Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2014 Nov 8.
Published in final edited form as: Sci China Life Sci. 2010 Mar 7;53(2):195–203. doi: 10.1007/s11427-010-0059-7

HIV-1 immunopathogenesis in humanized mouse models

Liguo Zhang 1, Lishan Su 1,2
PMCID: PMC4224686  NIHMSID: NIHMS636139  PMID: 20596827

Abstract

In recent years, the technology of constructing chimeric mice with humanized immune systems has markedly improved. Multiple lineages of human immune cells develop in immunodeficient mice that have been transplanted with human hematopoietic stem cells. More importantly, these mice mount functional humoral and cellular immune responses upon immunization and microbial infection. Human immunodeficiency virus type I (HIV-1) can establish an infection in humanized mice, resulting in CD4+ T-cell depletion and an accompanying nonspecific immune activation, which mimics the immunopathology in HIV-1-infected human patients. This makes humanized mice an optimal model for studying the mechanisms of HIV-1 immunopathogenesis and for developing novel immune-based therapies.

INTRODUCTION

Human immunodeficiency virus type I (HIV-1) infection is characterized by progressive CD4+ T-cell depletion and acquired immunodeficiency syndrome (AIDS). Approximately 60 million people have been infected with HIV-1, and half of them have died from AIDS-related diseases.1 After more than 30 years of extensive research, the precise mechanism by which HIV-1 infection leads to immunodeficiency is still poorly understood, mainly as a result of the lack of robust small animal models. The recent development of humanized mice with functional humanized immune systems may help to improve our understanding of HIV-1 pathogenesis and lead to new treatments.

A BRIEF HISTORY OF THE HUMANIZED MOUSE MODEL

In this review, humanized mice are defined as immunodeficient mice that have been transplanted with human hematopoietic stem cells (HSCs), lymphoid tissue or peripheral blood cells. Early attempts to reconstitute the human immune system in nude mice (which lack T cells) were unsuccessful because of the significant rejection mediated by the remaining mouse B and natural killer (NK) cells.2 The first breakthrough in this field came with the development of CB17-SCID (SCID) mice,3 which lack both T and B lymphocytes. Human peripheral blood leukocytes (SCID-hu PBL)4 and human fetal liver and thymus tissue (SCID-hu Thy/Liv)5 were successfully reconstituted in SCID mice. Non-obese diabetic (NOD)/SCID mice exhibit additional defects in T, B, NK cell and macrophage function6 and thus are superior to SCID mice at accommodating human peripheral mononuclear cells (PBMCs)7 and HSCs.8 However, these early models have limitations. The SCID-hu PBL mice lack human lymphoid organs and develop severe graft-versus-host disease mediated by xeno-reactive donor T cells. In contrast, the SCID-hu Thy/Liv mice have very low levels of human cells in the blood and peripheral organs. Collectively, the lack of human cells in the peripheral lymphoid organs and the inability to mount functional immune responses limit the applicability of these early humanized models.

RECENT PROGRESS IN HUMANIZED MOUSE MODELS

It was reported that depletion of NK cells by antibody treatment significantly increases human HSC engraftment efficiency in NOD/SCID mice.9 This finding encouraged the generation of mice that are completely devoid of T, B and NK cells (reviewed by Ito et al.10 and Shultz et al.11). These newly developed immunodeficient mice allowed much better human HSC reconstitution and significant improvements in human immune function. In addition to the development of novel immunodeficient mouse strains, more efforts have been made to enhance engraftment, such as by introducing human cytokines,1214 by using human leukocyte antigen (HLA) transgenics,15 and by inhibiting mouse macrophage function.16

Mice lacking T, B and NK cells

The interleukin-2 (IL-2) receptor gamma chain (IL2Rγ) is a common signaling component of IL-2, IL-4, IL-7, IL-9, IL-15 and IL-21 signaling. The absence of IL2Rγ blocks NK cell development as a result of the ablation of IL-7 and IL-15 signaling. Efficient multilineage hematopoiesis was first reported in NOD/Shi-scid Il2rgnull (NOG) mice after human HSC transplantation (NOG-hu HSC),17 and a subsequent study showed similar human immune cell differentiation in rag2−/− Il2rgnull mice (DKO-hu HSC). More importantly, functional human immune responses were observed in DKO-hu HSC mice, including antigen-specific T cells and antibody production in respond to immunization and microbial infection.18,19 Several other mutant mouse strains with an Il2rg gene knockout have been successfully developed, such as NOD/LtSZ-scid Il2rgnull (NSG),19,20 NOD-rag1−/−Il2rgnull (NRG)21 and rag1−/− Il2rgnull mice.22 It is worth noting that NSG mice have been shown to support increased human cell engraftment over the other strains.2224

Inhibition of mouse macrophages

In addition to T, B and NK cells, macrophages also contribute to xenograft rejection. Signal regulatory protein alpha (SIRPα) is an inhibitory receptor that is highly expressed on myeloid cells, whereas its ligand CD47 is expressed on all cell types. Ligation of SIRPα by CD47 inhibits macrophage phagocytosis, which contributes to the recognition of self and non-self by innate immunity.25 Additionally, this CD47–SIRPα interaction also plays an important role in macrophage-mediated xenograft rejection in humanized mice. The SIRPα of NOD mice shows enhanced binding to human CD47, which results in reduced rejection and improved human cell reconstitution.26 These polymorphisms of the sirpa gene may at least partially explain why NSG mice are more efficient than DKO mice in supporting human HSC transplant.22,27 It was recently reported that HSC transduction with mouse CD47 by a lentiviral vector led to increased engraftment in humanized mice.28 Meanwhile, human sirpa gene-transgenic DKO mice support improved human cell reconstitution and a stronger antigen-specific immune response.16

Improvement of graft efficiency by introducing human cytokines

Many mouse cytokines are poorly crossreactive with their human receptors, so supplementing human cytokines in trans can improve the development and differentiation of certain cell lineages in humanized mice: such cytokines include IL-7 for T cells,29 IL-15 for NK cells,12,30 erythropoietin for erythrocytes, and granulocyte-macrophage colony-stimulating factor (GM-CSF) IL-4 and macrophage colony-stimulating factor (M-CSF) for monocytes/macrophages.12,31

Recently, progress has been made by knock-in replacement of mouse cytokines with their human counterparts.32 Because transcription of the knock-in genes is controlled by mouse regulatory elements, the genes are expressed at the correct time, in the correct location and at physiological levels. Moreover, the replacements lead to defects in the targeted mouse cells, thus providing a competitive advantage to human cells. Three mouse strains have been developed with this technology to produce human thrombopoietin,14 human IL-3/GM-CSF13 and M-CSF.33 The thrombopoietin replacement results in better maintenance of human HSC and higher levels of human cell engraftment.14 The human IL-3/GM-CSF13 and M-CSF33 knock-in genes dramatically improve myeloid cell differentiation and function.

Human HLA transgenic mice

In humanized mice, human T cells are educated in the mouse thymus by both mouse thymic epithelial cells and human bone marrow-derived cells.18,19 The T-cell receptor affinity and specificity may be different from those in humans with matched MHC types.34 Transgenic expression of human HLA-A2 (MHC I) significantly improves human CD8+ T-cell responses to both Epstein–Barr virus (EBV)34,35 and dengue virus36 in infected mice. Interestingly, EBV-infected humanized mice with the HLA-A2 transgene generate antigen-specific T cells to lytic EBV antigens that predominate over T cells specific to latent antigens, which is similar to the T-cell response in human EBV carriers.34 Significantly increased human cell reconstitution and better immune responses, including immunoglobulin class switching and elevated human IgG responses, were also observed in HLA-DR4 (MHC II) transgenic mice.37,38

Other factors affecting human cell engraftment

In addition to the mouse genetic background, there are other factors that may affect human cell reconstitution. First, co-transplant of human fetal thymus with autologous HSC will significantly increase human immune reconstitution and function in NOD/SCID mice.39,40 Mice transplanted with human fetal thymus and liver tissue in addition to HSC are called BLT mice.39,40 BLT mice have been constructed on both NOD/SCID and NSG backgrounds, and the reconstitution of NSG-BLT has proved to be better than NOD/SCID-BLT.24 It has also been demonstrated that newborn mice (less than 3 days) support higher transplant efficiency.18,19,27,41 Mouse gender was found to play a role in accommodating human HSC grafts because engraftment of human hematopoietic stem cells was more efficient in female NSG recipient mice than in male mice.23,42

HIV-1 INFECTION IN HUMANIZED MICE

Early generations of humanized mice were developed to study HIV-1 infection,43,44 and the SCID-hu Thy/Liv model is still being used to test antiviral drugs (Table 1).4547 However, these models are limited in the modeling of HIV-1 immunopathogenesis owing to the lack of a functional immune system. In the improved humanized mice, several HIV-1 strains have been successfully used for infection. These include CCR5-tropic (JR-CSF,48,49 Yu-2,50 BAL,51,52 ADA53 and NFN-SX52,53), CXCR4-tropic (NL4-3)50,51 and dual-tropic (NL4-R3A) viruses.48,54 HIV-1 infection can be established by inoculation through intraperitoneal,50,51,53,55 intravenous48,49 or mucosal routes.56 Sustained viral replication and CD4+ T-cell depletion were observed by all routes of infection. As is the case for HIV-1 infected patients, CXCR4-tropic HIV-1 quickly depletes both CD45RA+ naive and CD45RA effector/memory CD4+ T lymphocytes, whereas CCR5-tropic HIV-1 preferentially depletes CD45RACD4+ T lymphocytes.57

Table 1.

HIV infection in current humanized mouse models

Research areas Models References
HIV-1 evolution DKO-hu HSC Ince et al.,58 2010
NOG-hu HSC Sato et al.,59 2010
Immune response NOG-hu HSC Nie et al.,57 2009
NOG-hu HSC Sato et al.,60 2010
NSG-BLT Brainard et al.,52 2009
NOD/SCID-BLT Brainard et al.,52 2009
Latency DKO-HSC Choudhary et al.,61 2012
NSG-BLT Denton et al.,62 2012
Marsden et al.,63 2012
Mucosal transmission and prevention DKO-hu HSC Berges et al.,64 2008
Hofer et al.,65 2008
Neff et al.,66 2010
Rag1−/− γC−/−-hu HSC Akkina et al.,67 2011
NSG-BLT Denton et al.,68 2011
Stoddart et al.,24 2011
Wheeler et al.,69 2011
NOD/SCID-BLT Sun et al.,56 2007
Denton et al.,70 2008
Denton et al.,71 2010
Denton et al.,68 2011
Stoddart et al.,24 2011
Immune activation and pathogenesis Tregs DKO-hu HSC Jiang et al.,54 2008
GALT and mucosal microbes DKO-hu HSC Hofer et al.,72 2010
pDCs DKO-hu HSC Zhang et al.,73 2011
Interferon-α NSG-BLT Long et al.,74 2012
Interferon-α SCID-hu Thy/Liv Stoddart et al.,75 2010
Neuropathology NSG-hu HSC Dash et al.,76 2011
Gong et al.,77 2011
Gorantla et al.,78 2010
Antiviral drug siRNA DKO-hu HSC Neff et al.,79 2011
Zhou et al.,80 2011
Ter Brake et al.,81 2009
NSG-hu HSC Kumar et al.,82 2008
Kim et al.,83 2010
NSG-BLT Wheeler et al.,69 2011
Small molecules DKO-hu HSC Choudhary et al.,84 2009
Sango et al.,85 2010
SCID-hu Thy/Liv Stoddart et al.,46 2007
Stoddart et al.,47 2007
Peptides DKO-hu HSC van Duyne et al.,86 2008
SCID-hu Thy/Liv Stoddart et al.,45 2012
Gene Therapy shRNA NSG-BLT Shimizu et al.,87 2010
HIV-1 neutralizing antibody NSG-hu HSC Joseph et al.,88 2010

Abbreviations: BLT, human thymus and liver tissues and HSC; DKO, rag2−/− Il2rg tm1Sug/Jic; hu HSC, human CD34+ hematopoietic stem/progenitor cells; hu Thy/Liv, human thymus and liver tissues; NOG, NOD. Cg-Prkdc scid Il2rg tm1Sug/Jic; NSG, NOD/LtSZ-scid Il2rgnull; pDCs, plasmacytoid dendritic cells; Tregs, regulatory T cells; shRNA, small hairpin RNA; siRNA, small interfering RNA.

Humanized mice have been used to study various aspects of HIV-1 infection (Table 1): the roles of regulatory T cells (Tregs)54 and plasmacytoid dendritic cells (pDCs)73 in HIV-1 infection, the immunopathogenesis of HIV-1, viral evolution in vivo,58,59 new antiviral treatments,7981,84,86 gene therapy,83,88 mucosal transmission56 and microbicide development.68,70 In the presence of antiviral drugs, latent infection can be established, making it a valuable model to study HIV-1 latency.6163

Most importantly, the anti-HIV-1 immune responses were observed in the infected mice. These include anti-HIV-1 antibodies49,50,52,56 and HIV-1-specific T-cell responses.52 HIV-1 infection resulted in increased CD8+ T cells in the blood, which were derived from CD45RA effector/memory T cells, not CD45RA+ naive T cells.60 The depletion of CD8+ T cells by antibody treatment resulted in increased viral load, robust immune cell activation and cytopathology in lymphoid tissues.89 These improvements make the new generation of humanized mice superior to the early models for studying HIV-1 immune responses and immunopathogenesis.

IMMUNE ACTIVATION AND HIV-1 PATHOGENESIS

Although HIV-1 infection kills target cells, the majority of CD4+ T-cell loss is not due to productive infection.90,91 It is widely accepted that chronic, generalized immune activation induced by HIV-1 infection is the major driving force of immunodeficiency.9294 The level of T-cell activation (the percentage HLA-DR+CD38+ T cells out of all the CD8+ T cells) predicts disease progression independent of and more accurately than CD4+ T cell count.95 Additionally, it was recently reported that anti-malarial drugs such as chloroquine96 and hydroxy-chloroquine97 inhibit immune activation in HIV-1-infected patients when used as a monotherapy96 or in combination with antiviral treatment.97 The reduction in immune activation correlates with an increase in CD4+ T cells.97

Additional data supporting the hypothesis that immune activation drives AIDS development come from simian immunodeficiency virus (SIV)-infected monkeys. SIV-infected Asian monkeys (e.g., rhesus macaques, cynomolgus macaques and pigtail macaques) experience a dramatic increase in immune activation, rapid CD4+ T-cell loss and progression to AIDS. Conversely, infected natural African hosts (e.g., green monkeys, sooty mangabeys and mandrills) exhibit minimal T-cell activation and rarely progress to immunodeficiency despite a viral load comparable to pathogenic SIV infections.98100 Moreover, experimental induction of immune activation by lipopolysaccharide (LPS) in SIV-infected African green monkeys has been shown to result in CD4+ T-cell loss.101 Interestingly, the transcriptomes of patients with preserved CD4+ T cell numbers in the presence of constant, high HIV-1 viral loads are very similar to the transcriptomes of SIV-infected sooty mangabeys.102

Long-term immune activation can cause damage even in the absence of viral infection. For example, transgenic mice expressing CD70 develop chronic immune activation and lethal immunodeficiency.103 Moreover, treatment with Toll-like receptor (TLR) 9104 or TLR7105 ligands in mice induces immune activation, lymphoid organ distraction and immune suppression.

The exact mechanism by which HIV infection leads to immune activation is not fully understood. It has been proposed that HIV-1 viral proteins, whole viral particles, infected cells and infection-induced cytokines contribute to immune cell activation.93 Other factors have also been proposed as the cause of immune activation, such as loss of tissue integrity during acute phase infection of gut-associated lymphoid tissue (GALT) and microbial products translocation,106 loss of Tregs,107,108 activation of pDCs,112 and production of type I interferons (IFN-I).109,110

GALT infection and intestinal bacteria translocation

HIV-1 infection causes massive depletion of T cells in GALT and breaks down the mucosal barrier, resulting in translocation of intestinal bacterial products (including LPS) and immune activation.106 Injection of LPS into SIV-infected African green monkeys resulted in increased immune activation and viral replication.101 It was recently reported that circulating LPS in the first years of chronic HIV-1 infection is a strong predictor of disease progression independent of CD4+ T-cell counts and HIV-1 viral load, so plasma LPS may serve as a candidate biomarker for HIV-1 monitoring and evaluation of treatments.111

IFN-I and pDC activation

IFN-I is a group of multifunctional cytokines that plays an essential role in antiviral immunity. pDCs constitute 0.2%–0.5% of human PBMCs, but they are capable of producing 100 times more IFN-I than other cell types. They preferentially express TLR7 and TLR9, sensing viral RNA and DNA, respectively, during infection. Upon viral infection or other stimulation, pDCs produce large amounts of IFN-I and other inflammatory cytokines.112 IFN-I play important roles in immune cell development and normal immune responses. However, persistent expression of IFN-I induces immune dysfunction and may lead to autoimmune disease.113

Elevated expression of IFN-I has been documented in HIV-1-infected patients.114116 HIV-1 infection also stimulates IFN-I production in cultured human PBMCs or purified pDCs.117119 As would be expected, both IFN-I120,121 and pDCs122 show the capacity to inhibit HIV-1 replication in vitro. pDCs are numerically decreased123125 and functionally impaired in the peripheral blood of HIV-1-infected individuals. The decreased capacity of pDCs to produce IFN-I correlates with opportunistic infection independent of CD4+ T-cell counts.126128 These observations suggest that pDCs and IFN-I are protective during HIV-1 infection, which is similar to their role in other viral infections.

Paradoxically, the high levels of IFN-I in HIV-1-infected patients do not correlate with viral control; rather, they are predictive of HIV-1 disease progression and AIDS development.115,129,130 Additionally, IFN-I is induced during the acute phase of SIV infection in both pathogenic and non-pathogenic hosts, but is rapidly controlled during non-pathogenic SIV infection. Only pathogenic SIV infection is characterized by sustained IFN-I production during a chronic infection, which correlates with immune activation and AIDS development.131134 However, it is still not clear if pDCs are the major source of IFN-I during chronic HIV-1 infection because the IFN-I-producing cells in the spleens of HIV-1 infected patients do not seem to express pDC-specific markers.135 The mechanisms of IFN-I production and pDC activation in HIV-1 pathogenesis are poorly understood. HIV-1 infection can stimulate pDCs to express TNF-related apoptosis-inducing ligand, which may contribute to CD4+ T-cell depletion.136138 However, the induction of CD4+ T-cell death by TNF-related apoptosis-inducing ligand-expressing pDCs remains controversial.139 These conflicting reports highlight that IFN-I and pDCs may play mixed roles in HIV-1 infection and immunopathogenesis.

Tregs

Human CD4+CD25+FoxP3+ Tregs are central players in balancing the induction and suppression of immune activation.140,141 During HIV infection, Tregs could be either beneficial, by inhibiting immune activation, or detrimental, by suppressing virus-specific T-cell responses.107,142 It has been reported that, during HIV infection, the absolute Treg count decreases and that Treg loss correlates with immune activation and disease progression.143,144 However, other studies have shown that Treg numbers are elevated in both the PBMCs145,146 and the GALT147 of HIV-1-infected patients, independently of immunological and virological status.145,146 One study in SIV-infected rhesus macaques demonstrated that Tregs are depleted from the GALT but accumulate in PBMCs and lymphoid organs.148 These conflicting reports underscore the complex role of Tregs in HIV infection and immune activation.

STUDYING THE MECHANISMS OF HIV-1 PATHOGENESIS IN HUMANIZED MOUSE MODELS

HIV-1 infection in humanized mice results in sustained viral replication and significant CD4+ T-cell depletion in the peripheral blood and lymphoid organs.48,50,51,53,55,56 Viral antigens have been observed in T cells, CD68+ macrophages50,56 and pDCs.73 Importantly, HIV-1 infection results in T-cell activation in the humanized mice, and the immune activation correlates with viral load74 and T-cell depletion.73 Several experiments to delineate the mechanisms of HIV-1 immunopathogenesis have been carried out in humanized mice and will be summarized in this section.

GALT infection and gut bacteria translocation

NOD/SCID-BLT or NSG-BLT mice support human cell reconstitution in the gut and virginal tissues through mucosal inoculation.24,56 These mice have been used to study microbicides and the prevention of HIV-1 mucosal transmission.24,68,70,71 DKO-hu HSC mice show very limited levels of human cells in the gut mucosa65 and whether these mice can support mucosal infection remains controversial.64,65 Application of dextran sodium sulfate induces bacterial endotoxin translocation in DKO-hu HSC mice but does not result in elevated plasma LPS levels unless phagocytic cells are depleted with clodronate liposomes or impaired by HIV-1 infection.72 This finding highlights the role of macrophages in modulating microbial translocation and immune activation.

pDCs and IFN-I in HIV-1 pathogenesis

Human pDCs in these chimeric mice phenotypically resemble their counterparts from human PBMCs in their expression of specific surface markers such as blood dendritic cell antigen 2, CD123, HLA-DR and CD4.18,52,73 Moreover, they function similarly to human pDCs by producing IFN-I and other inflammatory cytokines upon influenza virus or herpes simplex virus infection.18,52,73 HIV-1 infection in humanized mice can also activate pDCs to produce IFN-I and other cytokines. Importantly, the activation of pDCs positively correlates with immune activation and CD4+ T-cell depletion in infected mice.73 It has also been shown that IFN-I application to NSG-BLT mice causes immune activation similar to that induced by HIV-1 infection.74

It was recently reported that chloroquine118 and rapamycin151,152 inhibit IFN-I production by pDCs in vitro. Meanwhile, clinical studies show that chloroquine,96 hydroxychloroquine97 and rapamycin149,150 could reduce immune activation and inhibit pathogenesis in HIV-1-infected patients. Whether these drugs function through inhibiting pDCs in vivo needs to be examined further. Humanized mice provide a robust in vivo model for these studies and other hypothesis-driven experiments that test the roles of pDCs and IFN-I in HIV-1 pathogenesis.

Roles of Tregs in HIV-1 infection and pathogenesis

Tregs were observed in different organs of humanized mice, and purified Tregs have suppressive functions that are similar to those of their human PBMC-derived equivalents.54,153 During the acute phase of infection, CD4+FoxP3+ Tregs are preferentially infected and depleted by a pathogenic HIV-1 isolate in infected DKO-hu HSC mice. When Tregs are depleted with an IL-2-toxin fusion protein (denileukin diftitox, trade name Ontak), HIV-1 replication is significantly impaired in infected mice. This is observed in the reduced number of infected cells in lymphoid organs and lower plasma viremia.54 Notably, Ontak does not efficiently deplete Tregs in monkeys,101 which highlights the advantage of humanized mouse models.

AIDS-related neurological disorders

Neurocognitive disorders are common causes of morbidity in HIV-1-infected patients.154 SIV-infected rhesus macaques have been developed to study HIV-1-related neurological disorders.155 However, species specificity and high costs preclude their widespread usage. Recently, it was reported that HIV-1 infection in humanized mice induces neuroinflammatory responses, including leukocyte infiltration, microglial activation, meningitis and encephalitis.78 Structural changes in mouse cortical gray matter were also observed, evidenced by the loss of micro-tubule-associated protein 2, synaptophysin and neurofilament antigens.76 These reports suggest that humanized mice would be a valuable system for modeling AIDS-related neurodegeneration.

FUTURE DIRECTIONS

Substantial advances have been made in developing mice with humanized immune systems since the first report more than 20 years ago,5 although the functions of the human cells in these chimeric mice are still in need of further improvements.156,157 These mice have been shown to be invaluable for several aspects of HIV-1 research, especially for studying immune responses and immunopathogenesis.54,72,73 All of the human immune cell types that have been implicated in HIV-induced immune pathogenesis can be studied in humanized mice. Additionally, humanized mice can be genetically modified to test different hypotheses about immune activation and the underlying mechanisms. More importantly, data collected from humanized mice are readily translatable to clinical studies because the same agents can be used. In summary, humanized mouse models will increase our understanding of how HIV infection leads to AIDS and accelerate the development of therapeutic strategies.

References

  • 1.United Nations Programme on HIV/AIDS. Global Facts & Figures [PDF on Internet] 2009 Available from: http://data.unaids.org/pub/factsheet/2009/20091124_fs_global_en.pdf.
  • 2.Ganick DJ, Sarnwick RD, Shahidi NT, Manning DD. Inability of intravenously injected monocellular suspensions of human bone marrow to establish in the nude mouse. Int Arch Allergy Appl Immunol. 1980;62:330–333. doi: 10.1159/000232530. [DOI] [PubMed] [Google Scholar]
  • 3.Bosma GC, Custer RP, Bosma MJ. A severe combined immunodeficiency mutation in the mouse. Nature. 1983;301:527–530. doi: 10.1038/301527a0. [DOI] [PubMed] [Google Scholar]
  • 4.Mosier DE, Gulizia RJ, Baird SM, Wilson DB. Transfer of a functional human immune system to mice with severe combined immunodeficiency. Nature. 1988;335:256–259. doi: 10.1038/335256a0. [DOI] [PubMed] [Google Scholar]
  • 5.McCune JM, Namikawa R, Kaneshima H, Shultz LD, Lieberman M, Weissman IL. The SCID-hu mouse: murine model for the analysis of human hematolymphoid differentiation and function. Science. 1988;241:1632–1639. doi: 10.1126/science.241.4873.1632. [DOI] [PubMed] [Google Scholar]
  • 6.Shultz LD, Schweitzer PA, Christianson SW, Gott B, Schweitzer IB, Tennent B, et al. Multiple defects in innate and adaptive immunologic function in NOD/LtSz-scid mice. J Immunol. 1995;154:180–191. [PubMed] [Google Scholar]
  • 7.Hesselton RM, Greiner DL, Mordes JP, Rajan TV, Sullivan JL, Shultz LD. High levels of human peripheral blood mononuclear cell engraftment and enhanced susceptibility to human immunodeficiency virus type 1 infection in NOD/LtSz-scid/scid mice. J Infect Dis. 1995;172:974–982. doi: 10.1093/infdis/172.4.974. [DOI] [PubMed] [Google Scholar]
  • 8.Pflumio F, Izac B, Katz A, Shultz LD, Vainchenker W, Coulombel L. Phenotype and function of human hematopoietic cells engrafting immune-deficient CB17-severe combined immunodeficiency mice and nonobese diabetic-severe combined immunodeficiency mice after transplantation of human cord blood mononuclear cells. Blood. 1996;88:3731–3740. [PubMed] [Google Scholar]
  • 9.Yoshino H, Ueda T, Kawahata M, Kobayashi K, Ebihara Y, Manabe A, et al. Natural killer cell depletion by anti-asialo GM1 antiserum treatment enhances human hematopoietic stem cell engraftment in NOD/Shi-scid mice. Bone Marrow Transplant. 2000;26:1211–1216. doi: 10.1038/sj.bmt.1702702. [DOI] [PubMed] [Google Scholar]
  • 10.Ito M, Kobayashi K, Nakahata T. NOD/Shi-scid IL2rγnull (NOG) mice more appropriate for humanized mouse models. Curr Top Microbiol Immunol. 2008;324:53–76. doi: 10.1007/978-3-540-75647-7_3. [DOI] [PubMed] [Google Scholar]
  • 11.Shultz LD, Ishikawa F, Greiner DL. Humanized mice in translational biomedical research. Nat Rev Immunol. 2007;7:118–130. doi: 10.1038/nri2017. [DOI] [PubMed] [Google Scholar]
  • 12.Chen Q, Khoury M, Chen J. Expression of human cytokines dramatically improves reconstitution of specific human-blood lineage cells in humanized mice. Proc Natl Acad Sci USA. 2009;106:21783–2178. doi: 10.1073/pnas.0912274106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Willinger T, Rongvaux A, Takizawa H, Yancopoulos GD, Valenzuela DM, Murphy AJ, et al. Human IL-3/GM-CSF knock-in mice support human alveolar macrophage development and human immune responses in the lung. Proc Natl Acad Sci USA. 2011;108:2390–2395. doi: 10.1073/pnas.1019682108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Rongvaux A, Willinger T, Takizawa H, Rathinam C, Auerbach W, Murphy AJ, et al. Human thrombopoietin knockin mice efficiently support human hematopoiesis in vivo. Proc Natl Acad Sci USA. 2011;108:2378–2383. doi: 10.1073/pnas.1019524108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Whitfield-Larry F, Young EF, Talmage G, Fudge E, Azam A, Patel S, et al. HLA-A2-matched peripheral blood mononuclear cells from type 1 diabetic patients, but not nondiabetic donors, transfer insulitis to NOD-scid/γcnull/HLA-A2 transgenic mice concurrent with the expansion of islet-specific CD8+ T cells. Diabetes. 2011;60:1726–1733. doi: 10.2337/db10-1287. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Strowig T, Rongvaux A, Rathinam C, Takizawa H, Borsotti C, Philbrick W, et al. Transgenic expression of human signal regulatory protein alpha in Rag2−/−γc−/− mice improves engraftment of human hematopoietic cells in humanized mice. Proc Natl Acad Sci USA. 2011;108:13218–13223. doi: 10.1073/pnas.1109769108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Ito M, Hiramatsu H, Kobayashi K, Suzue K, Kawahata M, Hioki K, et al. NOD/SCID/γcnull mouse: an excellent recipient mouse model for engraftment of human cells. Blood. 2002;100:3175–3182. doi: 10.1182/blood-2001-12-0207. [DOI] [PubMed] [Google Scholar]
  • 18.Traggiai E, Chicha L, Mazzucchelli L, Bronz L, Piffaretti JC, Lanzavecchia A, et al. Development of a human adaptive immune system in cord blood cell-transplanted mice. Science. 2004;304:104–107. doi: 10.1126/science.1093933. [DOI] [PubMed] [Google Scholar]
  • 19.Ishikawa F, Yasukawa M, Lyons B, Yoshida S, Miyamoto T, Yoshimoto G, et al. Development of functional human blood and immune systems in NOD/SCID/IL2 receptor γ chainnull mice. Blood. 2005;106:1565–1573. doi: 10.1182/blood-2005-02-0516. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Shultz LD, Lyons BL, Burzenski LM, Gott B, Chen X, Chaleff S, et al. Human lymphoid and myeloid cell development in NOD/LtSz-scid IL2R γnull mice engrafted with mobilized human hemopoietic stem cells. J Immunol. 2005;174:6477–6489. doi: 10.4049/jimmunol.174.10.6477. [DOI] [PubMed] [Google Scholar]
  • 21.Pearson T, Shultz LD, Miller D, King M, Laning J, Fodor W, et al. Non-obese diabetic-recombination activating gene-1 (NOD-Rag1null) interleukin (IL)-2 receptor common gamma chain (IL2r γnull) null mice: a radioresistant model for human lymphohaematopoietic engraftment. Clin Exp Immunol. 2008;154:270–284. doi: 10.1111/j.1365-2249.2008.03753.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Lepus CM, Gibson TF, Gerber SA, Kawikova I, Szczepanik M, Hossain J, et al. Comparison of human fetal liver, umbilical cord blood, and adult blood hematopoietic stem cell engraftment in NOD-scid/γc−/−, Balb/c-Rag1−/−γc−/−, and C. B-17-scid/bg immunodeficient mice. Hum Immunol. 2009;70:790–802. doi: 10.1016/j.humimm.2009.06.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.McDermott SP, Eppert K, Lechman ER, Doedens M, Dick JE. Comparison of human cord blood engraftment between immunocompromised mouse strains. Blood. 2010;116:193–200. doi: 10.1182/blood-2010-02-271841. [DOI] [PubMed] [Google Scholar]
  • 24.Stoddart CA, Maidji E, Galkina SA, Kosikova G, Rivera JM, Moreno ME, et al. Superior human leukocyte reconstitution and susceptibility to vaginal HIV transmission in humanized NOD-scid IL-2Rγ−/− (NSG) BLT mice. Virology. 2011;417:154–160. doi: 10.1016/j.virol.2011.05.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Matozaki T, Murata Y, Okazawa H, Ohnishi H. Functions and molecular mechanisms of the CD47-SIRPalpha signalling pathway. Trends Cell Biol. 2009;19:72–80. doi: 10.1016/j.tcb.2008.12.001. [DOI] [PubMed] [Google Scholar]
  • 26.Takenaka K, Prasolava TK, Wang JC, Mortin-Toth SM, Khalouei S, Gan OI, et al. Polymorphism in Sirpa modulates engraftment of human hematopoietic stem cells. Nat Immunol. 2007;8:1313–1323. doi: 10.1038/ni1527. [DOI] [PubMed] [Google Scholar]
  • 27.Brehm MA, Cuthbert A, Yang C, Miller DM, DiIorio P, Laning J, et al. Parameters for establishing humanized mouse models to study human immunity: analysis of human hematopoietic stem cell engraftment in three immunodeficient strains of mice bearing the IL2rγnull mutation. Clin Immunol. 2010;135:84–98. doi: 10.1016/j.clim.2009.12.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Legrand N, Huntington ND, Nagasawa M, Bakker AQ, Schotte R, Strick-Marchand H, et al. Functional CD47/signal regulatory protein alpha (SIRPα) interaction is required for optimal human T- and natural killer- (NK) cell homeostasis in vivo. Proc Natl Acad Sci USA. 2011;108:13224–13229. doi: 10.1073/pnas.1101398108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.van Lent AU, Dontje W, Nagasawa M, Siamari R, Bakker AQ, Pouw SM, et al. IL-7 enhances thymic human T cell development in “human immune system” Rag2−/−IL-2Rγ−/− mice without affecting peripheral T cell homeostasis. J Immunol. 2009;183:7645–7655. doi: 10.4049/jimmunol.0902019. [DOI] [PubMed] [Google Scholar]
  • 30.Huntington ND, Legrand N, Alves NL, Jaron B, Weijer K, Plet A, et al. IL-15 trans-presentation promotes human NK cell development and differentiation in vivo. J Exp Med. 2009;206:25–34. doi: 10.1084/jem.20082013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Billerbeck E, Barry WT, Mu K, Dorner M, Rice CM, Ploss A. Development of human CD4+FoxP3+ regulatory T cells in human stem cell factor-, granulocyte-macrophage colony-stimulating factor-, and interleukin-3-expressing NOD-SCID IL2Rγnull humanized mice. Blood. 2011;117:3076–3086. doi: 10.1182/blood-2010-08-301507. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Willinger T, Rongvaux A, Strowig T, Manz MG, Flavell RA. Improving human hemato-lymphoid-system mice by cytokine knock-in gene replacement. Trends Immunol. 2011;32:321–327. doi: 10.1016/j.it.2011.04.005. [DOI] [PubMed] [Google Scholar]
  • 33.Rathinam C, Poueymirou WT, Rojas J, Murphy AJ, Valenzuela DM, Yancopoulos GD, et al. Efficient differentiation and function of human macrophages in humanized CSF-1 mice. Blood. 2011;118:3119–3128. doi: 10.1182/blood-2010-12-326926. [DOI] [PubMed] [Google Scholar]
  • 34.Strowig T, Gurer C, Ploss A, Liu YF, Arrey F, Sashihara J, et al. Priming of protective T cell responses against virus-induced tumors in mice with human immune system components. J Exp Med. 2009;206:1423–1434. doi: 10.1084/jem.20081720. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Shultz LD, Saito Y, Najima Y, Tanaka S, Ochi T, Tomizawa M, et al. Generation of functional human T-cell subsets with HLA-restricted immune responses in HLA class I expressing NOD/SCID/IL2rγnull humanized mice. Proc Natl Acad Sci USA. 2010;107:13022–13027. doi: 10.1073/pnas.1000475107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Jaiswal S, Pearson T, Friberg H, Shultz LD, Greiner DL, Rothman AL, et al. Dengue virus infection and virus-specific HLA-A2 restricted immune responses in humanized NOD-scid IL2rγnull mice. PLoS ONE. 2009;4:e7251. doi: 10.1371/journal.pone.0007251. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Covassin L, Laning J, Abdi R, Langevin DL, Phillips NE, Shultz LD, et al. Human peripheral blood CD4 T cell-engrafted non-obese diabetic-scid IL2rγnull H2-Ab1 (tm1Gru) Tg (human leucocyte antigen D-related 4) mice: a mouse model of human allogeneic graft-versus-host disease. Clin Exp Immunol. 2011;166:269–280. doi: 10.1111/j.1365-2249.2011.04462.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Danner R, Chaudhari SN, Rosenberger J, Surls J, Richie TL, Brumeanu TD, et al. Expression of HLA class II molecules in humanized NOD.Rag1KO.IL2RgcKO mice is critical for development and function of human T and B cells. PLoS ONE. 2011;6:e19826. doi: 10.1371/journal.pone.0019826. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Melkus MW, Estes JD, Padgett-Thomas A, Gatlin J, Denton PW, Othieno FA, et al. Humanized mice mount specific adaptive and innate immune responses to EBV and TSST-1. Nat Med. 2006;12:1316–1322. doi: 10.1038/nm1431. [DOI] [PubMed] [Google Scholar]
  • 40.Lan P, Tonomura N, Shimizu A, Wang S, Yang YG. Reconstitution of a functional human immune system in immunodeficient mice through combined human fetal thymus/liver and CD34+ cell transplantation. Blood. 2006;108:487–492. doi: 10.1182/blood-2005-11-4388. [DOI] [PubMed] [Google Scholar]
  • 41.Gimeno R, Weijer K, Voordouw A, Uittenbogaart CH, Legrand N, Alves NL, et al. Monitoring the effect of gene silencing by RNA interference in human CD34+ cells injected into newborn RAG2−/− γc−/− mice: functional inactivation of p53 in developing T cells. Blood. 2004;104:3886–3893. doi: 10.1182/blood-2004-02-0656. [DOI] [PubMed] [Google Scholar]
  • 42.Notta F, Doulatov S, Dick JE. Engraftment of human hematopoietic stem cells is more efficient in female NOD/SCID/IL-2Rgc-null recipients. Blood. 2010;115:3704–3707. doi: 10.1182/blood-2009-10-249326. [DOI] [PubMed] [Google Scholar]
  • 43.Namikawa R, Kaneshima H, Lieberman M, Weissman IL, McCune JM. Infection of the SCID-hu mouse by HIV-1. Science. 1988;242:1684–1686. doi: 10.1126/science.3201256. [DOI] [PubMed] [Google Scholar]
  • 44.McCune JM, Namikawa R, Shih CC, Rabin L, Kaneshima H. Suppression of HIV infection in AZT-treated SCID-hu mice. Science. 1990;247:564–566. doi: 10.1126/science.2300816. [DOI] [PubMed] [Google Scholar]
  • 45.Stoddart CA, Nault G, Galkina SA, Bousquet-Gagnon N, Bridon D, Quraishi O. Preexposure prophylaxis with albumin-conjugated C34 peptide HIV-1 fusion inhibitor in SCID-hu Thy/Liv mice. Antimicrob Agents Chemother. doi: 10.1128/AAC.05015-11. e-pub ahead of print January 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Stoddart CA, Joshi P, Sloan B, Bare JC, Smith PC, Allaway GP, et al. Potent activity of the HIV-1 maturation inhibitor bevirimat in SCID-hu Thy/Liv mice. PLoS ONE. 2007;2:e1251. doi: 10.1371/journal.pone.0001251. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Stoddart CA, Bales CA, Bare JC, Chkhenkeli G, Galkina SA, Kinkade AN, et al. Validation of the SCID-hu Thy/Liv mouse model with four classes of licensed antiretrovirals. PLoS ONE. 2007;2:e655. doi: 10.1371/journal.pone.0000655. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Zhang L, Kovalev GI, Su L. HIV-1 infection and pathogenesis in a novel humanized mouse model. Blood. 2007;109:2978–2981. doi: 10.1182/blood-2006-07-033159. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Watanabe S, Terashima K, Ohta S, Horibata S, Yajima M, Shiozawa Y, et al. Hematopoietic stem cell-engrafted NOD/SCID/IL2Rγnull mice develop human lymphoid systems and induce long-lasting HIV-1 infection with specific humoral immune responses. Blood. 2007;109:212–218. doi: 10.1182/blood-2006-04-017681. [DOI] [PubMed] [Google Scholar]
  • 50.Baenziger S, Tussiwand R, Schlaepfer E, Mazzucchelli L, Heikenwalder M, Kurrer MO, et al. Disseminated and sustained HIV infection in CD34+ cord blood cell-transplanted Rag2−/−γc−/−mice. Proc Natl Acad Sci USA. 2006;103:15951–15956. doi: 10.1073/pnas.0604493103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Berges BK, Wheat WH, Palmer BE, Connick E, Akkina R. HIV-1 infection and CD4 T cell depletion in the humanized Rag2−/−γc−/− (RAG-hu) mouse model. Retrovirology. 2006;3:76. doi: 10.1186/1742-4690-3-76. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Brainard DM, Seung E, Frahm N, Cariappa A, Bailey CC, Hart WK, et al. Induction of robust cellular and humoral virus-specific adaptive immune responses in human immunodeficiency virus-infected humanized BLT mice. J Virol. 2009;83:7305–7321. doi: 10.1128/JVI.02207-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Gorantla S, Sneller H, Walters L, Sharp JG, Pirruccello SJ, West JT, et al. Human immunodeficiency virus type 1 pathobiology studied in humanized BALB/c-Rag2−/−γc−/− mice. J Virol. 2007;81:2700–2712. doi: 10.1128/JVI.02010-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Jiang Q, Zhang L, Wang R, Jeffrey J, Washburn ML, Brouwer D, et al. FoxP3+CD4+ regulatory T cells play an important role in acute HIV-1 infection in humanized Rag2−/−γC−/− mice in vivo. Blood. 2008;112:2858–2868. doi: 10.1182/blood-2008-03-145946. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.An DS, Poon B, Ho Tsong Fang R, Weijer K, Blom B, Spits H, et al. Use of a novel chimeric mouse model with a functionally active human immune system to study human immunodeficiency virus type 1 infection. Clin Vaccine Immunol. 2007;14:391–396. doi: 10.1128/CVI.00403-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Sun Z, Denton PW, Estes JD, Othieno FA, Wei BL, Wege AK, et al. Intrarectal transmission, systemic infection, and CD4+ T cell depletion in humanized mice infected with HIV-1. J Exp Med. 2007;204:705–714. doi: 10.1084/jem.20062411. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Nie C, Sato K, Misawa N, Kitayama H, Fujino H, Hiramatsu H, et al. Selective infection of CD4+ effector memory T lymphocytes leads to preferential depletion of memory T lymphocytes in R5 HIV-1-infected humanized NOD/SCID/IL-2Rγnull mice. Virology. 2009;394:64–72. doi: 10.1016/j.virol.2009.08.011. [DOI] [PubMed] [Google Scholar]
  • 58.Ince WL, Zhang L, Jiang Q, Arrildt K, Su L, Swanstrom R. Evolution of the HIV-1 env gene in the Rag2−/−γC−/− humanized mouse model. J Virol. 2010;84:2740–2752. doi: 10.1128/JVI.02180-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Sato K, Izumi T, Misawa N, Kobayashi T, Yamashita Y, Ohmichi M, et al. Remarkable lethal G-to-A mutations in vif-proficient HIV-1 provirus by individual APOBEC3 proteins in humanized mice. J Virol. 2010;84:9546–9556. doi: 10.1128/JVI.00823-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Sato K, Nie C, Misawa N, Tanaka Y, Ito M, Koyanagi Y. Dynamics of memory and naive CD8+ T lymphocytes in humanized NOD/SCID/IL-2Rγnull mice infected with CCR5-tropic HIV-1. Vaccine. 2010;28(Suppl 2):B32–B37. doi: 10.1016/j.vaccine.2009.10.154. [DOI] [PubMed] [Google Scholar]
  • 61.Choudhary SK, Archin NM, Cheema M, Dahl NP, Garcia JV, Margolis DM. Latent HIV-1 infection of resting CD4 T cells in the humanized Rag2−/−γc−/− mouse. J Virol. 2012;86:114–120. doi: 10.1128/JVI.05590-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Denton PW, Olesen R, Choudhary SK, Archin NM, Wahl A, Swanson MD, et al. Generation of HIV latency in humanized BLT mice. J Virol. 2012;86:630–634. doi: 10.1128/JVI.06120-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Marsden MD, Kovochich M, Suree N, Shimizu S, Mehta R, Cortado R, et al. HIV latency in the humanized BLT mouse. J Virol. 2012;86:339–347. doi: 10.1128/JVI.06366-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Berges BK, Akkina SR, Folkvord JM, Connick E, Akkina R. Mucosal transmission of R5 and X4 tropic HIV-1 via vaginal and rectal routes in humanized Rag2−/−γc−/− (RAG-hu) mice. Virology. 2008;373:342–351. doi: 10.1016/j.virol.2007.11.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Hofer U, Baenziger S, Heikenwalder M, Schlaepfer E, Gehre N, Regenass S, et al. RAG2−/−γc−/− mice transplanted with CD34+ cells from human cord blood show low levels of intestinal engraftment and are resistant to rectal transmission of human immunodeficiency virus. J Virol. 2008;82:12145–12153. doi: 10.1128/JVI.01105-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Neff CP, Ndolo T, Tandon A, Habu Y, Akkina R. Oral pre-exposure prophylaxis by anti-retrovirals raltegravir and maraviroc protects against HIV-1 vaginal transmission in a humanized mouse model. PLoS ONE. 2010;5:e15257. doi: 10.1371/journal.pone.0015257. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Akkina R, Berges BK, Palmer BE, Remling L, Neff CP, Kuruvilla J, et al. Humanized Rag1−/−γc−/− mice support multilineage hematopoiesis and are susceptible to HIV-1 infection via systemic and vaginal routes. PLoS ONE. 2011;6:e20169. doi: 10.1371/journal.pone.0020169. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Denton PW, Othieno F, Martinez-Torres F, Zou W, Krisko JF, Fleming E, et al. One percent tenofovir applied topically to humanized BLT mice and used according to the CAPRISA 004 experimental design demonstrates partial protection from vaginal HIV infection, validating the BLT model for evaluation of new microbicide candidates. J Virol. 2011;85:7582–7593. doi: 10.1128/JVI.00537-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Wheeler LA, Trifonova R, Vrbanac V, Basar E, McKernan S, Xu Z, et al. Inhibition of HIV transmission in human cervicovaginal explants and humanized mice using CD4 aptamer-siRNA chimeras. J Clin Invest. 2011;121:2401–2412. doi: 10.1172/JCI45876. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Denton PW, Estes JD, Sun Z, Othieno FA, Wei BL, Wege AK, et al. Antiretroviral pre-exposure prophylaxis prevents vaginal transmission of HIV-1 in humanized BLT mice. PLoS Med. 2008;5:e16. doi: 10.1371/journal.pmed.0050016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Denton PW, Krisko JF, Powell DA, Mathias M, Kwak YT, Martinez-Torres F, et al. Systemic administration of antiretrovirals prior to exposure prevents rectal and intravenous HIV-1 transmission in humanized BLT mice. PLoS ONE. 2010;5:e8829. doi: 10.1371/journal.pone.0008829. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Hofer U, Schlaepfer E, Baenziger S, Nischang M, Regenass S, Schwendener R, et al. Inadequate clearance of translocated bacterial products in HIV-infected humanized mice. PLoS Pathog. 2010;6:e1000867. doi: 10.1371/journal.ppat.1000867. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Zhang L, Jiang Q, Li G, Jeffrey J, Kovalev GI, Su L. Efficient infection, activation, and impairment of pDCs in the BM and peripheral lymphoid organs during early HIV-1 infection in humanized rag2/gamma C/mice in vivo. Blood. 2011;117:6184–6192. doi: 10.1182/blood-2011-01-331173. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Long BR, Stoddart CA. Interferon alpha and HIV infection cause activation of human T cells in NSG-BLT mice. J Virol. 2012;86:3327–3336. doi: 10.1128/JVI.06676-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Stoddart CA, Keir ME, McCune JM. IFN-alpha-induced upregulation of CCR5 leads to expanded HIV tropism in vivo. PLoS Pathog. 2010;6:e1000766. doi: 10.1371/journal.ppat.1000766. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Dash PK, Gorantla S, Gendelman HE, Knibbe J, Casale GP, Makarov E, et al. Loss of neuronal integrity during progressive HIV-1 infection of humanized mice. J Neurosci. 2011;31:3148–3157. doi: 10.1523/JNEUROSCI.5473-10.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Gong N, Liu J, Reynolds AD, Gorantla S, Mosley RL, Gendelman HE. Brain ingress of regulatory T cells in a murine model of HIV-1 encephalitis. J Neuroimmunol. 2011;230:33–41. doi: 10.1016/j.jneuroim.2010.08.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Gorantla S, Makarov E, Finke-Dwyer J, Castanedo A, Holguin A, Gebhart CL, et al. Links between progressive HIV-1 infection of humanized mice and viral neuropathogenesis. Am J Pathol. 2010;177:2938–2949. doi: 10.2353/ajpath.2010.100536. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Neff CP, Zhou J, Remling L, Kuruvilla J, Zhang J, Li H, et al. An aptamer-siRNA chimera suppresses HIV-1 viral loads and protects from helper CD4+ T cell decline in humanized mice. Sci Transl Med. 2011;3:66ra6. doi: 10.1126/scitranslmed.3001581. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Zhou J, Neff CP, Liu X, Zhang J, Li H, Smith DD, et al. Systemic administration of combinatorial dsiRNAs via nanoparticles efficiently suppresses HIV-1 infection in humanized mice. Mol Ther. 2011;19:2228–2238. doi: 10.1038/mt.2011.207. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Ter Brake O, Legrand N, von Eije KJ, Centlivre M, Spits H, Weijer K, et al. Evaluation of safety and efficacy of RNAi against HIV-1 in the human immune system (Rag-2−/−γc−/− mouse model. Gene Ther. 2009;16:148–153. doi: 10.1038/gt.2008.124. [DOI] [PubMed] [Google Scholar]
  • 82.Kumar P, Ban HS, Kim SS, Wu H, Pearson T, Greiner DL, et al. T cell-specific siRNA delivery suppresses HIV-1 infection in humanized mice. Cell. 2008;134:577–586. doi: 10.1016/j.cell.2008.06.034. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Kim SS, Peer D, Kumar P, Subramanya S, Wu H, Asthana D, et al. RNAi-mediated CCR5 silencing by LFA-1-targeted nanoparticles prevents HIV infection in BLT mice. Mol Ther. 2010;18:370–376. doi: 10.1038/mt.2009.271. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Choudhary SK, Rezk NL, Ince WL, Cheema M, Zhang L, Su L, et al. Suppression of human immunodeficiency virus type 1 (HIV-1) viremia with reverse transcriptase and integrase inhibitors, CD4+ T-cell recovery, and viral rebound upon interruption of therapy in a new model for HIV treatment in the humanized Rag2−/−γc−/− mouse. J Virol. 2009;83:8254–8258. doi: 10.1128/JVI.00580-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Sango K, Joseph A, Patel M, Osiecki K, Dutta M, Goldstein H. Highly active antiretroviral therapy potently suppresses HIV infection in humanized Rag2−/−γc−/− mice. AIDS Res Hum Retroviruses. 2010;26:735–746. doi: 10.1089/aid.2009.0136. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.van Duyne R, Cardenas J, Easley R, Wu W, Kehn-Hall K, Klase Z, et al. Effect of transcription peptide inhibitors on HIV-1 replication. Virology. 2008;376:308–322. doi: 10.1016/j.virol.2008.02.036. [DOI] [PubMed] [Google Scholar]
  • 87.Shimizu S, Hong P, Arumugam B, Pokomo L, Boyer J, Koizumi N, et al. A highly efficient short hairpin RNA potently down-regulates CCR5 expression in systemic lymphoid organs in the hu-BLT mouse model. Blood. 2010;115:1534–1544. doi: 10.1182/blood-2009-04-215855. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Joseph A, Zheng JH, Chen K, Dutta M, Chen C, Stiegler G, et al. Inhibition of in vivo HIV infection in humanized mice by gene therapy of human hematopoietic stem cells with a lentiviral vector encoding a broadly neutralizing anti-HIV antibody. J Virol. 2010;84:6645–6653. doi: 10.1128/JVI.02339-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Gorantla S, Makarov E, Finke-Dwyer J, Gebhart CL, Domm W, Dewhurst S, et al. CD8+ cell depletion accelerates HIV-1 immunopathology in humanized mice. J Immunol. 2010;184:7082–7091. doi: 10.4049/jimmunol.1000438. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Meyaard L, Otto SA, Jonker RR, Mijnster MJ, Keet RP, Miedema F. Programmed death of T cells in HIV-1 infection. Science. 1992;257:217–219. doi: 10.1126/science.1352911. [DOI] [PubMed] [Google Scholar]
  • 91.Finkel TH, Tudor-Williams G, Banda NK, Cotton MF, Curiel T, Monks C, et al. Apoptosis occurs predominantly in bystander cells and not in productively infected cells of HIV-and SIV-infected lymph nodes. Nat Med. 1995;1:129–134. doi: 10.1038/nm0295-129. [DOI] [PubMed] [Google Scholar]
  • 92.Ascher MS, Sheppard HW. AIDS as immune system activation: a model for pathogenesis. Clin Exp Immunol. 1988;73:165–167. [PMC free article] [PubMed] [Google Scholar]
  • 93.Sodora DL, Silvestri G. Immune activation and AIDS pathogenesis. AIDS. 2008;22:439–446. doi: 10.1097/QAD.0b013e3282f2dbe7. [DOI] [PubMed] [Google Scholar]
  • 94.Moir S, Buckner CM, Ho J, Wang W, Chen J, Waldner AJ, et al. B cells in early and chronic HIV infection: evidence for preservation of immune function associated with early initiation of antiretroviral therapy. Blood. 2010;116:5571–5579. doi: 10.1182/blood-2010-05-285528. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Giorgi JV, Liu Z, Hultin LE, Cumberland WG, Hennessey K, Detels R The Los Angeles Center, Multicenter AIDS Cohort Study. Elevated levels of CD38+CD8+ T cells in HIV infection add to the prognostic value of low CD4+ T cell levels: results of 6 years of follow-up. J Acquir Immune Defic Syndr. 1993;6:904–912. [PubMed] [Google Scholar]
  • 96.Murray SM, Down CM, Boulware DR, Stauffer WM, Cavert WP, Schacker TW, et al. Reduction of immune activation with chloroquine therapy during chronic HIV infection. J Virol. 2010;84:12082–12086. doi: 10.1128/JVI.01466-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Piconi S, Parisotto S, Rizzardini G, Passerini S, Terzi R, Argenteri B, et al. Hydroxychloroquine drastically reduces immune activation in HIV-infected, antiretroviral therapy-treated immunologic nonresponders. Blood. 2011;118:3263–3272. doi: 10.1182/blood-2011-01-329060. [DOI] [PubMed] [Google Scholar]
  • 98.Sodora DL, Allan JS, Apetrei C, Brenchley JM, Douek DC, Else JG, et al. Toward an AIDS vaccine: lessons from natural simian immunodeficiency virus infections of African nonhuman primate hosts. Nat Med. 2009;15:861–865. doi: 10.1038/nm.2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Apetrei C, Sumpter B, Souquiere S, Chahroudi A, Makuwa M, Reed P, et al. Immunovirological analyses of chronically SIVmnd-1- and SIVmnd-2-infected mandrills (Mandrillus sphinx) J Virol. 2011;85:13077–13087. doi: 10.1128/JVI.05693-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Klatt NR, Canary LA, Vanderford TH, Vinton CL, Engram JC, Dunham RM, et al. Dynamics of simian immunodeficiency virus SIVmac239 infection in pigtail macaques. J Virol. 2012;86:1203–1213. doi: 10.1128/JVI.06033-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Pandrea I, Gaufin T, Brenchley JM, Gautam R, Monjure C, Gautam A, et al. Cutting edge: experimentally induced immune activation in natural hosts of simian immunodeficiency virus induces significant increases in viral replication and CD4+ T cell depletion. J Immunol. 2008;181:6687–6691. doi: 10.4049/jimmunol.181.10.6687. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Rotger M, Dalmau J, Rauch A, McLaren P, Bosinger SE, Martinez R, et al. Comparative transcriptomics of extreme phenotypes of human HIV-1 infection and SIV infection in sooty mangabey and rhesus macaque. J Clin Invest. 2011;121:2391–2400. doi: 10.1172/JCI45235. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Tesselaar K, Arens R, van Schijndel GM, Baars PA, van der Valk MA, Borst J, et al. Lethal T cell immunodeficiency induced by chronic costimulation via CD27–CD70 interactions. Nat Immunol. 2003;4:49–54. doi: 10.1038/ni869. [DOI] [PubMed] [Google Scholar]
  • 104.Heikenwalder M, Polymenidou M, Junt T, Sigurdson C, Wagner H, Akira S, et al. Lymphoid follicle destruction and immunosuppression after repeated CpG oligodeoxynucleotide administration. Nat Med. 2004;10:187–192. doi: 10.1038/nm987. [DOI] [PubMed] [Google Scholar]
  • 105.Baenziger S, Heikenwalder M, Johansen P, Schlaepfer E, Hofer U, Miller RC, et al. Triggering TLR7 in mice induces immune activation and lymphoid system disruption, resembling HIV-mediated pathology. Blood. 2009;113:377–388. doi: 10.1182/blood-2008-04-151712. [DOI] [PubMed] [Google Scholar]
  • 106.Brenchley JM, Price DA, Schacker TW, Asher TE, Silvestri G, Rao S, et al. Microbial translocation is a cause of systemic immune activation in chronic HIV infection. Nat Med. 2006;12:1365–1371. doi: 10.1038/nm1511. [DOI] [PubMed] [Google Scholar]
  • 107.Holmes D, Jiang Q, Zhang L, Su L. Foxp3 and Treg cells in HIV-1 infection and immunopathogenesis. Immunol Res. 2008;41:248–266. doi: 10.1007/s12026-008-8037-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Fazekas de St Groth B, Landay AL. Regulatory T cells in HIV infection: pathogenic or protective participants in the immune response? AIDS. 2008;22:671–683. doi: 10.1097/QAD.0b013e3282f466da. [DOI] [PubMed] [Google Scholar]
  • 109.Skurkovich S, Skurkovich B, Bellanti JA. A disturbance of interferon synthesis with the hyperproduction of unusual kinds of interferon can trigger autoimmune disease and play a pathogenetic role in AIDS: the removal of these interferons can be therapeutic. Med Hypotheses. 1993;41:177–185. doi: 10.1016/0306-9877(93)90066-y. [DOI] [PubMed] [Google Scholar]
  • 110.Fitzgerald-Bocarsly P, Jacobs ES. Plasmacytoid dendritic cells in HIV infection: striking a delicate balance. J Leukoc Biol. 2010;87:609–620. doi: 10.1189/jlb.0909635. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Marchetti G, Cozzi-Lepri A, Merlini E, Bellistri GM, Castagna A, Galli M, et al. Microbial translocation predicts disease progression of HIV-infected antiretroviral-naive patients with high CD4+ cell count. AIDS. 2011;25:1385–1394. doi: 10.1097/QAD.0b013e3283471d10. [DOI] [PubMed] [Google Scholar]
  • 112.Liu YJ. IPC: professional type 1 interferon-producing cells and plasmacytoid dendritic cell precursors. Annu Rev Immunol. 2005;23:275–306. doi: 10.1146/annurev.immunol.23.021704.115633. [DOI] [PubMed] [Google Scholar]
  • 113.Theofilopoulos AN, Baccala R, Beutler B, Kono DH. Type I interferons (alpha/beta) in immunity and autoimmunity. Annu Rev Immunol. 2005;23:307–336. doi: 10.1146/annurev.immunol.23.021704.115843. [DOI] [PubMed] [Google Scholar]
  • 114.DeStefano E, Friedman RM, Friedman-Kien AE, Goedert JJ, Henriksen D, Preble OT, et al. Acid-labile human leukocyte interferon in homosexual men with Kaposi’s sarcoma and lymphadenopathy. J Infect Dis. 1982;146:451–459. doi: 10.1093/infdis/146.4.451. [DOI] [PubMed] [Google Scholar]
  • 115.Buimovici-Klein E, Lange M, Klein RJ, Grieco MH, Cooper LZ. Long-term follow-up of serum-interferon and its acid-stability in a group of homosexual men. AIDS Res. 1986;2:99–108. doi: 10.1089/aid.1.1986.2.99. [DOI] [PubMed] [Google Scholar]
  • 116.Badolato R, Ghidini C, Facchetti F, Serana F, Sottini A, Chiarini M, et al. Type I interferon-dependent gene MxA in perinatal HIV-infected patients under antiretroviral therapy as marker for therapy failure and blood plasmacytoid dendritic cells depletion. J Transl Med. 2008;6:49. doi: 10.1186/1479-5876-6-49. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Capobianchi MR, de Marco F, Di Marco P, Dianzani F. Acid-labile human interferon alpha production by peripheral blood mononuclear cells stimulated by HIV-infected cells. Arch Virol. 1988;99:9–19. doi: 10.1007/BF01311019. [DOI] [PubMed] [Google Scholar]
  • 118.Beignon AS, McKenna K, Skoberne M, Manches O, DaSilva I, Kavanagh DG, et al. Endocytosis of HIV-1 activates plasmacytoid dendritic cells via Toll-like receptor-viral RNA interactions. J Clin Invest. 2005;115:3265–3275. doi: 10.1172/JCI26032. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Martinelli E, Cicala C, van Ryk D, Goode DJ, Macleod K, Arthos J, et al. HIV-1 gp120 inhibits TLR9-mediated activation and IFN-α secretion in plasmacytoid dendritic cells. Proc Natl Acad Sci USA. 2007;104:3396–3401. doi: 10.1073/pnas.0611353104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Poli G, Orenstein JM, Kinter A, Folks TM, Fauci AS. Interferon-alpha but not AZT suppresses HIV expression in chronically infected cell lines. Science. 1989;244:575–577. doi: 10.1126/science.2470148. [DOI] [PubMed] [Google Scholar]
  • 121.Baca-Regen L, Heinzinger N, Stevenson M, Gendelman HE. Alpha interferon-induced antiretroviral activities: restriction of viral nucleic acid synthesis and progeny virion production in human immunodeficiency virus type 1-infected monocytes. J Virol. 1994;68:7559–7565. doi: 10.1128/jvi.68.11.7559-7565.1994. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Gurney KB, Colantonio AD, Blom B, Spits H, Uittenbogaart CH. Endogenous IFN-alpha production by plasmacytoid dendritic cells exerts an antiviral effect on thymic HIV-1 infection. J Immunol. 2004;173:7269–7276. doi: 10.4049/jimmunol.173.12.7269. [DOI] [PubMed] [Google Scholar]
  • 123.Donaghy H, Pozniak A, Gazzard B, Qazi N, Gilmour J, Gotch F, et al. Loss of blood CD11c+ myeloid and CD11c− plasmacytoid dendritic cells in patients with HIV-1 infection correlates with HIV-1 RNA virus load. Blood. 2001;98:2574–2576. doi: 10.1182/blood.v98.8.2574. [DOI] [PubMed] [Google Scholar]
  • 124.Pacanowski J, Kahi S, Baillet M, Lebon P, Deveau C, Goujard C, et al. Reduced blood CD123+ (lymphoid) and CD11c+ (myeloid) dendritic cell numbers in primary HIV-1 infection. Blood. 2001;98:3016–3021. doi: 10.1182/blood.v98.10.3016. [DOI] [PubMed] [Google Scholar]
  • 125.Soumelis V, Scott I, Gheyas F, Bouhour D, Cozon G, Cotte L, et al. Depletion of circulating natural type 1 interferon-producing cells in HIV-infected AIDS patients. Blood. 2001;98:906–912. doi: 10.1182/blood.v98.4.906. [DOI] [PubMed] [Google Scholar]
  • 126.Siegal FP, Lopez C, Fitzgerald PA, Shah K, Baron P, Leiderman IZ, et al. Opportunistic infections in acquired immune deficiency syndrome result from synergistic defects of both the natural and adaptive components of cellular immunity. J Clin Invest. 1986;78:115–123. doi: 10.1172/JCI112539. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Feldman S, Stein D, Amrute S, Denny T, Garcia Z, Kloser P, et al. Decreased interferon-alpha production in HIV-infected patients correlates with numerical and functional deficiencies in circulating type 2 dendritic cell precursors. Clin Immunol. 2001;101:201–210. doi: 10.1006/clim.2001.5111. [DOI] [PubMed] [Google Scholar]
  • 128.Lichtner M, Rossi R, Rizza MC, Mengoni F, Sauzullo I, Massetti AP, et al. Plasmacytoid dendritic cells count in antiretroviral-treated patients is predictive of HIV load control independent of CD4+ T-cell count. Curr HIV Res. 2008;6:19–27. doi: 10.2174/157016208783571937. [DOI] [PubMed] [Google Scholar]
  • 129.Buimovici-Klein E, Lange M, Klein RJ, Cooper LZ, Grieco MH. Is presence of interferon predictive for AIDS? Lancet. 1983;2:344. doi: 10.1016/s0140-6736(83)90322-7. [DOI] [PubMed] [Google Scholar]
  • 130.Meier A, Chang JJ, Chan ES, Pollard RB, Sidhu HK, Kulkarni S, et al. Sex differences in the Toll-like receptor-mediated response of plasmacytoid dendritic cells to HIV-1. Nat Med. 2009;15:955–959. doi: 10.1038/nm.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Bosinger SE, Li Q, Gordon SN, Klatt NR, Duan L, Xu L, et al. Global genomic analysis reveals rapid control of a robust innate response in SIV-infected sooty mangabeys. J Clin Invest. 2009;119:3556–3572. doi: 10.1172/JCI40115. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Jacquelin B, Mayau V, Targat B, Liovat AS, Kunkel D, Petitjean G, et al. Nonpathogenic SIV infection of African green monkeys induces a strong but rapidly controlled type I IFN response. J Clin Invest. 2009;119:3544–3555. doi: 10.1172/JCI40093. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Harris LD, Tabb B, Sodora DL, Paiardini M, Klatt NR, Douek DC, et al. Downregulation of robust acute type I interferon responses distinguishes nonpathogenic simian immunodeficiency virus (SIV) infection of natural hosts from pathogenic SIV infection of rhesus macaques. J Virol. 2010;84:7886–7891. doi: 10.1128/JVI.02612-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Campillo-Gimenez L, Laforge M, Fay M, Brussel A, Cumont MC, Monceaux V, et al. Nonpathogenesis of simian immunodeficiency virus infection is associated with reduced inflammation and recruitment of plasmacytoid dendritic cells to lymph nodes, not to lack of an interferon type I response, during the acute phase. J Virol. 2010;84:1838–1846. doi: 10.1128/JVI.01496-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Nascimbeni M, Perie L, Chorro L, Diocou S, Kreitmann L, Louis S, et al. Plasmacytoid dendritic cells accumulate in spleens from chronically HIV-infected patients but barely participate in interferon-alpha expression. Blood. 2009;113:6112–6119. doi: 10.1182/blood-2008-07-170803. [DOI] [PubMed] [Google Scholar]
  • 136.Herbeuval JP, Hardy AW, Boasso A, Anderson SA, Dolan MJ, Dy M, et al. Regulation of TNF-related apoptosis-inducing ligand on primary CD4+ T cells by HIV-1: role of type I IFN-producing plasmacytoid dendritic cells. Proc Natl Acad Sci USA. 2005;102:13974–13979. doi: 10.1073/pnas.0505251102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Herbeuval JP, Nilsson J, Boasso A, Hardy AW, Kruhlak MJ, Anderson SA, et al. Differential expression of IFN-alpha and TRAIL/DR5 in lymphoid tissue of progressor versus nonprogressor HIV-1-infected patients. Proc Natl Acad Sci USA. 2006;103:7000–7005. doi: 10.1073/pnas.0600363103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Stary G, Klein I, Kohlhofer S, Koszik F, Scherzer T, Mullauer L, et al. Plasmacytoid dendritic cells express TRAIL and induce CD4+ T-cell apoptosis in HIV-1 viremic patients. Blood. 2009;114:3854–3863. doi: 10.1182/blood-2009-04-217927. [DOI] [PubMed] [Google Scholar]
  • 139.Chehimi J, Papasavvas E, Tomescu C, Gekonge B, Abdulhaqq S, Raymond A, et al. Inability of plasmacytoid dendritic cells to directly lyse HIV-infected autologous CD4+ T cells despite induction of tumor necrosis factor-related apoptosis-inducing ligand. J Virol. 2010;84:2762–2773. doi: 10.1128/JVI.01350-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Rouse BT, Sarangi PP, Suvas S. Regulatory T cells in virus infections. Immunol Rev. 2006;212:272–286. doi: 10.1111/j.0105-2896.2006.00412.x. [DOI] [PubMed] [Google Scholar]
  • 141.Belkaid Y, Rouse BT. Natural regulatory T cells in infectious disease. Nat Immunol. 2005;6:353–360. doi: 10.1038/ni1181. [DOI] [PubMed] [Google Scholar]
  • 142.Seddiki N, Kelleher AD. Regulatory T cells in HIV infection: who’s suppressing what? Curr HIV/AIDS Rep. 2008;5:20–26. doi: 10.1007/s11904-008-0004-6. [DOI] [PubMed] [Google Scholar]
  • 143.Eggena MP, Barugahare B, Jones N, Okello M, Mutalya S, Kityo C, et al. Depletion of regulatory T cells in HIV infection is associated with immune activation. J Immunol. 2005;174:4407–4414. doi: 10.4049/jimmunol.174.7.4407. [DOI] [PubMed] [Google Scholar]
  • 144.Chase AJ, Yang HC, Zhang H, Blankson JN, Siliciano RF. Preservation of FoxP3+ regulatory T cells in the peripheral blood of human immunodeficiency virus type 1-infected elite suppressors correlates with low CD4+ T-cell activation. J Virol. 2008;82:8307–8315. doi: 10.1128/JVI.00520-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Kolte L, Gaardbo JC, Skogstrand K, Ryder LP, Ersboll AK, Nielsen SD. Increased levels of regulatory T cells (Tregs) in human immunodeficiency virus-infected patients after 5 years of highly active anti-retroviral therapy may be due to increased thymic production of naive Tregs. Clin Exp Immunol. 2009;155:44–52. doi: 10.1111/j.1365-2249.2008.03803.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Gaardbo JC, Nielsen SD, Vedel SJ, Ersboll AK, Harritshoj L, Ryder LP, et al. Regulatory T cells in human immunodeficiency virus-infected patients are elevated and independent of immunological and virological status, as well as initiation of highly active anti-retroviral therapy. Clin Exp Immunol. 2008;154:80–86. doi: 10.1111/j.1365-2249.2008.03725.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Shaw JM, Hunt PW, Critchfield JW, McConnell DH, Garcia JC, Pollard RB, et al. Increased frequency of regulatory T cells accompanies increased immune activation in rectal mucosae of HIV-positive noncontrollers. J Virol. 2011;85:11422–11434. doi: 10.1128/JVI.05608-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Chase AJ, Sedaghat AR, German JR, Gama L, Zink MC, Clements JE, et al. Severe depletion of CD4+CD25+ regulatory T cells from the intestinal lamina propria but not peripheral blood or lymph nodes during acute simian immunodeficiency virus infection. J Virol. 2007;81:12748–12757. doi: 10.1128/JVI.00841-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Moreno A, Barcena R, Quereda C, Casado JL, Perez-Elias MJ, Fortun J, et al. Safe use of raltegravir and sirolimus in an HIV-infected patient with renal impairment after orthotopic liver transplantation. AIDS. 2008;22:547–548. doi: 10.1097/QAD.0b013e3282f37478. [DOI] [PubMed] [Google Scholar]
  • 150.Di Benedetto F, Di Sandro S, de Ruvo N, Montalti R, Ballarin R, Guerrini GP, et al. First report on a series of HIV patients undergoing rapamycin monotherapy after liver transplantation. Transplantation. 2010;89:733–738. doi: 10.1097/TP.0b013e3181c7dcc0. [DOI] [PubMed] [Google Scholar]
  • 151.Guiducci C, Ghirelli C, Marloie-Provost MA, Matray T, Coffman RL, Liu YJ, et al. PI3K is critical for the nuclear translocation of IRF-7 and type I IFN production by human plasmacytoid predendritic cells in response to TLR activation. J Exp Med. 2008;205:315–322. doi: 10.1084/jem.20070763. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Cao W, Manicassamy S, Tang H, Kasturi SP, Pirani A, Murthy N, et al. Toll-like receptor-mediated induction of type I interferon in plasmacytoid dendritic cells requires the rapamycin-sensitive PI3K–mTOR–p70S6K pathway. Nat Immunol. 2008;9:1157–1164. doi: 10.1038/ni.1645. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Onoe T, Kalscheuer H, Danzl N, Chittenden M, Zhao G, Yang YG, et al. Human natural regulatory T cell development, suppressive function, and postthymic maturation in a humanized mouse model. J Immunol. 2011;187:3895–3903. doi: 10.4049/jimmunol.1100394. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Antinori A, Arendt G, Becker JT, Brew BJ, Byrd DA, Cherner M, et al. Updated research nosology for HIV-associated neurocognitive disorders. Neurology. 2007;69:1789–1799. doi: 10.1212/01.WNL.0000287431.88658.8b. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Williams R, Bokhari S, Silverstein P, Pinson D, Kumar A, Buch S. Nonhuman primate models of NeuroAIDS. J Neurovirol. 2008;14:292–300. doi: 10.1080/13550280802074539. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Watanabe Y, Takahashi T, Okajima A, Shiokawa M, Ishii N, Katano I, et al. The analysis of the functions of human B and T cells in humanized NOD/shi-scid/γcnull (NOG) mice (hu-HSC NOG mice) Int Immunol. 2009;21:843–858. doi: 10.1093/intimm/dxp050. [DOI] [PubMed] [Google Scholar]
  • 157.Andre MC, Erbacher A, Gille C, Schmauke V, Goecke B, Hohberger A, et al. Long-term human CD34+ stem cell-engrafted nonobese diabetic/SCID/IL-2R γnull mice show impaired CD8+ T cell maintenance and a functional arrest of immature NK cells. J Immunol. 2010;185:2710–2720. doi: 10.4049/jimmunol.1000583. [DOI] [PubMed] [Google Scholar]

RESOURCES