on mycobacterium bovis bacillus calmette-guerin infection in mice. J Immunol. 1989;143(9):2837–2843. [PubMed] [Google Scholar]
  • 250. Kaplan DH, Jenison MC, Saeland S, Shlomchik WD, Shlomchik MJ. Epidermal langerhans cell-deficient mice develop enhanced contact hypersensitivity. Immunity. 2005;23(6):611–620. 10.1016/j.immuni.2005.10.008. [DOI] [PubMed] [Google Scholar]
  • 251. Miyauchi-Hashimoto H, Tanaka K, Horio T. Enhanced inflammation and immunosuppression by ultraviolet radiation in xeroderma pigmentosum group A (XPA) model mice. J Invest Dermatol. 1996;107(3):343–348. [DOI] [PubMed] [Google Scholar]
  • 252. Miyauchi-Hashimoto H, Sugihara A, Tanaka K, Horio T. Ultraviolet radiation-induced impairment of tumor rejection is enhanced in xeroderma pigmentosum a gene-deficient mice. J Invest Dermatol. 2005;124(6):1313–1317. 10.1111/j.0022-202x.2005.23717.x. [DOI] [PubMed] [Google Scholar]
  • 253. Obhrai JS, Oberbarnscheidt M, Zhang N, et al. . Langerhans cells are not required for efficient skin graft rejection. J Invest Dermatol. 2008;128(8):1950–1955. 10.1038/jid.2008.52. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 254. Pronk C, Oldenziel H, Lequin HC. A method for determination of serum iron, total iron binding capacity and iron in urine by atomic absorption spectrophotometry with manganese as internal standard. Clin Chim Acta. 1974;50(1):35–41. [DOI] [PubMed] [Google Scholar]
  • 255. Rana S, Rogers LJ, Halliday GM. Immunosuppressive ultraviolet-A radiation inhibits the development of skin memory CD8 T cells. Photochem Photobiol Sci. 2010;9(1):25–30. 10.1039/b9pp00051h. [DOI] [PubMed] [Google Scholar]
  • 256. Reeve VE, Tyrrell RM. Heme oxygenase induction mediates the photoimmunoprotective activity of UVA radiation in the mouse. Proc Natl Acad Sci USA. 1999;96(16):9317–9321. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 257. Shen J, Bao S, Reeve VE. Modulation of IL-10, IL-12, and IFN-gamma in the epidermis of hairless mice by UVA (320-400 nm) and UVB (280–320 nm) radiation. J Invest Dermatol. 1999;113(6):1059–1064. 10.1046/j.1523-1747.1999.00782.x. [DOI] [PubMed] [Google Scholar]
  • 258. Yuen KS, Nearn MR, Halliday GM. Nitric oxide-mediated depletion of langerhans cells from the epidermis may be involved in UVA radiation-induced immunosuppression. Nitric Oxide. 2002;6(3):313–318. 10.1006/niox.2001.0414. [DOI] [PubMed] [Google Scholar]
  • 259. Altman A, Hochberg Z, Silbermann M. Interactions between growth hormone and dexamethasone in skeletal growth and bone structure of the young mouse. Calcif Tissue Int. 1992;51(4):298–304. [DOI] [PubMed] [Google Scholar]
  • 260. Cain DW, Cidlowski JA. Immune regulation by glucocorticoids. Nat Rev Immunol. 2017;17(4):233–247. 10.1038/nri.2017.1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 261. Ersek A, Santo AIE, Vattakuzhi Y, George S, Clark AR, Horwood NJ. Strain dependent differences in glucocorticoid-induced bone loss between C57BL/6J and CD-1 mice. Sci Rep. 2016;6:36513 10.1038/srep36513. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 262. McLaughlin F, Mackintosh J, Hayes BP, et al. . Glucocorticoid-induced osteopenia in the mouse as assessed by histomorphometry, microcomputed tomography, and biochemical markers. Bone. 2002;30(6):924–930. [DOI] [PubMed] [Google Scholar]
  • 263. Purton JF, Monk JA, Liddicoat DR, et al. . Expression of the glucocorticoid receptor from the 1A promoter correlates with T lymphocyte sensitivity to glucocorticoid-induced cell death. J Immunol. 2004;173(6):3816–3824. [DOI] [PubMed] [Google Scholar]
  • 264. Rousseaux CG, Bolon B. Chapter 62—embryo and fetus In: Haschek W, Rousseaux C, Wallig M, eds. Haschek and Rousseaux’s handbook of toxicologic pathology. 3rd ed.Boston: Academic Press; 2013:2695–2759. https://www.sciencedirect.com/science/article/pii/B9780124157590000625. Accessed Jun 1, 2018. [Google Scholar]
  • 265. Silverstone AE, Frazier DE, Fiore NC, Soults JA, Gasiewicz TA. Dexamethasone, beta-estradiol, and 2,3,7,8-tetrachlorodibenzo-p-dioxin elicit thymic atrophy through different cellular targets. Toxicol Appl Pharmacol. 1994;126(2):248–259. 10.1006/taap.1994.1114. [DOI] [PubMed] [Google Scholar]
  • 266. Anton E. Delayed toxicity of cyclophosphamide on the bladder of DBA/2 and C57BL/6 female mouse. Int J Exp Pathol. 2002;83(1):47–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 267. Bracci L, Moschella F, Sestili P, et al. . Cyclophosphamide enhances the antitumor efficacy of adoptively transferred immune cells through the induction of cytokine expression, B-cell and T-cell homeostatic proliferation, and specific tumor infiltration. Clin Cancer Res. 2007;13(2 Pt 1):644–653. 10.1158/1078-0432.CCR-06-1209. [DOI] [PubMed] [Google Scholar]
  • 268. Golubeva AV, Zhdanov AV, Mallel G, Dinan TG, Cryan JF. The mouse cyclophosphamide model of bladder pain syndrome: tissue characterization, immune profiling, and relationship to metabotropic glutamate receptors. Physiol Rep. 2014;2(3):e00260 10.1002/phy2.260. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 269. Kawakami T, Nakamura Y, Karibe H. Cyclophosphamide inhibits root development of molar teeth in growing mice. Odontology. 2015;103(2):143–151. 10.1007/s10266-014-0158-1. [DOI] [PubMed] [Google Scholar]
  • 270. Nakahara T, Uchi H, Lesokhin AM, et al. . Cyclophosphamide enhances immunity by modulating the balance of dendritic cell subsets in lymphoid organs. Blood. 2010;115(22):4384–4392. 10.1182/blood-2009-11-251231. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 271. Xu X, Zhang X. Effects of cyclophosphamide on immune system and gut microbiota in mice. Microbiol Res. 2015;171:97–106. 10.1016/j.micres.2014.11.002. [DOI] [PubMed] [Google Scholar]
  • 272. Yasunami R, Bach JF. Anti-suppressor effect of cyclophosphamide on the development of spontaneous diabetes in NOD mice. Eur J Immunol. 1988;18(3):481–484. 10.1002/eji.1830180325. [DOI] [PubMed] [Google Scholar]
  • 273. Zhao D, Wang C, Zhao Y, et al. . Cyclophosphamide causes osteoporosis in C57BL/6 male mice: suppressive effects of cyclophosphamide on osteoblastogenesis and osteoclastogenesis. Oncotarget. 2017;8(58):98163–98183. 10.18632/oncotarget.21000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 274. Swindell WR. Meta-analysis of 29 experiments evaluating the effects of rapamycin on life span in the laboratory mouse. J Gerontol A Biol Sci Med Sci. 2017;72(8):1024–1032. 10.1093/gerona/glw153. [DOI] [PubMed] [Google Scholar]
  • 275. Thomson AW, Turnquist HR, Raimondi G. Immunoregulatory functions of mTOR inhibition. Nat Rev Immunol. 2009;9(5):324–337. 10.1038/nri2546. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 276. Bhoopalam N, Price KS, Norgello H, Fried W. Busulfan-induced suppression of natural killer cell activity. Exp Hematol. 1985;13(11):1127–1132. [PubMed] [Google Scholar]
  • 277. Gibson FM, Andrews CM, Diamanti P, et al. . A new model of busulphan-induced chronic bone marrow aplasia in the female BALB/c mouse. Int J Exp Pathol. 2003;84(1):31–48. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 278. Hassan M. The role of busulfan in bone marrow transplantation. Med Oncol. 1999;16(3):166–176. [DOI] [PubMed] [Google Scholar]
  • 279. Molyneux G, Andrews M, Sones W, et al. . Haemotoxicity of busulphan, doxorubicin, cisplatin and cyclophosphamide in the female BALB/c mouse using a brief regimen of drug administration. Cell Biol Toxicol. 2011;27(1):13–40. 10.1007/s10565-010-9167-1. [DOI] [PubMed] [Google Scholar]
  • 280. Sjöö F, Hassan Z, Abedi-Valugerdi M, et al. . Myeloablative and immunosuppressive properties of treosulfan in mice. Exp Hematol. 2006;34(1):115–121. 10.1016/j.exphem.2005.09.015. [DOI] [PubMed] [Google Scholar]
  • 281. Buettner M, Bornemann M, Bode U. Skin tolerance is supported by the spleen. Scand J Immunol. 2013;77(4):238–245. 10.1111/sji.12034. [DOI] [PubMed] [Google Scholar]
  • 282. Fotiadis C, Zografos G, Aronis K, et al. . The effect of various types of splenectomy on the development of B-16 melanoma in mice. Anticancer Res. 1999;19(5B):4235–4239. [PubMed] [Google Scholar]
  • 283. Chung DT, Korn T, Richard J, et al. . Anti-thymocyte globulin (ATG) prevents autoimmune encephalomyelitis by expanding myelin antigen-specific Foxp3+ regulatory T cells. Int Immunol. 2007;19(8):1003–1010. 10.1093/intimm/dxm078. [DOI] [PubMed] [Google Scholar]
  • 284. Bakheet SA, Attia SM, Alwetaid MY, et al. . Β-1,3-glucan reverses aflatoxin B1-mediated suppression of immune responses in mice. Life Sci. 2016;152:1–13. 10.1016/j.lfs.2016.03.030. [DOI] [PubMed] [Google Scholar]
  • 285. Norton EB, Clements JD, Voss TG, Cárdenas-Freytag L. Prophylactic administration of bacterially derived immunomodulators improves the outcome of influenza virus infection in a murine model. J Virol. 2010;84(6):2983–2995. 10.1128/JVI.01805-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 286. Maki T, Ichikawa T, Blanco R, Porter J. Long-term abrogation of autoimmune diabetes in nonobese diabetic mice by immunotherapy with anti-lymphocyte serum. Proc Natl Acad Sci USA. 1992;89(8):3434–3438. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 287. Bouaziz J, Yanaba K, Venturi GM, et al. . Therapeutic B cell depletion impairs adaptive and autoreactive CD4+ T cell activation in mice. Proc Natl Acad Sci USA. 2007;104(52):20878–20883. 10.1073/pnas.0709205105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 288. Chen L, Mehta ND, Zhao Y, DiPietro LA. Absence of CD4 or CD8 lymphocytes changes infiltration of inflammatory cells and profiles of cytokine expression in skin wounds, but does not impair healing. Exp Dermatol. 2014;23(3):189–194. 10.1111/exd.12346. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 289. Jing W, Gershan JA, Johnson BD. Depletion of CD4 T cells enhances immunotherapy for neuroblastoma after syngeneic HSCT but compromises development of antitumor immune memory. Blood. 2009;113(18):4449–4457. 10.1182/blood-2008-11-190827. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 290. Rosenblum MD, Remedios KA, Abbas AK. Mechanisms of human autoimmunity. J Clin Invest. 2015;125:2228–2233. http://www.ncbi.nlm.nih.gov/pubmed/25893595. Accessed Dec 1, 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 291. Melli K, Friedman RS, Martin AE, et al. . Amplification of autoimmune response through induction of dendritic cell maturation in inflamed tissues. J Immunol. 2009;182:2590–2600. http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3057894/. Accessed Dec 1, 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 292. Schmidt RE, Grimbacher B, Witte T. Autoimmunity and primary immunodeficiency: two sides of the same coin? Nat Rev Rheumatol. 2017;14(1):7–18. [DOI] [PubMed] [Google Scholar]
  • 293. Petkova SB, Yuan R, Tsaih SW, Schott W, Roopenian DC, Paigen B. Genetic influence on immune phenotype revealed strain-specific variations in peripheral blood lineages. Physiol Genomics. 2008;34(3):304–314. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 294. deLuca LES, Pikor NB, O’Leary J, et al. . Substrain differences reveal novel disease-modifying gene candidates that alter the clinical course of a rodent model of multiple sclerosis. J Immunol. 2010;184:3174–3185. http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3160975/. Accessed Dec 1, 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 295. Begley CG, Ellis LM. Drug development: raise standards for preclinical cancer research. Nature. 2012;483(7391):531–533. [DOI] [PubMed] [Google Scholar]
  • 296. Rice AS, Cimino-Brown D, Eisenach JC, et al. . Animal models and the prediction of efficacy in clinical trials of analgesic drugs: a critical appraisal and call for uniform reporting standards. Pain. 2008;139(2):243–247. [DOI] [PubMed] [Google Scholar]
  • 297. Bevaart L, Vervoordeldonk MJ, Tak PP. Evaluation of therapeutic targets in animal models of arthritis: how does it relate to rheumatoid arthritis? Arthritis Rheum. 2010;62:2192–2205. http://www.ncbi.nlm.nih.gov/pubmed/20506322. [DOI] [PubMed] [Google Scholar]
  • 298. Farkas B, Boldizsar F, Tarjanyi O, et al. . BALB/c mice genetically susceptible to proteoglycan-induced arthritis and spondylitis show colony-dependent differences in disease penetrance. Arthritis Res Ther. 2009;11:R21 http://www.ncbi.nlm.nih.gov/pubmed/19220900. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 299. Glant TT, Radacs M, Nagyeri G, et al. . Proteoglycan (PG)-induced arthritis (PGIA) and recombinant human PG-G1 domain-induced arthritis (GIA) in BALB/c mice resembling two subtypes of rheumatoid arthritis. Arthritis Rheum. 2011;63:1312–1321. http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3086933/. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 300. Boldizsar F, Tarjanyi O, Nemeth P, Mikecz K, Glant TT. T(h)1/T(h)17 polarization and acquisition of an arthritogenic phenotype in arthritis-susceptible BALB/c, but not in MHC-matched, arthritis-resistant DBA/2 mice. Int Immunol. 2009;21:511–522. http://www.ncbi.nlm.nih.gov/pmc/articles/PMC2675029/. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 301. Voskuhl RR, Gold SM. Sex-related factors in multiple sclerosis: genetic, hormonal and environmental contributions. Nat Rev Neurol. 2012;8:255–263. http://www.ncbi.nlm.nih.gov/pmc/articles/PMC4435786/. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 302. Bernard CC, Mackay IR. Experimental autoimmune encephalomyelitis in mice: lyt and ia phenotypes of the effector and suppressor cells. Prog Clin Biol Res. 1984;146:277–284. http://www.ncbi.nlm.nih.gov/pubmed/6232623. [PubMed] [Google Scholar]
  • 303. Bernard CC. Suppressor T cells prevent experimental autoimmune encephalomyelitis in mice. Clin Exp Immunol. 1977;29:100–109. http://www.ncbi.nlm.nih.gov/pubmed/302168. [PMC free article] [PubMed] [Google Scholar]
  • 304. Bernard CC, Leydon J, Mackay IR. T cell necessity in the pathogenesis of experimental autoimmune encephalomyelitis in mice. Eur J Immunol. 1976;6:655–660. http://www.ncbi.nlm.nih.gov/pubmed/1087243. [DOI] [PubMed] [Google Scholar]
  • 305. Croxford AL, Kurschus FC, Waisman A. Mouse models for multiple sclerosis: historical facts and future implications. Biochim Biophys Acta. 2011;1812(2):177–183. 10.1016/j.bbadis.2010.06.010. [DOI] [PubMed] [Google Scholar]
  • 306. Bernard CC, Carnegie PR. Experimental autoimmune encephalomyelitis in mice: immunologic response to mouse spinal cord and myelin basic proteins. J Immunol. 1975;114:1537–1540. http://www.ncbi.nlm.nih.gov/pubmed/47366. [PubMed] [Google Scholar]
  • 307. Behlke MA, Loh DY. Alternative splicing of murine T-cell receptor β-chain transcripts. Nature. 1986;322(6077):379 10.1038/322379a0. https://proxy.library.upenn.edu:2611/articles/322379a0. Accessed May 3, 2018. [DOI] [PubMed] [Google Scholar]
  • 308. Bernard CC. Experimental autoimmune encephalomyelitis in mice: genetic control of susceptibility. J Immunogenet. 1976;3:263–274. http://www.ncbi.nlm.nih.gov/pubmed/1109134. [DOI] [PubMed] [Google Scholar]
  • 309. Kawamura K, Yao K, Shukaliak-Quandt JA, et al. . Different development of myelin basic protein agonist- and antagonist-specific human TCR transgenic T cells in the thymus and periphery. J Immunol. 2008;181(8):5462–5472. [DOI] [PubMed] [Google Scholar]
  • 310. Sato F, Tanaka H, Hasanovic F, Tsunoda I. Theiler’s virus infection: pathophysiology of demyelination and neurodegeneration. Pathophysiology. 2011;18(1):31–41. 10.1016/j.pathophys.2010.04.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 311. Turrin NP. Central nervous system toll-like receptor expression in response to Theiler’s murine encephalomyelitis virus-induced demyelination disease in resistant and susceptible mouse strains. Virol J. 2008;5:154 10.1186/1743-422x-5-154. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 312. McCarthy DP, Richards MH, Miller SD. Mouse models of multiple sclerosis: experimental autoimmune encephalomyelitis and Theiler’s virus-induced demyelinating disease. Methods Mol Biol. 2012;900:381–401. 10.1007/978-1-60761-720-4_19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 313. Cowley TJ, Weiss SR. Murine coronavirus neuropathogenesis: determinants of virulence. J Neurovirol. 2010;16(6):427–434. 10.3109/13550284.2010.529238. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 314. Kishore A, Kanaujia A, Nag S, et al. . Different mechanisms of inflammation induced in virus and autoimmune-mediated models of multiple sclerosis in C57BL6 mice. Biomed Res Int. 2013;2013:589048 10.1155/2013/589048. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 315. Knobler RL, Tunison LA, Lampert PW, Oldstone MB. Selected mutants of mouse hepatitis virus type 4 (JHM strain) induce different CNS diseases. pathobiology of disease induced by wild type and mutants ts8 and ts15 in BALB/c and SJL/J mice. Am J Pathol. 1982;109(2):157–168. [PMC free article] [PubMed] [Google Scholar]
  • 316. [NRC] National Research Council Guide for the care and use of laboratory animals: eighth edition. Washington, DC: The National Academies Press; 2011. 10.17226/12910. https://www.nap.edu/catalog/12910/guide-for-the-care-and-use-of-laboratory-animals-eighth. [DOI] [Google Scholar]
  • 317. Clough G. Suggested guidelines for the housing and husbandry of rodents for aging studies. Neurobiol Aging. 1991;12(6):653–658. [DOI] [PubMed] [Google Scholar]
  • 318. Everds NE, Snyder PW, Bailey KL, et al. . Interpreting stress responses during routine toxicity studies: a review of the biology, impact, and assessment. Toxicol Pathol. 2013;41(4):560–614. 10.1177/0192623312466452. [DOI] [PubMed] [Google Scholar]
  • 319. Rowan AN. Refinement of animal research technique and validity of research data. Fundam Appl Toxicol. 1990;15(1):25–32. [DOI] [PubMed] [Google Scholar]
  • 320. Laber K, Veatch LM, Lopez MF, Mulligan JK, Lathers DMR. Effects of housing density on weight gain, immune function, behavior, and plasma corticosterone concentrations in BALB/c and C57BL/6 mice. J Am Assoc Lab Anim Sci. 2008;47(2):16–23. [PMC free article] [PubMed] [Google Scholar]
  • 321. Anisman H, Prakash P, Merali Z, Poulter MO. Corticotropin releasing hormone receptor alterations elicited by acute and chronic unpredictable stressor challenges in stressor-susceptible and resilient strains of mice. Behav Brain Res. 2007;181(2):180–190. 10.1016/j.bbr.2007.04.002. [DOI] [PubMed] [Google Scholar]
  • 322. David JM, Knowles S, Lamkin DM, Stout DB. Individually ventilated cages impose cold stress on laboratory mice: a source of systemic experimental variability. J Am Assoc Lab Anim Sci. 2013;52(6):738–744. [PMC free article] [PubMed] [Google Scholar]
  • 323. Ferrecchia CE, Jensen K, Van Andel R. Intracage ammonia levels in static and individually ventilated cages housing C57BL/6 mice on 4 bedding substrates. J Am Assoc Lab Anim Sci. 2014;53(2):146–151. [PMC free article] [PubMed] [Google Scholar]
  • 324. Mexas AM, Brice AK, Caro AC, Hillanbrand TS, Gaertner DJ. Nasal histopathology and intracage ammonia levels in female groups and breeding mice housed in static isolation cages. J Am Assoc Lab Anim Sci. 2015;54(5):478–486. [PMC free article] [PubMed] [Google Scholar]
  • 325. Rosenbaum MD, VandeWoude S, Volckens J, Johnson T. Disparities in ammonia, temperature, humidity, and airborne particulate matter between the micro-and macroenvironments of mice in individually ventilated caging. J Am Assoc Lab Anim Sci. 2010;49(2):177–183. [PMC free article] [PubMed] [Google Scholar]
  • 326. Bartolomucci A. Social stress, immune functions and disease in rodents. Front Neuroendocrinol. 2007;28(1):28–49. 10.1016/j.yfrne.2007.02.001. [DOI] [PubMed] [Google Scholar]
  • 327. Gordon CJ, Becker P, Ali JS. Behavioral thermoregulatory responses of single- and group-housed mice. Physiol Behav. 1998;65(2):255–262. [DOI] [PubMed] [Google Scholar]
  • 328. Maher RL, Barbash SM, Lynch DV, Swoap SJ. Group housing and nest building only slightly ameliorate the cold stress of typical housing in female C57BL/6J mice. Am J Physiol Regul Integr Comp Physiol. 2015;308(12):1070 10.1152/ajpregu.00407.2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 329. Nicholson A, Malcolm RD, Russ PL, et al. . The response of C57BL/6J and BALB/cJ mice to increased housing density. J Am Assoc Lab Anim Sci. 2009;48(6):740–753. [PMC free article] [PubMed] [Google Scholar]
  • 330. Weber EM, Dallaire JA, Gaskill BN, Pritchett-Corning KR, Garner JP. Aggression in group-housed laboratory mice: why can’t we solve the problem? Lab Anim (NY). 2017;46(4):157–161. 10.1038/laban.1219. [DOI] [PubMed] [Google Scholar]
  • 331. Gaskill BN, Gordon CJ, Pajor EA, Lucas JR, Davis JK, Garner JP. Heat or insulation: behavioral titration of mouse preference for warmth or access to a nest. PLoS One. 2012;7(3):e32799 10.1371/journal.pone.0032799. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 332. Johnson JS, Taylor DJ, Green AR, Gaskill BN. Effects of nesting material on energy homeostasis in BALB/cAnNCrl, C57BL/6NCrl, and crl:CD1(ICR) mice housed at 20 °C. J Am Assoc Lab Anim Sci. 2017;56(3):254–259. [PMC free article] [PubMed] [Google Scholar]
  • 333. Song Y, Gan Y, Wang Q, et al. . Enriching the housing environment for mice enhances their NK cell antitumor immunity via sympathetic nerve-dependent regulation of NKG2D and CCR5. Cancer Res. 2017;77(7):1611–1622. 10.1158/0008-5472.CAN-16-2143. [DOI] [PubMed] [Google Scholar]
  • 334. Van Loo PLP, Van der Meer E, Kruitwagen CLJJ, Koolhaas JM, Van Zutphen LFM, Baumans V. Long-term effects of husbandry procedures on stress-related parameters in male mice of two strains. Lab Anim. 2004;38(2):169–177. 10.1258/002367704322968858. [DOI] [PubMed] [Google Scholar]
  • 335. Arranz L, De Castro NM, Baeza I et al. . Environmental enrichment improves age-related immune system impairment: long-term exposure since adulthood increases life span in mice. Rejuvenation Res. 2010;13(4):415–428. 10.1089/rej.2009.0989. [DOI] [PubMed] [Google Scholar]
  • 336. Brod S, Gobbetti T, Gittens B, Ono M, Perretti M, D’Acquisto F. Findings from Queen Mary University of London in granulocytes provides new insights (the impact of environmental enrichment on the murine inflammatory immune response). Obes Fit Wellness Week. 2017;2(7):817 https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5374068/. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 337. Messmer MN, Kokolus KM, Eng JW, Abrams SI, Repasky EA. Mild cold-stress depresses immune responses: implications for cancer models involving laboratory mice. Bioessays. 2014;36(9):884–891. 10.1002/bies.201400066. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 338. Bailoo JD, Murphy E, Varholick JA, Novak J, Palme R, Würbel H. Evaluation of the effects of space allowance on measures of animal welfare in laboratory mice. Sci Rep. 2018;8(1):713 10.1038/s41598-017-18493-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 339. Hylander BL, Repasky EA. Thermoneutrality, mice, and cancer: a heated opinion. Trends Cancer. 2016;2(4):166–175. 10.1016/j.trecan.2016.03.005. [DOI] [PubMed] [Google Scholar]
  • 340. Hylander BL, Eng JW, Repasky EA. The impact of housing temperature-induced chronic stress on preclinical mouse tumor models and therapeutic responses: an important role for the nervous system. Adv Exp Med Biol. 2017;1036:173–189. 10.1007/978-3-319-67577-0_12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 341. Toth LA, Trammell RA, Ilsley-Woods M. Interactions between housing density and ambient temperature in the cage environment: effects on mouse physiology and behavior. J Am Assoc Lab Anim Sci. 2015;54(6):708 http://www.ncbi.nlm.nih.gov/pubmed/26632780. [PMC free article] [PubMed] [Google Scholar]
  • 342. Eichner M, Purcell JE, Fortman JD. Effects of intracage ammonia on markers of pulmonary endothelial integrity in mice housed in static microisolation cages. J Am Assoc Lab Anim Sci. 2018;57(1):18 https://www.ncbi.nlm.nih.gov/pubmed/29402347. [PMC free article] [PubMed] [Google Scholar]
  • 343. Langgartner D, Foertsch S, Füchsl AM, Reber SO. Light and water are not simple conditions: fine tuning of animal housing in male C57BL/6 mice. Stress. 2017;20(1):10–18. 10.1080/10253890.2016.1254186. [DOI] [PubMed] [Google Scholar]
  • 344. Peirson SN, Brown LA, Pothecary CA, Benson LA, Fisk AS. Light and the laboratory mouse. J Neurosci Methods. 2018;300:26–36. 10.1016/j.jneumeth.2017.04.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 345. Scheiermann C, Kunisaki Y, Frenette PS. Circadian control of the immune system. Nat Rev Immunol. 2013;13(3):190–198. 10.1038/nri3386. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 346. Kim S, Neuendorff N, Alaniz RC, Sun Y, Chapkin RS, Earnest DJ. Shift work cycle-induced alterations of circadian rhythms potentiate the effects of high-fat diet on inflammation and metabolism. FASEB J. 2018;32(6):3085–3095. 10.1096/fj.201700784R. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 347. Maestroni GJ. The photoperiod transducer melatonin and the immune-hematopoietic system. J Photochem Photobiol B. 1998;43(3):186–192. [DOI] [PubMed] [Google Scholar]
  • 348. Turner JG, Parrish JL, Hughes LF, Toth LA, Caspary DM. Hearing in laboratory animals: strain differences and nonauditory effects of noise. Comp Med. 2005;55(1):12–23. [PMC free article] [PubMed] [Google Scholar]
  • 349. Norton JN, Kinard WL, Reynolds RP. Comparative vibration levels perceived among species in a laboratory animal facility. J Am Assoc Lab Anim Sci. 2011;50(5):653–659. [PMC free article] [PubMed] [Google Scholar]
  • 350. Münzel T, Daiber A, Steven S, et al. . Effects of noise on vascular function, oxidative stress, and inflammation: mechanistic insight from studies in mice. Eur Heart J. 2017;38(37):2838–2849. 10.1093/eurheartj/ehx081. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 351. Atanasov NA, Sargent JL, Parmigiani JP, Palme R, Diggs HE. Characterization of train-induced vibration and its effect on fecal corticosterone metabolites in mice. J Am Assoc Lab Anim Sci. 2015;54(6):737–744. [PMC free article] [PubMed] [Google Scholar]
  • 352. Ambery AG, Tackett L, Penque BA, Hickman DL, Elmendorf JS. Effect of corncob bedding on feed conversion efficiency in a high-fat diet-induced prediabetic model in C57Bl/6J mice. J Am Assoc Lab Anim Sci. 2014;53(5):449–451. [PMC free article] [PubMed] [Google Scholar]
  • 353. Jacobs BB, Dieter DK. Spontaneous hepatomas in mice inbred from ha:ICR Swiss stock: effects of sex, cedar shavings in bedding, and immunization with fetal liver or hepatoma cells. J Natl Cancer Inst. 1978;61(6):1531–1534. [PubMed] [Google Scholar]
  • 354. Potgieter FJ, Törrönen R, Wilke PI. The in vitro enzyme-inducing and cytotoxic properties of South African laboratory animal contact bedding and nesting materials. Lab Anim. 1995;29(2):163–171. 10.1258/002367795780740212. [DOI] [PubMed] [Google Scholar]
  • 355. Sabine JR. Exposure to an environment containing the aromatic red cedar, Juniperus virginiana: procarcinogenic, enzyme-inducing and insecticidal effects. Toxicology. 1975;5(2):221–235. [DOI] [PubMed] [Google Scholar]
  • 356. Villalon Landeros R, Morisseau C, Yoo HJ, Fu SH, Hammock BD, Trainor BC. Corncob bedding alters the effects of estrogens on aggressive behavior and reduces estrogen receptor-α expression in the brain. Endocrinology. 2012;153(2):949–953. 10.1210/en.2011-1745. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 357. Whiteside TE, Thigpen JE, Kissling GE, Grant MG, Forsythe D. Endotoxin, coliform, and dust levels in various types of rodent bedding. J Am Assoc Lab Anim Sci. 2010;49(2):184–189. [PMC free article] [PubMed] [Google Scholar]
  • 358. [NRC] National Research Council Nutrient requirements of laboratory animals. 4th revised ed Washington, DC: The National Academies Press; 1995. 10.17226/4758. https://www.nap.edu/catalog/4758/nutrient-requirements-of-laboratory-animals-fourth-revised-edition-1995. [DOI] [PubMed] [Google Scholar]
  • 359. [NRC] National Research Council Mineral tolerance of domestic animals. Washington, DC: The National Academies Press; 1980. 10.17226/25. https://www.nap.edu/catalog/25/mineral-tolerance-of-domestic-animals. [DOI] [Google Scholar]
  • 360. [NRC] National Research Council Vitamin tolerance of animals. Washington, DC: The National Academies Press; 1987. 10.17226/949. https://www.nap.edu/catalog/949/vitamin-tolerance-of-animals. [DOI] [Google Scholar]
  • 361. Clarke HE, Coates ME, Eva JK, et al. . Dietary standards for laboratory animals: report of the laboratory animals centre diets advisory committee. Lab Anim. 1977;11(1):1–28. 10.1258/002367777780959175. [DOI] [PubMed] [Google Scholar]
  • 362. Newberne PM, Fox JG. Nutritional adequacy and quality control of rodent diets. Lab Anim Sci. 1980;30(2 Pt 2):352–365. [PubMed] [Google Scholar]
  • 363. Rao GN, Knapka JJ. Contaminant and nutrient concentrations of natural ingredient rat and mouse diet used in chemical toxicology studies. Fundam Appl Toxicol. 1987;9(2):329–338. [DOI] [PubMed] [Google Scholar]
  • 364. Mesnage R, Defarge N, Rocque L, Spiroux de Vendômois J, Séralini G. Laboratory rodent diets contain toxic levels of environmental contaminants: implications for regulatory tests. PLoS One. 2015;10(7):e0128429 10.1371/journal.pone.0128429. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 365. Jensen MN, Ritskes-Hoitinga M. How isoflavone levels in common rodent diets can interfere with the value of animal models and with experimental results. Lab Anim. 2007;41(1):1–18. 10.1258/002367707779399428. [DOI] [PubMed] [Google Scholar]
  • 366. Thigpen JE, Setchell KDR, Kissling GE, et al. . The estrogenic content of rodent diets, bedding, cages, and water bottles and its effect on bisphenol A studies. J Am Assoc Lab Anim Sci. 2013;52(2):130–141. [PMC free article] [PubMed] [Google Scholar]
  • 367. Silverman J, Adams JD. N-nitrosamines in laboratory animal feed and bedding. Lab Anim Sci. 1983;33(2):161–164. [PubMed] [Google Scholar]
  • 368. Pellizzon MA, Ricci MR. The common use of improper control diets in diet-induced metabolic disease research confounds data interpretation: the fiber factor. Nutr Metab (Lond). 2018;15:3 10.1186/s12986-018-0243-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 369. NTP Effect of dietary restriction on toxicology and carcinogenesis studies in F344/N rats and B6C3F1 mice. Natl Toxicol Program Tech Rep Ser. 1997;460:1–414. https://www.ncbi.nlm.nih.gov/pubmed/12587016. [PubMed] [Google Scholar]
  • 370. Moraal M, Leenaars PP, Arnts H, et al. . The influence of food restriction versus ad libitum feeding of chow and purified diets on variation in body weight, growth and physiology of female Wistar rats. Lab Anim. 2012;46(2):101–107. 10.1258/la.2011.011011. [DOI] [PubMed] [Google Scholar]
  • 371. Spindler SR. Review of the literature and suggestions for the design of rodent survival studies for the identification of compounds that increase health and life span. Age (Dordr). 2012;34(1):111–120. 10.1007/s11357-011-9224-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 372. Hermann LM, White WJ, Lang CM. Prolonged exposure to acid, chlorine, or tetracycline in the drinking water: effects on delayed-type hypersensitivity, hemagglutination titers, and reticuloendothelial clearance rates in mice. Lab Anim Sci. 1982;32(6):603–608. [PubMed] [Google Scholar]
  • 373. McIntyre AR, Lipman NS. Amoxicillin-clavulanic acid and trimethoprim- sulfamethoxazole in rodent feed and water: effects of compounding on antibiotic stability. J Am Assoc Lab Anim Sci. 2007;46(5):26–32. [PubMed] [Google Scholar]
  • 374. Redelsperger IM, Taldone T, Riedel ER, Lepherd ML, Lipman NS, Wolf FR. Stability of doxycycline in feed and water and minimal effective doses in tetracycline-inducible systems. J Am Assoc Lab Anim Sci. 2016;55(4):467–474. [PMC free article] [PubMed] [Google Scholar]
  • 375. Franklin CL. Microbial considerations in genetically engineered mouse research. ILAR J. 2006;47(2):141–155. 10.1093/ilar.47.2.141. https://academic.oup.com/ilarjournal/article/47/2/141/671013. Accessed May 13, 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 376. Nicklas W, Keubler L, Bleich A. Maintaining and monitoring the defined microbiota status of gnotobiotic rodents. ILAR J. 2015;56(2):241–249. https://academic.oup.com/ilarjournal/article/56/2/241/651412 10.1093/ilar/ilv029. Accessed May 13, 2018. [DOI] [PubMed] [Google Scholar]
  • 377. Hedrich HJ, Nicklas W. Chapter 4. 1—housing and maintenance In: Hedrich H, ed. The laboratory mouse. 2nd ed Boston: Academic Press; 2012:521–545. https://www.sciencedirect.com/science/article/pii/B9780123820082000222. Accessed May 13, 2018. [Google Scholar]
  • 378. Treuting PM, Clifford CB, Sellers RS, Brayton CF. Of mice and microflora: considerations for genetically engineered mice. Vet Pathol. 2012;49(1):44–63. [DOI] [PubMed] [Google Scholar]
  • 379. Dagnæs-Hansen F, Poulsen K. Pneumocystis murina infection in immunodeficient mice in a closed barrier unit: a case report. Scand J Lab Anim Sci. 2011;38(2):91–96. [Google Scholar]
  • 380. Burr HN, Lipman NS, White JR, Zheng J, Wolf FR. Strategies to prevent, treat, and provoke corynebacterium-associated hyperkeratosis in athymic nude mice. J Am Assoc Lab Anim Sci. 2011;50(3):378 https://www.ncbi.nlm.nih.gov/pubmed/21640035. Accessed May 13, 2018. [PMC free article] [PubMed] [Google Scholar]
  • 381. Santagostino SF, Arbona RJR, Nashat MA, White JR, Monette S. Pathology of aging in NOD scid gamma female mice. Vet Pathol. 2017;54(5):855–869. 10.1177/0300985817698210. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 382. Scanziani E, Gobbi A, Crippa L, et al. . Hyperkeratosis-associated coryneform infection in severe combined immunodeficient mice. Lab Anim. 1998;32(3):330–336. 10.1258/002367798780559239. [DOI] [PubMed] [Google Scholar]
  • 383. Acuff NV, LaGatta M, Nagy T, Watford WT. Severe dermatitis associated with spontaneous staphylococcus xylosus infection in rag-/-Tpl2-/- mice. Comp Med. 2017;67(4):344–349. [PMC free article] [PubMed] [Google Scholar]
  • 384. Russo M, Invernizzi A, Gobbi A, Radaelli E. Diffuse scaling dermatitis in an athymic nude mouse. Vet Pathol. 2013;50(4):722–726. 10.1177/0300985812463408. [DOI] [PubMed] [Google Scholar]
  • 385. Tao L, Reese TA. Making mouse models that reflect human immune responses. Trends Immunol. 2017;38(3):181–193. 10.1016/j.it.2016.12.007. [DOI] [PubMed] [Google Scholar]
  • 386. Ivanov II, Littman DR. Segmented filamentous bacteria take the stage. Mucosal Immunol. 2010;3(3):209–212. 10.1038/mi.2010.3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 387. Ivanov II, Atarashi K, Manel N, et al. . Induction of intestinal Th17 cells by segmented filamentous bacteria. Cell. 2009;139(3):485–498. 10.1016/j.cell.2009.09.033. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 388. Kriegel MA, Sefik E, Hill JA, Wu H, Benoist C, Mathis D. Naturally transmitted segmented filamentous bacteria segregate with diabetes protection in nonobese diabetic mice. Proc Natl Acad Sci USA. 2011;108(28):11548–11553. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 389. Lee YK, Menezes JS, Umesaki Y, Mazmanian SK. Proinflammatory T-cell responses to gut microbiota promote experimental autoimmune encephalomyelitis. Proc Natl Acad Sci USA. 2011;108(Suppl 1):4615–4622. 10.1073/pnas.1000082107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 390. Wu H, Ivanov II, Darce J, et al. . Gut-residing segmented filamentous bacteria drive autoimmune arthritis via T helper 17 cells. Immunity. 2010;32(6):815–827. 10.1016/j.immuni.2010.06.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 391. Ericsson AC, Hagan CE, Davis DJ, Franklin CL. Segmented filamentous bacteria: commensal microbes with potential effects on research. Comp Med. 2014;64(2):90–98. [PMC free article] [PubMed] [Google Scholar]
  • 392. Jiang HQ, Bos NA, Cebra JJ. Timing, localization, and persistence of colonization by segmented filamentous bacteria in the neonatal mouse gut depend on immune status of mothers and pups. Infect Immun. 2001;69(6):3611–3617. 10.1128/IAI.69.6.3611-3617.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 393. Suzuki K, Meek B, Doi Y, et al. . Aberrant expansion of segmented filamentous bacteria in IgA-deficient gut. Proc Natl Acad Sci USA. 2004;101(7):1981–1986. 10.1073/pnas.0307317101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 394. Schnupf P, Gaboriau-Routhiau V, Gros M, et al. . Growth and host interaction of mouse segmented filamentous bacteria in vitro. Nature. 2015;520(7545):99–103. 10.1038/nature14027. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 395. Wymore Brand M, Wannemuehler MJ, Phillips GJ, et al. . The altered schaedler flora: continued applications of a defined murine microbial community. ILAR J. 2015;56(2):169–178. 10.1093/ilar/ilv012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 396. Ericsson AC, Franklin CL. Manipulating the gut microbiota: methods and challenges. ILAR J. 2015;56(2):205–217. 10.1093/ilar/ilv021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 397. Friswell MK, Gika H, Stratford IJ, et al. . Site and strain-specific variation in gut microbiota profiles and metabolism in experimental mice. PLoS One. 2010;5(1):e8584 10.1371/journal.pone.0008584. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 398. Hufeldt MR, Nielsen DS, Vogensen FK, Midtvedt T, Hansen AK. Variation in the gut microbiota of laboratory mice is related to both genetic and environmental factors. Comp Med. 2010;60(5):336–347. [PMC free article] [PubMed] [Google Scholar]
  • 399. Cadwell K, Liu JY, Brown SL, et al. . A key role for autophagy and the autophagy gene Atg16l1 in mouse and human intestinal paneth cells. Nature. 2008;456(7219):259–263. 10.1038/nature07416. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 400. Cadwell K, Patel KK, Maloney NS, et al. . Virus-plus-susceptibility gene interaction determines Crohn’s disease gene Atg16L1 phenotypes in intestine. Cell. 2010;141(7):1135–1145. 10.1016/j.cell.2010.05.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 401. Kverka M, Zakostelska Z, Klimesova K, et al. . Oral administration of parabacteroides distasonis antigens attenuates experimental murine colitis through modulation of immunity and microbiota composition. Clin Exp Immunol. 2011;163(2):250–259. 10.1111/j.1365-2249.2010.04286.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 402. Denning TL, Norris BA, Medina-Contreras O, et al. . Functional specializations of intestinal dendritic cell and macrophage subsets that control Th17 and regulatory T cell responses are dependent on the T cell/APC ratio, source of mouse strain, and regional localization. J Immunol. 2011;187(2):733–747. 10.4049/jimmunol.1002701. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 403. Hart ML, Ericsson AC, Franklin CL. Differing complex microbiota alter disease severity of the IL-10-/- mouse model of inflammatory bowel disease. Front Microbiol. 2017;8:792 10.3389/fmicb.2017.00792. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 404. Peloquin JM, Nguyen DD. The microbiota and inflammatory bowel disease: insights from animal models. Anaerobe. 2013;24:102–106. 10.1016/j.anaerobe.2013.04.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 405. Chudnovskiy A, Mortha A, Kana V, et al. . Host-protozoan interactions protect from mucosal infections through activation of the inflammasome. Cell. 2016;167(2):456.e14 10.1016/j.cell.2016.08.076. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 406. Escalante NK, Lemire P, Cruz Tleugabulova M, et al. . The common mouse protozoa tritrichomonas muris alters mucosal T cell homeostasis and colitis susceptibility. J Exp Med. 2016;213(13):2841–2850. 10.1084/jem.20161776. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 407. Ericsson AC, Davis JW, Spollen W, et al. . Effects of vendor and genetic background on the composition of the fecal microbiota of inbred mice. PLoS One. 2015;10(2):e0116704 10.1371/journal.pone.0116704. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 408. Franklin CL, Ericsson AC. Microbiota and reproducibility of rodent models. Lab Anim (NY). 2017;46(4):114–122. 10.1038/laban.1222. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 409. Rosshart SP, Vassallo BG, Angeletti D, et al. . Wild mouse gut microbiota promotes host fitness and improves disease resistance. Cell. 2017;171(5):1028.e13 10.1016/j.cell.2017.09.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 410. Barthold SW. Microbes and the evolution of scientific fancy mice. ILAR J. 2008;49(3):265–271. [DOI] [PubMed] [Google Scholar]
  • 411. Arapović J, Arapović M, Golemac M, et al. . The specific NK cell response in concert with perforin prevents CD8(+) T cell-mediated immunopathology after mouse cytomegalovirus infection. Med Microbiol Immunol. 2015;204(3):335–344. 10.1007/s00430-015-0409-y. [DOI] [PubMed] [Google Scholar]
  • 412. Wetzel JL, Fensterl V, Sen GC. Sendai virus pathogenesis in mice is prevented by Ifit2 and exacerbated by interferon. J Virol. 2014;88(23):13593–13601. 10.1128/JVI.02201-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 413. Morse SS, Sakaguchi N, Sakaguchi S. Virus and autoimmunity: induction of autoimmune disease in mice by mouse T lymphotropic virus (MTLV) destroying CD4+ T cells. J Immunol. 1999;162(9):5309–5316. [PubMed] [Google Scholar]
  • 414. Jacoby Ro, Ball-Goodrich L. Chapter 4—parvoviruses In: Fox JG, Davisson MT, Quimby FW, Barthold SW, Newcomer CE, Smith AL, eds. The mouse in biomedical research. (2nd ed). Burlington: Academic Press; 2007:93–103. https://www.sciencedirect.com/science/article/pii/B9780123694546500327. Accessed May 14, 2018. [Google Scholar]
  • 415. Riolobos L, Valle N, Hernando E, Maroto B, Kann M, Almendral JM. Viral oncolysis that targets raf-1 signaling control of nuclear transport. J Virol. 2010;84(4):2090–2099. 10.1128/JVI.01550-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 416. Henderson KS, Pritchett-Corning KR, Perkins CL, et al. . A comparison of mouse parvovirus 1 infection in BALB/c and C57BL/6 mice: susceptibility, replication, shedding, and seroconversion. Comp Med. 2015;65(1):5–14. [PMC free article] [PubMed] [Google Scholar]
  • 417. Watson J. Unsterilized feed as the apparent cause of a mouse parvovirus outbreak. J Am Assoc Lab Anim Sci. 2013;52(1):83–88. [PMC free article] [PubMed] [Google Scholar]
  • 418. Beura LK, Hamilton SE, Bi K, et al. . Normalizing the environment recapitulates adult human immune traits in laboratory mice. Nature. 2016;532(7600):512 10.1038/nature17655. https://www.nature.com/articles/nature17655. Accessed Mar 4, 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 419. Roble GS, Gillespie V, Lipman NS. Infectious disease survey of mus musculus from pet stores in New York City. J Am Assoc Lab Anim Sci. 2012;51(1):37–41. [PMC free article] [PubMed] [Google Scholar]
  • 420. Mahabir E, Bauer B, Schmidt J. Rodent and germplasm trafficking: risks of microbial contamination in a high-tech biomedical world. ILAR J. 2008;49(3):347–355. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 421. Parker SE, Malone S, Bunte RM, Smith AL. Infectious diseases in wild mice (mus musculus) collected on and around the University of Pennsylvania (Philadelphia) campus. Comp Med. 2009;59(5):424 https://www.ncbi.nlm.nih.gov/pubmed/19887025. Accessed Jun 10, 2018. [PMC free article] [PubMed] [Google Scholar]
  • 422. Ehlers B, Küchler J, Yasmum N, et al. . Identification of novel rodent herpesviruses, including the first gammaherpesvirus of mus musculus. J Virol. 2007;81(15):8091–8100. 10.1128/JVI.00255-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 423. Patel SJ, Zhao G, Penna VR, et al. . A murine herpesvirus closely related to ubiquitous human herpesviruses causes T-cell depletion. J Virol. 2017;91(9). doi: 10.1128/JVI.02463-16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 424. Patel SJ, Yokoyama WM. Reply to “murine roseolovirus, historically known as murine thymic lymphotropic virus”. J Virol. 2017;91(18). doi: 10.1128/JVI.00956-17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 425. Krmpotic A, Bubic I, Polic B, Lucin P, Jonjic S. Pathogenesis of murine cytomegalovirus infection. Microbes Infect. 2003;5(13):1263–1277. [DOI] [PubMed] [Google Scholar]
  • 426. Thomas AC, Forster MR, Bickerstaff AA, et al. . Occult cytomegalovirus in vivarium-housed mice may influence transplant allograft acceptance. Transpl Immunol. 2010;23(1–2):86–91. 10.1016/j.trim.2010.03.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 427. Hughes DJ, Kipar A, Sample JT, Stewart JP. Pathogenesis of a model gammaherpesvirus in a natural host. J Virol. 2010;84(8):3949–3961. 10.1128/JVI.02085-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 428. Olivadoti M, Toth LA, Weinberg J, Opp MR. Murine gammaherpesvirus 68: a model for the study of epstein-barr virus infections and related diseases. Comp Med. 2007;57(1):44–50. [PubMed] [Google Scholar]
  • 429. Aligo J, Brosnan K, Walker M, et al. . Murine gammaherpesvirus-68 (MHV-68) is not horizontally transmitted amongst laboratory mice by cage contact. J Immunotoxicol. 2015;12(4):330–341. 10.3109/1547691x.2014.980020. [DOI] [PubMed] [Google Scholar]
  • 430. Dong S, Forrest JC, Liang X. Murine gammaherpesvirus 68: a small animal model for gammaherpesvirus-associated diseases. Adv Exp Med Biol. 2017;1018:225–236. 10.1007/978-981-10-5765-6_14. [DOI] [PubMed] [Google Scholar]
  • 431. Walsh NC, Kenney LL, Jangalwe S, et al. . Humanized mouse models of clinical disease. Annu Rev Pathol. 2017;12:187–215. 10.1146/annurev-pathol-052016-100332. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 432. Ahmed EH, Baiocchi RA. Murine models of Epstein-Barr virus-associated lymphomagenesis. ILAR J. 2016;57(1):55–62. 10.1093/ilar/ilv074. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 433. Wagar EJ, Cromwell MA, Shultz LD, et al. . Regulation of human cell engraftment and development of EBV-related lymphoproliferative disorders in hu-PBL-scid mice. J Immunol. 2000;165(1):518–527. [DOI] [PubMed] [Google Scholar]
  • 434. Radaelli E, Hermans E, Omodho L, et al. . Spontaneous post-transplant disorders in NOD.cg- prkdcscid Il2rgtm1Sug/JicTac (NOG) mice engrafted with patient-derived metastatic melanomas. PLoS One. 2015;10(5):e0124974 10.1371/journal.pone.0124974. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 435. Taurozzi AJ, Beekharry R, Wantoch M, et al. . Spontaneous development of Epstein-Barr virus associated human lymphomas in a prostate cancer xenograft program. PLoS One. 2017;12(11):e0188228 10.1371/journal.pone.0188228. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 436. Facompre ND, Sahu V, Montone KT, et al. . Barriers to generating PDX models of HPV-related head and neck cancer. Laryngoscope. 2017;127(12):2777–2783. 10.1002/lary.26679. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 437. Butler KA, Hou X, Becker MA, et al. . Prevention of human lymphoproliferative tumor formation in ovarian cancer patient-derived xenografts. Neoplasia. 2017;19(8):628–636. 10.1016/j.neo.2017.04.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 438. Corso S, Cargnelutti M, Durando S, et al. . Rituximab treatment prevents lymphoma onset in gastric cancer patient-derived xenografts. Neoplasia. 2018;20(5):443–455. 10.1016/j.neo.2018.02.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 439. Morse HC Chapter 10—retroelements in the mouse In: Fox JG, Davisson MT, Quimby FW, Barthold SW, Newcomer CE, Smith AL, eds. The mouse in biomedical research. 2nd ed Burlington: Academic Press; 2007:269–279. https://www.sciencedirect.com/science/article/pii/B9780123694546500388. Accessed May 14, 2018. [Google Scholar]
  • 440. Barthold SW, Griffey SM, Percy DH. Mouse In: Barthold SW, Griffey SM, Percy DH, eds. Pathology of laboratory rodents and rabbits. John Wiley & Sons; 2016:1–118. [Google Scholar]
  • 441. Nagata S, Suda T. Fas and fas ligand: lpr and gld mutations. Immunol Today. 1995;16(1):39–43. [DOI] [PubMed] [Google Scholar]
  • 442. Hiromatsu K, Usami J, Aoki Y, Makino M, Yoshikai Y. Accelerated progression of a murine retrovirus-induced immunodeficiency syndrome in fas mutant C57BL/6 lpr/lpr mice. Microbiol Immunol. 1997;41(3):221–227. https://www.ncbi.nlm.nih.gov/pubmed/9130234. Accessed May 14, 2018. [DOI] [PubMed] [Google Scholar]
  • 443. Rastad JL, Green WR. Myeloid-derived suppressor cells in murine AIDS inhibit B-cell responses in part via soluble mediators including reactive oxygen and nitrogen species, and TGF-β. Virology. 2016;499:9–22. 10.1016/j.virol.2016.08.031. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 444. Radaelli E, Castiglioni V, Recordati C, et al. . The pathology of aging 129S6/SvEvTac mice. Vet Pathol. 2016;53(2):477–492. 10.1177/0300985815608673. [DOI] [PubMed] [Google Scholar]
  • 445. Baudino L, Yoshinobu K, Morito N, Santiago-Raber ML, Izui S. Role of endogenous retroviruses in murine SLE. Autoimmun Rev. 2010;10(1):27–34. 10.1016/j.autrev.2010.07.012. https://www.ncbi.nlm.nih.gov/pubmed/20659589. Accessed May 14, 2018. [DOI] [PubMed] [Google Scholar]
  • 446. Hanafusa T, Miyagawa J, Nakajima H, et al. . The NOD mouse. Diabetes Res Clin Pract. 1994;24(Suppl):S307–S311. https://www.ncbi.nlm.nih.gov/pubmed/7859625. Accessed May 14, 2018. [DOI] [PubMed] [Google Scholar]
  • 447. Ito K, Baudino L, Kihara M, et al. . Three sgp loci act independently as well as synergistically to elevate the expression of specific endogenous retroviruses implicated in murine lupus. J Autoimmun. 2013;43:10 10.1016/j.jaut.2013.01.014. https://www.ncbi.nlm.nih.gov/pubmed/23465716. Accessed May 14, 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 448. Bashratyan R, Regn D, Rahman MJ, et al. . Type 1 diabetes pathogenesis is modulated by spontaneous autoimmune responses to endogenous retrovirus antigens in NOD mice. Eur J Immunol. 2017;47(3):575–584. 10.1002/eji.201646755. [DOI] [PubMed] [Google Scholar]
  • 449. Graham DM. A walk on the wild side. Lab Anim (NY). 2017;46:423–427. 10.1038/laban.1372. [DOI] [PubMed] [Google Scholar]
  • 450. Zeiss CJ, Brayton CF. Immune responses to the real world. Lab Anim (NY). 2017;47(1):13–14. 10.1038/laban.1384. [DOI] [PubMed] [Google Scholar]
  • 451. Peterson NC. From bench to cageside: risk assessment for rodent pathogen contamination of cells and biologics. ILAR J. 2008;49(3):310–315. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 452. Nicklas W, Kraft V, Meyer B. Contamination of transplantable tumors, cell lines, and monoclonal antibodies with rodent viruses. Lab Anim Sci. 1993;43(4):296–300. [PubMed] [Google Scholar]
  • 453. Almeida JL, Cole KD, Plant AL. Standards for cell line authentication and beyond. PLoS Biol. 2016;14(6):e1002476 10.1371/journal.pbio.1002476. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 454. Geraghty RJ, Capes-Davis A, Davis JM, et al. . Guidelines for the use of cell lines in biomedical research. Br J Cancer. 2014;111(6):1021–1046. 10.1038/bjc.2014.166. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 455. Meehan TF, Conte N, Goldstein T, et al. . PDX-MI: minimal information for patient-derived tumor xenograft models. Cancer Res. 2017;77(21):e66 10.1158/0008-5472.CAN-17-0582. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 456. Yu M, Selvaraj SK, Liang-Chu MMY, et al. . A resource for cell line authentication, annotation and quality control. Nature. 2015;520(7547):307–311. 10.1038/nature14397. [DOI] [PubMed] [Google Scholar]
  • 457. Pepin G, Ferrand J, Honing K, et al. . Cre-dependent DNA recombination activates a STING-dependent innate immune response. Nucleic Acids Res. 2016;44(11):5356–5364. http://www.ncbi.nlm.nih.gov/pubmed/27166376 10.1093/nar/gkw405. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 458. Behjati S, Frank MH. The effects of tamoxifen on immunity. Curr Med Chem. 2009;16(24):3076–3080. https://www.ncbi.nlm.nih.gov/pubmed/19689284 10.2174/092986709788803042. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 459. Sthoeger ZM, Zinger H, Mozes E. Beneficial effects of the anti-oestrogen tamoxifen on systemic lupus erythematosus of (NZBxNZW)F1 female mice are associated with specific reduction of IgG3 autoantibodies. Ann Rheum Dis. 2003;62(4):341–346. http://www.ncbi.nlm.nih.gov/pubmed/12634234 10.1136/ard.62.4.341. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 460. Corbo-Rodgers E, Staub ES, Zou T, Smith A, Kambayashi T, Maltzman JS. Oral ivermectin as an unexpected initiator of CreT2-mediated deletion in T cells. Nat Immunol. 2012;13(3):197–198. 10.1038/ni.2232. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 461. Bellahsene A, Forsgren A. Effect of doxycycline on immune response in mice. Infect Immun. 1985;48(2):556–559. http://iai.asm.org/content/48/2/556.abstract. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 462. Milano S, Arcoleo F, D’Agostino P, Cillari E. Intraperitoneal injection of tetracyclines protects mice from lethal endotoxemia downregulating inducible nitric oxide synthase in various organs and cytokine and nitrate secretion in blood. Antimicrob Agents Chemother. 1997;41(1):117–121. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 463. Boynton FDD, Ericsson AC, Uchihashi M, Dunbar ML, Wilkinson JE. Doxycycline induces dysbiosis in female C57BL/6NCrl mice. BMC Res Notes. 2017;10(1):644 https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5708113/ 10.1186/s13104-017-2960-7. Accessed Feb 7, 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 464. Ottina E, Peperzak V, Schoeler K, et al. . DNA-binding of the tet-transactivator curtails antigen-induced lymphocyte activation in mice. Nat Commun. 2017;8:1 10.1038/s41467-017-01022-4. https://www.nature.com/articles/s41467-017-01022-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 465. Ansari A, Ahmed A, Matsangos A, et al. . Cellular GFP toxicity and immunogenicity: potential confounders in in vivo cell tracking experiments. Stem Cell Rev. 2016;12(5):553–559. 10.1007/s12015-016-9670-8. https://www.ncbi.nlm.nih.gov/pubmed/27435468. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 466. Jeon YH, Choi Y, Kang JH, et al. . Immune response to firefly luciferase as a naked DNA. Cancer Biol Ther. 2007;6(5):781–786. [DOI] [PubMed] [Google Scholar]
  • 467. McPherson SW, Yang J, Chan C, Dou C, Gregerson DS. Resting CD8 T cells recognize beta-galactosidase expressed in the immune-privileged retina and mediate autoimmune disease when activated. Immunology. 2003;110(3):386–396. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 468. Podetz-Pedersen KM, Vezys V, Somia NV, Russell SJ, McIvor RS. Cellular immune response against firefly luciferase after sleeping beauty-mediated gene transfer in vivo. Hum Gene Ther. 2014;25(11):955–965. 10.1089/hum.2014.048. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 469. Stripecke R, Carmen Villacres M, Skelton D, Satake N, Halene S, Kohn D. Immune response to green fluorescent protein: implications for gene therapy. Gene Ther. 1999;6(7):1305–1312. [DOI] [PubMed] [Google Scholar]
  • 470. Gossa S, Nayak D, Zinselmeyer BH, McGavern DB. Development of an immunologically tolerated combination of fluorescent proteins for in vivo two-photon imaging. Sci Rep. 2014;4:6664 10.1038/srep06664. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 471. Ledford H. How the immune system could stymie some CRISPR gene therapies. http://www.nature.com/articles/d41586-018-00335-8 Updated 2018. Accessed Feb 17, 2018.
  • 472. Chew WL, Tabebordbar M, Cheng JKW, et al. . A multifunctional AAV-CRISPR-Cas9 and its host response. Nat Methods. 2016;13(10):868–874. 10.1038/nmeth.3993. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 473. Kalenova LF, Sukhovei YG, Fisher TA. Specific and nonspecific reactions of mouse immune system under the effect of short-term exposure in warm and/or cold water. Bull Exp Biol Med. 2005;140(6):720–722. 10.1007/s10517-006-0065-8. https://link.springer.com/article/10.1007/s10517-006-0065-8. Accessed Jun 17, 2018. [DOI] [PubMed] [Google Scholar]
  • 474. Karp CL. Unstressing intemperate models: how cold stress undermines mouse modeling. J Exp Med. 2012;209(6):1069 10.1084/jem.20120988. https://www.ncbi.nlm.nih.gov/pubmed/22665703. Accessed Jun 17, 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 475. Kokolus KM, Capitano ML, Lee C, et al. . Baseline tumor growth and immune control in laboratory mice are significantly influenced by subthermoneutral housing temperature. Proc Natl Acad Sci USA. 2013;110(50):20176 10.1073/pnas.1304291110. https://www.pnas.org/content/110/50/20176. Accessed Jun 17, 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 476. Nguyen KD, Qiu Y, Cui X, et al. . Alternatively activated macrophages produce catecholamines to sustain adaptive thermogenesis. Nature. 2011;480(7375):104 10.1038/nature10653. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3371761/. Accessed Jun 17, 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 477. Baldwin AL. Effects of noise on rodent physiology. East Eur J Enterp Technol. 2014;2(12(68)):72. https://escholarship.org/uc/item/04m5m3h1 10.15587/1729-4061.2014.23865. [DOI] [Google Scholar]
  • 478. Fonken LK, Lieberman RA, Weil ZM, Nelson RJ. Dim light at night exaggerates weight gain and inflammation associated with a high-fat diet in male mice. Endocrinology. 2013;154(10):3817–3825. 10.1210/en.2013-1121. https://www.ncbi.nlm.nih.gov/pubmed/23861373. Accessed Jun 17, 2018. [DOI] [PubMed] [Google Scholar]
  • 479. Gardner EM. Caloric restriction decreases survival of aged mice in response to primary influenza infection. J Gerontol A Biol Sci Med Sci. 2005;60(6):688–694. 10.1093/gerona/60.6.688. https://academic.oup.com/biomedgerontology/article/60/6/688/590315. Accessed Jun 17, 2018. [DOI] [PubMed] [Google Scholar]
  • 480. Suárez-Souto MA, Lara-Padilla E, Reyna-Garfias H, et al. . Caloric restriction modifies both innate and adaptive immunity in the mouse small intestine. J Physiol Biochem. 2012;68(2):163–173. 10.1007/s13105-011-0128-9. [DOI] [PubMed] [Google Scholar]
  • 481. Rogers NH, Walsh H, Alvarez-Garcia O, et al. . Metabolic benefit of chronic caloric restriction and activation of hypothalamic AGRP/NPY neurons in male mice is independent of ghrelin. Endocrinology. 2016;157(4):1430–1442. 10.1210/en.2015-1745. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 482. Spaulding CC, Walford RL, Effros RB. Calorie restriction inhibits the age-related dysregulation of the cytokines TNF-alpha and IL-6 in C3B10RF1 mice. Mech Ageing Dev. 1997;93(1–3):87–94. [DOI] [PubMed] [Google Scholar]
  • 483. Vega VL, De Cabo R, De Maio A. Age and caloric restriction diets are confounding factors that modify the response to lipopolysaccharide by peritoneal macrophages in C57BL/6 mice. Shock. 2004;22(3):248–253. [DOI] [PubMed] [Google Scholar]
  • 484. Brown EM, Wlodarska M, Willing BP, et al. . Diet and specific microbial exposure trigger features of environmental enteropathy in a novel murine model. Nat Commun. 2015;6:7806 10.1038/ncomms8806. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 485. Iyer SS, Chatraw JH, Tan WG, et al. . Protein energy malnutrition impairs homeostatic proliferation of memory CD8 T cells. J Immunol. 2012;188(1):77–84. 10.4049/jimmunol.1004027. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 486. Maier EA, Weage KJ, Guedes MM, et al. . Protein-energy malnutrition alters IgA responses to rotavirus vaccination and infection but does not impair vaccine efficacy in mice. Vaccine. 2013;32(1):48–53. 10.1016/j.vaccine.2013.10.072. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 487. Gruver AL, Sempowski GD. Cytokines, leptin, and stress-induced thymic atrophy. J Leukoc Biol. 2008;84(4):915–923. 10.1189/jlb.0108025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 488. Barouei J, Bendiks Z, Martinic A, et al. . Microbiota, metabolome, and immune alterations in obese mice fed a high-fat diet containing type 2 resistant starch. Mol Nutr Food Res. 2017;61(11). doi: 10.1002/mnfr.201700184. [DOI] [PubMed] [Google Scholar]
  • 489. Crevel RW, Friend JV, Goodwin BF, Parish WE. High-fat diets and the immune response of C57Bl mice. Br J Nutr. 1992;67(1):17–26. [DOI] [PubMed] [Google Scholar]
  • 490. Olefsky JM, Glass CK. Macrophages, inflammation, and insulin resistance. Annu Rev Physiol. 2010;72:219–246. 10.1146/annurev-physiol-021909-135846. [DOI] [PubMed] [Google Scholar]
  • 491. Cheng D, Wan Z, Zhang X, Li J, Li H, Wang C. Dietary chlorella vulgaris ameliorates altered immunomodulatory functions in cyclophosphamide-induced immunosuppressive mice. Nutrients. 2017;9(7). doi: 10.3390/nu9070708. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 492. Weise C, Heunemann C, Loddenkemper C et al. . Dietary docosahexaenoic acid in combination with arachidonic acid ameliorates allergen-induced dermatitis in mice. Pediatr Allergy Immunol. 2011;22(5):497–504. 10.1111/j.1399-3038.2010.01133.x. [DOI] [PubMed] [Google Scholar]
  • 493. Blewett HJ, Taylor CG. Dietary zinc deficiency in rodents: effects on T-cell development, maturation and phenotypes. Nutrients. 2012;4(6):449–466. 10.3390/nu4060449. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 494. Fraker PJ, King LE, Laakko T, Vollmer TL. The dynamic link between the integrity of the immune system and zinc status. J Nutr. 2000;130(5S Suppl):406S 10.1093/jn/130.5.1399S. [DOI] [PubMed] [Google Scholar]
  • 495. Fraker PJ, King LE. A distinct role for apoptosis in the changes in lymphopoiesis and myelopoiesis created by deficiencies in zinc. FASEB J. 2001;15(14):2572–2578. 10.1096/fj.01-0430rev. [DOI] [PubMed] [Google Scholar]
  • 496. King LE, Osati-Ashtiani F, Fraker PJ. Apoptosis plays a distinct role in the loss of precursor lymphocytes during zinc deficiency in mice. J Nutr. 2002;132(5):974–979. 10.1093/jn/132.5.974. [DOI] [PubMed] [Google Scholar]
  • 497. Ross AC. Vitamin A and retinoic acid in T cell-related immunity. Am J Clin Nutr. 2012;96(5):72S 10.3945/ajcn.112.034637. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 498. Spencer SP, Wilhelm C, Yang Q, et al. . Adaptation of innate lymphoid cells to a micronutrient deficiency promotes type 2 barrier immunity. Science. 2014;343(6169):432–437. 10.1126/science.1247606. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 499. Stephensen CB, Moldoveanu Z, Gangopadhyay NN. Vitamin A deficiency diminishes the salivary immunoglobulin A response and enhances the serum immunoglobulin G response to influenza A virus infection in BALB/c mice. J Nutr. 1996;126(1):94–102. 10.1093/jn/126.1.94. [DOI] [PubMed] [Google Scholar]
  • 500. Mora JR, Iwata M, von Andrian UH. Vitamin effects on the immune system: vitamins A and D take centre stage. Nat Rev Immunol. 2008;8(9):685–698. 10.1038/nri2378. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 501. Cooke PS, Simon L, Denslow ND. Chapter 37—endocrine disruptors In: Haschek WM, Rousseaux CG, Wallig MA, eds. Haschek and Rousseaux’s handbook of toxicologic pathology. 3rd ed Boston: Academic Press; 2013:1123–1154. https://www.sciencedirect.com/science/article/pii/B9780124157590000376. Accessed Jun 18, 2018. [Google Scholar]
  • 502. Pestka JJ. Deoxynivalenol-induced IgA production and IgA nephropathy-aberrant mucosal immune response with systemic repercussions. Toxicol Lett. 2003;140–141:287–295. [DOI] [PubMed] [Google Scholar]
  • 503. Xu J, Huang G, Guo TL. Developmental bisphenol A exposure modulates immune-related diseases. Toxics. 2016;4(4). doi: 10.3390/toxics4040023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 504. Kuper CF, Ruehl-Fehlert C, Elmore SA, Parker GA. Chapter 49—immune system In: Haschek WM, Rousseaux CG, Wallig MA, eds. Haschek and Rousseaux’s handbook of toxicologic pathology. 3rd ed Boston: Academic Press; 2013:1795–1862. https://www.sciencedirect.com/science/article/pii/B9780124157590000492. Accessed Jun 18, 2018. [Google Scholar]
  • 505. Schecter AJ, Olson J, Papke O. Exposure of laboratory animals to polychlorinated dibenzodioxins and polychlorinated dibenzofurans from commerical rodent chow. Chemosphere. 1996;32(3):501–508. [DOI] [PubMed] [Google Scholar]
  • 506. Kozul CD, Ely KH, Enelow RI, Hamilton JW. Low-dose arsenic compromises the immune response to influenza A infection in vivo. Environ Health Perspect. 2009;117(9):1441–1447. 10.1289/ehp.0900911. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 507. de Souza MS, Smith AL. Characterization of accessory cell function during acute infection of BALB/cByJ mice with mouse hepatitis virus (MHV), strain JHM. Lab Anim Sci. 1991;41(2):112–118. [PubMed] [Google Scholar]
  • 508. Godfraind C, Holmes KV, Coutelier JP. Thymus involution induced by mouse hepatitis virus A59 in BALB/c mice. J Virol. 1995;69(10):6541–6547. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 509. Jolicoeur P, Lamontagne L. Impairment of bone marrow pre-B and B cells in MHV3 chronically-infected mice. Adv Exp Med Biol. 1995;380:193–195. [DOI] [PubMed] [Google Scholar]
  • 510. Brownstein DG, Weir EC. Immunostimulation in mice infected with sendai virus. Am J Vet Res. 1987;48(12):1692–1696. [PubMed] [Google Scholar]
  • 511. Clark EA, Russell PH, Egghart M, Horton MA. Characteristics and genetic control of NK-cell-mediated cytotoxicity activated by naturally acquired infection in the mouse. Int J Cancer. 1979;24(5):688–699. [DOI] [PubMed] [Google Scholar]
  • 512. Streilein JW, Shadduck JA, Pakes SP. Effects of splenectomy and sendai virus infection on rejection of male skin isografts by pathogen-free C57BL/6 female mice. Transplantation. 1981;32(1):34–37. [DOI] [PubMed] [Google Scholar]
  • 513. Kay MM. Long term subclinical effects of parainfluenza (SENDAI) infection on immune cells of aging mice. Proc Soc Exp Biol Med. 1978;158(3):326–331. [DOI] [PubMed] [Google Scholar]
  • 514. Hsu CC, Paik J, Treuting PM, et al. . Infection with murine norovirus 4 does not alter helicobacter-induced inflammatory bowel disease in Il10(-/-) mice. Comp Med. 2014;64(4):256–263. [PMC free article] [PubMed] [Google Scholar]
  • 515. Mumphrey SM, Changotra H, Moore TN, et al. . Murine norovirus 1 infection is associated with histopathological changes in immunocompetent hosts, but clinical disease is prevented by STAT1-dependent interferon responses. J Virol. 2007;81(7):3251–3263. 10.1128/JVI.02096-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 516. Hsu CC, Piotrowski SL, Meeker SM, Smith KD, Maggio-Price L, Treuting PM. Histologic lesions induced by murine norovirus infection in laboratory mice. Vet Pathol. 2016;53(4):754–763. 10.1177/0300985815618439. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 517. Andrews DM, Andoniou CE, Granucci F, Ricciardi-Castagnoli P, Degli-Esposti MA. Infection of dendritic cells by murine cytomegalovirus induces functional paralysis. Nat Immunol. 2001;2(11):1077–1084. 10.1038/ni724. [DOI] [PubMed] [Google Scholar]
  • 518. O’Donoghue HL, Lawson CM, Reed WD. Autoantibodies to cardiac myosin in mouse cytomegalovirus myocarditis. Immunology. 1990;71(1):20–28. [PMC free article] [PubMed] [Google Scholar]
  • 519. Popkin DL, Watson MA, Karaskov E, Dunn GP, Bremner R, Virgin HW. Murine cytomegalovirus paralyzes macrophages by blocking IFN gamma-induced promoter assembly. Proc Natl Acad Sci USA. 2003;100(24):14309–14314. 10.1073/pnas.1835673100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 520. Trgovcich J, Stimac D, Polić B, et al. . Immune responses and cytokine induction in the development of severe hepatitis during acute infections with murine cytomegalovirus. Arch Virol. 2000;145(12):2601–2618. [DOI] [PubMed] [Google Scholar]
  • 521. Wood BA, Dutz W, Cross SS. Neonatal infection with mouse thymic virus: spleen and lymph node necrosis. J Gen Virol. 1981;57(Pt 1):139–147. 10.1099/0022-1317-57-1-139. [DOI] [PubMed] [Google Scholar]
  • 522. McKisic MD, Macy JD, Delano ML, Jacoby RO, Paturzo FX, Smith AL. Mouse parvovirus infection potentiates allogeneic skin graft rejection and induces syngeneic graft rejection. Transplantation. 1998;65(11):1436–1446. [DOI] [PubMed] [Google Scholar]
  • 523. McKisic MD, Paturzo FX, Smith AL. Mouse parvovirus infection potentiates rejection of tumor allografts and modulates T cell effector functions. Transplantation. 1996;61(2):292–299. [DOI] [PubMed] [Google Scholar]
  • 524. Segovia JC, Bueren JA, Almendral JM. Myeloid depression follows infection of susceptible newborn mice with the parvovirus minute virus of mice (strain i). J Virol. 1995;69(5):3229–3232. http://jvi.asm.org/content/69/5/3229.abstract. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 525. Gaskins HR, Prochazka M, Hamaguchi K, Serreze DV, Leiter EH. Beta cell expression of endogenous xenotropic retrovirus distinguishes diabetes-susceptible NOD/lt from resistant NON/lt mice. J Clin Invest. 1992;90(6):2220–2227. 10.1172/JCI116107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 526. Morse HC, Yetter RA, Via CS, et al. . Functional and phenotypic alterations in T cell subsets during the course of MAIDS, a murine retrovirus-induced immunodeficiency syndrome. J Immunol. 1989;143(3):844–850. [PubMed] [Google Scholar]
  • 527. Morse HC, Chattopadhyay SK, Makino M, Fredrickson TN, Hügin AW, Hartley JW. Retrovirus-induced immunodeficiency in the mouse: MAIDS as a model for AIDS. AIDS. 1992;6(7):607–621. [DOI] [PubMed] [Google Scholar]
  • 528. Mosier DE, Yetter RA, Morse HC. Retroviral induction of acute lymphoproliferative disease and profound immunosuppression in adult C57BL/6 mice. J Exp Med. 1985;161(4):766–784. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 529. Tsumura H, Miyazawa M, Ogawa S, Wang JZ, Ito Y, Shimura K. Detection of endogenous retrovirus antigens in NOD mouse pancreatic beta-cells. Lab Anim. 1998;32(1):86–94. 10.1258/002367798780559464. [DOI] [PubMed] [Google Scholar]
  • 530. Li W, Hofer MJ, Jung SR, Lim S, Campbell IL. IRF7-dependent type I interferon production induces lethal immune-mediated disease in STAT1 knockout mice infected with lymphocytic choriomeningitis virus. J Virol. 2014;88(13):7578–7588. 10.1128/JVI.03117-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 531. Oldstone MBA. Biology and pathogenesis of lymphocytic choriomeningitis virus infection. Curr Top Microbiol Immunol. 2002;263:83–117. [DOI] [PubMed] [Google Scholar]
  • 532. Zhou X, Ramachandran S, Mann M, Popkin DL. Role of lymphocytic choriomeningitis virus (LCMV) in understanding viral immunology: past, present and future. Viruses. 2012;4(11):2650–2669. 10.3390/v4112650. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 533. Foreman O, Kavirayani AM, Griffey SM, Reader R, Shultz LD. Opportunistic bacterial infections in breeding colonies of the NSG mouse strain. Vet Pathol. 2011;48(2):495–499. 10.1177/0300985810378282. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 534. Godfrey VL. Chapter 20—fungal diseases in laboratory mice In: Fox JG, Davisson MT, Quimby FW, Barthold SW, Newcomer CE, Smith AL, eds. The mouse in biomedical research. 2nd ed Burlington: Academic Press; 2007:507–515. https://www.sciencedirect.com/science/article/pii/B9780123694546500480. Accessed Jun 22, 2018. [Google Scholar]
  • 535. Kobayashi-Sakamoto M, Tamai R, Isogai E, Kiyoura Y. Gastrointestinal colonisation and systemic spread of Candida albicans in mice treated with antibiotics and prednisolone. Microb Pathog. 2018;117:191–199. 10.1016/j.micpath.2018.02.043. [DOI] [PubMed] [Google Scholar]
  • 536. Odds FC. Candida and candidosis: F. C. Odds. Baltimore: University Park Press; 1978. https://trove.nla.gov.au/version/22995412. Accessed Jun 22, 2018. [Google Scholar]
  • 537. Furuta T, Nagata T, Kikuchi T, Kikutani H. Fatal spontaneous pneumocystosis in CD40 knockout mice. J Eukaryot Microbiol. 2001;Suppl:129S–130S. https://www.ncbi.nlm.nih.gov/pubmed/11906023. [DOI] [PubMed] [Google Scholar]
  • 538. Sakala IG, Chaudhri G, Scalzo AA, et al. . Evidence for persistence of ectromelia virus in inbred mice, recrudescence following immunosuppression and transmission to naïve mice. PLoS Pathog. 2015;11(12):e1005342 10.1371/journal.ppat.1005342. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 539. Ingle A, Ghim S, Joh J, Chepkoech I, Bennett Jenson A, Sundberg JP. Novel laboratory mouse papillomavirus (MusPV) infection. Vet Pathol. 2011;48(2):500–505. 10.1177/0300985810377186. [DOI] [PubMed] [Google Scholar]
  • 540. MacDuff DA, Reese TA, Kimmey JM, et al. . Phenotypic complementation of genetic immunodeficiency by chronic herpesvirus infection. Elife. 2015;4 10.7554/eLife.04494. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 541. Labelle P, Hahn NE, Fraser JK, et al. . Mousepox detected in a research facility: case report and failure of mouse antibody production testing to identify ectromelia virus in contaminated mouse serum. Comp Med. 2009;59(2):180–186. [PMC free article] [PubMed] [Google Scholar]
  • 542. Fenske TS, McMahon C, Edwin D, et al. . Identification of candidate alkylator-induced cancer susceptibility genes by whole genome scanning in mice. Cancer Res. 2006;66(10):5029–5038. 10.1158/0008-5472.CAN-05-3404. [DOI] [PubMed] [Google Scholar]
  • 543. Haggerty HG, Holsapple MP. Role of metabolism in dimethylnitrosamine-induced immunosuppression: a review. Toxicology. 1990;63(1):1–23. [DOI] [PubMed] [Google Scholar]
  • 544. Shimada Y, Nishimura M, Kakinuma S, et al. . Genetic susceptibility to thymic lymphomas and K-ras gene mutation in mice after exposure to X-rays and N-ethyl-N-nitrosourea. Int J Radiat Biol. 2003;79(6):423–430. [DOI] [PubMed] [Google Scholar]
  • 545. Freund YR, Dousman L, MacGregor JT, Mohagheghpour N. Oral treatment with trimethoprim-sulfamethoxazole and zidovudine suppresses murine accessory cell-dependent immune responses. Toxicol Sci. 2000;55(2):335–342. [DOI] [PubMed] [Google Scholar]
  • 546. Blakley BR, Rousseaux CG. Effect of ivermectin on the immune response in mice. Am J Vet Res. 1991;52(4):593–595. [PubMed] [Google Scholar]
  • 547. Yan S, Ci X, Chen N, et al. . Anti-inflammatory effects of ivermectin in mouse model of allergic asthma. Inflamm Res. 2011;60(6):589–596. 10.1007/s00011-011-0307-8. [DOI] [PubMed] [Google Scholar]
  • 548. Berndt A, Ackert-Bicknell C, Silva KA, et al. . Genetic determinants of fibro-osseous lesions in aged inbred mice. Exp Mol Pathol. 2016;100(1):92–100. 10.1016/j.yexmp.2015.11.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 549. Brown N, Nagarkatti M, Nagarkatti PS. Diethylstilbestrol alters positive and negative selection of T cells in the thymus and modulates T-cell repertoire in the periphery. Toxicol Appl Pharmacol. 2006;212(2):119–126. 10.1016/j.taap.2005.07.012. [DOI] [PubMed] [Google Scholar]
  • 550. Kalland T, Forsberg JG. Natural killer cell activity and tumor susceptibility in female mice treated neonatally with diethylstilbestrol. Cancer Res. 1981;41(12 Pt 1):5134–5140. [PubMed] [Google Scholar]
  • 551. Khan D, Ansar Ahmed S. The immune system is a natural target for estrogen action: opposing effects of estrogen in two prototypical autoimmune diseases. Front Immunol. 2015;6:635 10.3389/fimmu.2015.00635. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 552. Lindberg MK, Weihua Z, Andersson N, et al. . Estrogen receptor specificity for the effects of estrogen in ovariectomized mice. J Endocrinol. 2002;174(2):167–178. [DOI] [PubMed] [Google Scholar]
  • 553. Arredouani MS. New insights into androgenic immune regulation. Oncoimmunology. 2014;3(9):e954968 10.4161/21624011.2014.954968. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 554. Kissick HT, Sanda MG, Dunn LK, et al. . Androgens alter T-cell immunity by inhibiting T-helper 1 differentiation. Proc Natl Acad Sci USA. 2014;111(27):9887–9892. 10.1073/pnas.1402468111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 555. Ramaiah L, Bounous DI, Elmore SA. Chapter 50—hematopoietic system In: Haschek WM, Rousseaux CG, Wallig MA, eds. Haschek and Rousseaux’s handbook of toxicologic pathology. 3rd ed Boston: Academic Press; 2013:1863–1933. https://www.sciencedirect.com/science/article/pii/B9780124157590000509. Accessed Jun 23, 2018. [Google Scholar]
  • 556. Roden AC, Moser MT, Tri SD, et al. . Augmentation of T cell levels and responses induced by androgen deprivation. J Immunol. 2004;173(10):6098–6108. [DOI] [PubMed] [Google Scholar]
  • 557. Muller YD, Golshayan D, Ehirchiou D, et al. . Immunosuppressive effects of streptozotocin-induced diabetes result in absolute lymphopenia and a relative increase of T regulatory cells. Diabetes. 2011;60(9):2331–2340. 10.2337/db11-0159. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 558. Chen H, Dorrigan A, Saad S, Hare DJ, Cortie MB, Valenzuela SM. In vivo study of spherical gold nanoparticles: inflammatory effects and distribution in mice. PLoS One. 2013;8(2):e58208 10.1371/journal.pone.0058208. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 559. Ilinskaya AN, Dobrovolskaia MA. Immunosuppressive and anti-inflammatory properties of engineered nanomaterials. Br J Pharmacol. 2014;171(17):3988–4000. 10.1111/bph.12722. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 560. Mitchell LA, Lauer FT, Burchiel SW, McDonald JD. Mechanisms for how inhaled multiwalled carbon nanotubes suppress systemic immune function in mice. Nat Nanotechnol. 2009;4(7):451–456. 10.1038/nnano.2009.151. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 561. Shen C, Liang H, Wang C, Liao M, Jan T. Iron oxide nanoparticles suppressed T helper 1 cell-mediated immunity in a murine model of delayed-type hypersensitivity. Int J Nanomedicine. 2012;7:2729–2737. 10.2147/IJN.S31054. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 562. Su C, Chen T, Chang C, et al. . Comparative proteomics of inhaled silver nanoparticles in healthy and allergen provoked mice. Int J Nanomedicine. 2013;8:2783–2799. 10.2147/IJN.S46997. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 563. Sumbayev VV, Yasinska IM, Garcia CP, et al. . Gold nanoparticles downregulate interleukin-1β-induced pro-inflammatory responses. Small. 2013;9(3):472–477. 10.1002/smll.201201528. [DOI] [PubMed] [Google Scholar]
  • 564. Baklaushev VP, Kilpeläinen A, Petkov S, et al. . Luciferase expression allows bioluminescence imaging but imposes limitations on the orthotopic mouse (4T1) model of breast cancer. Sci Rep. 2017;7(1):7715 10.1038/s41598-017-07851-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 565. Yang Y, Haecker SE, Su Q, Wilson JM. Immunology of gene therapy with adenoviral vectors in mouse skeletal muscle. Hum Mol Genet. 1996;5(11):1703–1712. [DOI] [PubMed] [Google Scholar]
  • 566. Ernst W. Humanized mice in infectious diseases. Comp Immunol Microbiol Infect Dis. 2016;49:29–38. 10.1016/j.cimid.2016.08.006. [DOI] [PubMed] [Google Scholar]
  • 567. Skelton JK, Ortega-Prieto AM, Dorner M. A hitchhiker’s guide to humanized mice: new pathways to studying viral infections. Immunology. 2018;154(1):50–61. 10.1111/imm.12906. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 568. Festing MFW. Genetically defined strains in drug development and toxicity testing. Methods Mol Biol. 2016;1438:1–17. 10.1007/978-1-4939-3661-8_1. [DOI] [PubMed] [Google Scholar]
  • 569. Festing MFW, Nevalainen T. The design and statistical analysis of animal experiments: introduction to this issue. ILAR J. 2014;55(3):379–382. 10.1093/ilar/ilu046. [DOI] [PubMed] [Google Scholar]
  • 570. Brodin P, Davis MM. Human immune system variation. Nat Rev Immunol. 2017;17(1):21 10.1038/nri.2016.125. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5328245/. Accessed Mar 3, 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 571. Davis MM, Tato CT, Furman D. Systems immunology: just getting started. Nat Immunol 2017;18(7):725 10.1038/ni.3768. https://search.proquest.com/docview/1917969297. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 572. Graham JB, Swarts JL, Mooney M, et al. . Extensive homeostatic T cell phenotypic variation within the collaborative cross. Cell Rep. 2017;21(8):2313–2325. 10.1016/j.celrep.2017.10.093. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 573. Phillippi J, Xie Y, Miller DR, et al. . Using the emerging collaborative cross to probe the immune system. Genes Immun. 2014;15(1):38–46. 10.1038/gene.2013.59. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4004367/. Accessed Dec 15, 2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 574. Durrant C, Tayem H, Yalcin B, et al. . Collaborative cross mice and their power to map host susceptibility to aspergillus fumigatus infection. Genome Res. 2011;21(8):1239–1248. 10.1101/gr.118786.110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 575. Graham JB, Thomas S, Swarts J, et al. . Genetic diversity in the collaborative cross model recapitulates human west nile virus disease outcomes. mBio. 2015;6(3):493 10.1128/mBio.00493-15. http://mbio.asm.org/content/6/3/e00493-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 576. Gralinski LE, Ferris MT, Aylor DL, et al. . Genome wide identification of SARS-CoV susceptibility loci using the collaborative cross. PLoS Genet. 2015;11(10):e1005504 10.1371/journal.pgen.1005504. https://www.ncbi.nlm.nih.gov/pubmed/26452100. Accessed Dec 15, 2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 577. Green R, Wilkins C, Thomas S, et al. . Transcriptional profiles of WNV neurovirulence in a genetically diverse collaborative cross population. Genom Data. 2016;10:137–140. 10.1016/j.gdata.2016.10.005. http://europepmc.org/abstract/MED/27872814, http://europepmc.org/articles/PMC5107684/?report=abstract. Accessed Dec 15, 2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 578. Churchill GA, Gatti DM, Munger SC, Svenson KL. The diversity outbred mouse population. Mamm Genome. 2012;23(9–10):713–718. 10.1007/s00335-012-9414-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 579. Gatti DM, Svenson KL, Shabalin A, et al. . Quantitative trait locus mapping methods for diversity outbred mice. G3 (Bethesda). 2014;4(9):1623–1633. 10.1534/g3.114.013748. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 580. Logan RW, Robledo RF, Recla JM, et al. . High-precision genetic mapping of behavioral traits in the diversity outbred mouse population. Genes Brain Behav. 2013;12(4):424–437. 10.1111/gbb.12029. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 581. Svenson KL, Gatti DM, Valdar W, et al. . High-resolution genetic mapping using the mouse diversity outbred population. Genetics. 2012;190(2):437–447. 10.1534/genetics.111.132597. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 582. Chia R, Achilli F, Festing MFW, Fisher EMC. The origins and uses of mouse outbred stocks. Nat Genet. 2005;37(11):1181–1186. 10.1038/ng1665. [DOI] [PubMed] [Google Scholar]
  • 583. Yalcin B, Flint J. Association studies in outbred mice in a new era of full-genome sequencing. Mamm Genome. 2012;23(9–10):719–726. 10.1007/s00335-012-9409-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 584. Rivera J, Tessarollo L. Genetic background and the dilemma of translating mouse studies to humans. Immunity. 2008;28(1):1–4. 10.1016/j.immuni.2007.12.008. http://www.sciencedirect.com/science/article/pii/S1074761307005912. Accessed Feb 26, 2018. [DOI] [PubMed] [Google Scholar]
  • 585. Marx JO, Gaertner DJ, Smith AL. Results of survey regarding prevalence of adventitial infections in mice and rats at biomedical research facilities. J Am Assoc Lab Anim Sci. 2017;56(5):527–533. [PMC free article] [PubMed] [Google Scholar]
  • 586. Mähler Convenor M, Berard M, Feinstein R, et al. . FELASA recommendations for the health monitoring of mouse, rat, hamster, guinea pig and rabbit colonies in breeding and experimental units. Lab Anim. 2014;48(3):178–192. 10.1177/0023677213516312. [DOI] [PubMed] [Google Scholar]
  • 587. Pritchett-Corning KR, Cosentino J, Clifford CB. Contemporary prevalence of infectious agents in laboratory mice and rats. Lab Anim. 2009;43(2):165–173. 10.1258/la.2008.008009. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

ily026_Suppl_Table-1_ER_06-05-2019
ily026_Suppl-Table-2_Revision_ER_06-06-2019

Articles from ILAR Journal are provided here courtesy of Oxford University Press

RESOURCES