Abstract
Emigration of mature naïve CD4 SP T cells from the human thymus to the periphery is not fully understood, although elucidation of the mechanisms that govern egress of T cells is crucial to understanding both basic immunology and the immune response in diseases such as HIV infection. Recent work has brought to light the requirement for sphingosine-1-phosphate (S1P) and its receptors in a variety of fields including mature naïve T-cell egress from the thymus of mice. We are examining the expression and function of this novel requisite T-cell egress receptor within the human thymus, characterizing changes observed in the expression and function of this receptor in infectious diseases. To perform this work, we use a variety of humanized murine models reviewed in this article. Future work in the field of T-cell egress, especially as it pertains to S1P receptors, should advance the fields of basic T-cell immunology and immunopathology and open new avenues for exploration into novel therapeutics.
Keywords: human immunodeficiency virus, human immunology, humanized mice, migration, murine models, S1P-R1, S1P-R2, sphingosine-1-phosphate, T-cell reconstitution, thymic egress, thymocyte development, thymus
I. HISTORY AND BASICS OF T-CELL IMMUNOLOGY
The field of immunology is relatively young compared to other biological sciences. Edward Jenner, an English physician-scientist and discoverer of the concept of vaccination, which continues to be used today, is credited with stimulating the origination of the field and is often referred to as one of the “fathers of immunology,” along with Paul Ehrlich and Louis Pasteur. Although the theory of vaccination had circulated for some time before his work, Jenner cemented the validity of the notion when he intrepidly attempted to protect individuals from deadly smallpox infection by using a closely related nonfatal infectious agent, cowpox, that he administered to healthy individuals in 1796. Jenner documented that treatment with cowpox resulted in immunity to smallpox. Although he was successful, it would be some time before the underlying mechanisms of his discovery were fully understood.1 This pattern of an observation followed by toils to uncover a mechanism would set the tone for the field, in which researchers continue to strive to understand the processes governing the human immune system and the mechanisms of our incredibly intricate and complex response to pathogens that threaten to perturb the delicate balance of a healthy state.
Our current understanding is that our immune system is comprised of two branches known as innate and adaptive immunity. Innate immunity refers to immune system cells that are always ready to respond to a wide range of pathogens. This response is nonspecific and involves cells such as granulocytes, NK cells, and macrophages (phagocytic cells capable of engulfing many potentially harmful microorganisms).2 Elie Metchnikoff, a Russian immunologist, is responsible for primarily characterizing these types of reactions.1,3 Adaptive immunity refers to a more specific response that the immune system generates over time. Immunologic memory is developed upon encounter with a particular antigen (harmful microorganism or other substance), such that if the antigen presents again, specific cells are primed to deal with it. Adaptive immunity is further divided into two branches: humoral and cellular.
B lymphocytes (B cells, from “bursa” of the chicken, where they were discovered), responsible for humoral immunity, generate antibodies that recognize antigens and can neutralize recurring pathogens. T lymphocytes or T cells (from thymus, where these cells complete their development) are key players in adaptive immunity, specifically cell-mediated immunity. Briefly, T cells are comprised of several subsets, each of which is responsible for a specific task in the immune response. These tasks include cytotoxic activity (direct cell killing by Cluster of Differentiation 8+ [CD8+] T cells or cytotoxic T lymphocytes [CTLs]), regulation of other immune cells (CD4 and CD8 regulatory T cells [CD4/CD8 Tregs]), helper activity for CD4 T lymphocytes (Th1, Th2, and T follicular helper [Tfh] cells), and protective immunity against extracellular bacteria and fungi (Th1, Th2, Th17, Th22, Tfh, and γd T cells). Development of these various subsets begins in the thymus, but differentiation into the specific subsets described above occurs in the periphery after egress from the thymus.1 T-cell development in the thymus is discussed below.
II. T-CELL DEVELOPMENT IN THE THYMUS AND T-CELL SELECTION
As discussed in the preceding section, the adaptive branch of the human immune system is quite specific and potent. Therefore, the participants in adaptive immunity must be trained to avoid self-reactivity and to identify and target foreign invaders with an adequately strong response. T and B lymphocytes undergo intense selection processes during development. In this section, we focus on T cells, although it bears mentioning that B cells undergo an analogous process of selection and maturation within the bone marrow4,5 and the germinal centers of the secondary lymphoid organs (spleen and lymph nodes).6–8
T cells ultimately develop from CD34+ hematopoietic stem-cell progenitors that originate in bone marrow.9 In fact, both T- and B-cell precursors originate within bone marrow, but whereas B cells continue their development therein, T-cell precursors emigrate to the thymus, a lymphoid organ located in the thoracic cavity over the heart, where they progress through several stages of development.1 Upon entry of these precursors into the thymus, they have not yet undergone rearrangement of the genes encoding the T cell receptor (TCR) (a heterodimer consisting of an α and β chain embedded in a CD3 complex that allows recognition of the major histocompatibility complex [MHC]-bound antigen presented to T cells, the expression of which is a defining characteristic of T cells),10 a process that occurs during their maturation in the thymus. Additionally, at this stage they lack most of the surface molecules used to characterize mature T cells. Various surface-expressed receptors and other molecules are used to characterize the maturation stages of T cells, because in varied combinations they are quite specific to stages of T-cell development in the thymus. T-cell precursors in the thymus undergo extensive proliferation, but the majority of these cells die and do not progress along the differentiation pathway to become mature T cells.11
Within the thymic cortex, or the outer portion of its lobes, initial differentiation toward the T-cell pathway occurs, encouraged by the interaction between T-cell precursors and thymic stromal cells. The TCR gene begins to rearrange, and CD4, a coreceptor for the TCR, is expressed on thymocytes. T cells are considered double-positive cells once CD8 is also up-regulated. The first selection process, termed positive selection, selects for CD4+/CD8+ thymocytes that recognize the MHC displayed via specialized antigen-presenting cells in the thymus.12,13 Next, thymocytes up-regulate CCR7, which is necessary for their translocation to the thymic medulla,14,15 guided by additional cytokines produced by stromal cells.16 Once in the medulla, the thymocytes undergo a second selection process, negative selection, whereby double-positive cells that react too strongly to self-MHC fail to receive survival signals17,18 and undergo programmed cell death, or apoptosis,19 whereas cells that react mildly are selected for survival.18 The selection process is not yet fully understood, but recent evidence suggests that in developing thymocytes, microRNAs have a role in selection.20 Upon loss of expression of CD4 or CD8, thymocytes are considered single-positive (SP) cells.
Typical developmental markers for thymocytes include CD27 (tumor necrosis factor [TNF] receptor), CD69 (C-type lectin transmembrane protein), CD45RA (leukocyte common antigen), and CD62 ligand (CD62L, also known as L selectin). CD27 is also expressed on all thymic medullary cells and is used as a marker of relatively mature human thymocytes.21,22 CD45 is found in two isoforms, CD45RA and CD45RO, that are expressed differentially on various developing thymocyte subsets. CD45RA is expressed during the most immature stage of thymocyte development, not expressed in intermediate stages, and finally reexpressed in mature thymocytes. CD45RO is expressed on the majority of CD4+CD8+ (double-positive), intermediate-stage thymocytes.23 CD69, an activation marker that begins to be expressed during positive selection2 and is also expressed on activated peripheral T cells, has recently been found to have a role in regulation of T-cell egress.2 CD69 is down-regulated on mature thymocytes before egress and is not present on recent thymic emigrants.24 Finally, thymocytes up-regulate CD62L and are able to egress the thymus for the periphery to encounter their antigens as mature SP T cells. Egress from the thymus is discussed in the following section. See Figure 1 for the markers used to identify maturing thymocyte populations.
FIG. 1.
Expression of cell-surface and intracellular markers on thymocytes across developmental stages. Yellow, Thymic cortex; brown, medulla.
During thymocyte maturation, transient expression of KLF2, a master transcriptional regulator, has been shown in the mouse to control the expression of several migration-associated surface molecules expressed on thymocytes, including CD62L and CCR5.26–28 KLF2 begins to be expressed on mature thymocytes nearly prepared to egress the thymus for the periphery and has been demonstrated to regulate transcription of sphingosine-1-phosphate receptor 1 mRNA (S1P-R1, an egress-associated receptor discussed in detail below) in murine models.26
III. MATURE THYMOCYTE EGRESS FROM THE THYMUS
After completing their development in the thymus, thymocytes that are prepared to egress the thymus for the periphery are considered mature naïve T lymphocytes. Our work and that of other investigators has determined the phenotype of these cells to be CD3hiCD27+CD45RA+CD62L+CD69−.29–31 As shown by Vanhecke et al., CD69 is lost before thymocyte egress of mature naïve cells24 and can therefore be used reliably as a marker of fully mature thymocytes prepared for egress, in conjunction with the additional surface markers above. Interestingly, these mature T cells are refractory to death by apoptosis (programmed cell death), in contrast to less mature subsets, which are susceptible to apoptosis. Instead, mature SP cells respond to TCR stimulation by proliferating.32 They also must be able to respond to signals that “tell” them it is time to egress the thymus for the periphery. Once they leave the thymus, these cells are known as recent thymic emigrants (RTEs; Figure 2).
FIG. 2.
Expression of S1P-R1 across developmental stages in the human thymus.
Many of the chemokine receptors and chemokines involved in egress from the thymus have been well characterized, but this work has been performed predominantly in murine models and may not necessarily reflect the situation in humans. Therefore, it is important to consider that the current understanding of the process of thymic egress in humans is quite fluid and thus subject to fine-tuning as methods emerge to permit in-depth work in humans or as more researchers begin to work with human tissue samples. Briefly, it is known that the chemokine receptor CXCR4 and its ligand CXCL12 are important molecules involved in egress from the thymus to the periphery. CXCL12, also known as stromal-derived factor 1 (SDF-1), is a potent lymphocyte chemoattractant known to retain thymocytes at the corticomedullary junction,33 the region of the thymus microenvironment at which thymocytes enter and exit the thymus until the cells are “told” to leave.34 Additional factors such as the early growth response gene (Egr1, a transcriptional regulator involved in differentiation and mitogenesis),35 integrin α5β1 (a mediator of migration and proliferation),36,37 aryl hydrocarbon receptor (AHR, a helix-loop-helix transcription factor),38,39 laminin-5 (a mediator of migration, organization, and attachment in tissues),40 CCR7 (a mediator of migration from the cortex to the medulla during selection),14,41,42 KLF2 (see above),27 PI3 kinase (PI3K, a negative regulator of KLF2),43 and phosphatase and tensin homolog (PTEN, a regulator of PI3K)44 also bear mentioning because they have been shown to have roles in the process of egress (although much of the research is murine work). According to “textbook knowledge,” this maturation process takes ~2 weeks.45 However, it has recently been discovered that the time between positive selection and emigration is much less than previously believed: 4 or 5 days as opposed to the earlier estimate of 14 days. Additionally, emigration may not be stochastic as previously believed, but, rather, some data point toward the most immature thymocytes leaving before less mature medullary cells, although this has not been unequivocally demonstrated.32,46,47
There are limited data on the mechanisms regulating egress of mature human thymocytes from the thymus to the periphery. Murine studies investigated the S1P/S1P-R1 ligation prerequisite for thymocyte egress and demonstrated that S1P-R1 is essential for thymic egress of mature SP murine thymocytes.48,49 However, no reports to date have demonstrated whether the S1P/S1P receptor pathway has a role in the response of mature thymocytes to S1P or egress from the human thymus to the peripheral blood and lymphoid tissues; moreover, the exact population of thymocytes that expresses S1P-R1 has not been fully characterized in either mice or humans.
The following section describes the role of sphingosine-1-phosphate in the egress of mature, SP CD4 and CD8 thymocytes from the thymus to the periphery in mice and humans, the characterization of which is the main focus of this review.
IV. SPHINGOSINE-1-PHOSPHATE AND ITS ROLE IN CHEMOTAXIS
S1P is a signaling sphingolipid molecule, also known as a lysosphingolipid, with multiple tasks throughout the body.50 The name “sphingosine” in fact originates from the mythological Greek character the sphinx, in reference to the molecule’s promiscuous nature in the human body.51 S1P functions in a myriad of roles in humans, from regulation of cell death (specifically, suppression of apoptosis)52 and survival,53,54 immune responses,55 autoimmune conditions and allergies,56–58 B-cell development,59 insulin modulation,60 cell motility, 61,62 and response to viral infections.63,64 In addition, S1P is an “integral constituent” of high-density lipoprotein (HDL) complexes in the plasma and not only plays a role in regulation of its bioactivity but also contributes to protection against atherosclerosis.65–67 The S1P molecule is comprised of a long saturated carbohydrate chain and one alcohol, amino, and phosphate group.68 S1P is produced by phosphorylation of its precursor, sphingosine, by sphingosine kinase, which is found ubiquitously in the cytoplasm and endoplasmic reticulum of many cell types69,70 and can also be released from ceramides via their conversion to sphingosine and subsequent phosphorylation.71
Among its many roles, S1P is crucial for proper lymphocyte trafficking between the lymphoid organs and about the periphery48,72 and the trafficking of various other cell types such as smooth muscle and endothelial cells.73 An S1P gradient is present in which S1P is concentrated in the blood (about 100 nm) where it is taken up, phosphorylated, stored, transported, and released by erythrocytes74,75 and activated platelets76 in response to factors in the serum75 but is at a low concentration in lymphoid tissues. S1P is degraded by sphingosine lyase in lymphoid tissues, thus keeping tissue concentrations low77; sphingosine phosphatase also aids in breaking down S1P in tissues.78 Sphingosine kinase, sphingosine phosphatase, and sphingosine lyase together maintain the S1P gradient (see also Figure 3), which promotes the influx of lymphocytes bearing one of its five G-protein-coupled receptors to the lymph nodes and other lymphoid tissues. The S1P receptor family is known to include S1P-R1-5; these receptors are present on various cell types.79 S1P receptors had not been characterized on human thymocytes before our recent work (Resop et al. Journal of Allergy and Clinical Immunology. In press, DOI 10.1016/j.jaci.2015.12.1339), although murine work alluded to their importance in emigration from the thymus.48
FIG. 3.
Migration of mature naïve T cells toward S1P in the blood.
In murine models, it has been shown that S1P/S1P-R1 ligation is required for mature T cells to egress the thymus to the periphery. Mice with siRNA-ablated S1P-R1 have virtually no naïve CD4 or CD8 SP cells in the peripheral lymph nodes; treatment of mice with FTY720, an immune modulator that targets S1P-R1, 3, 4, and 5, recapitulates this effect.48,49 S1P-R1 also has a role in migration of T cells to and from peripheral lymphoid organs.80 S1P-R2 has been shown to inhibit migration of maturing B cells in the germinal centers when ligated to S1P.81 However, no studies have demonstrated the role of S1P and its receptors in human thymocyte egress.
V. ADDITIONAL MARKERS FOR MATURATION STAGES OF THYMOCYTES IN THE HUMAN THYMUS
As described above, various markers are used to identify the stages of thymocyte development, including CD3, CD4, CD8, CD45RA, CD27, CD69, and CD62L. CD31, also known as PECAM-1, is well known as a marker of RTEs and interestingly as a marker of CD4+ (but not CD8+) RTE in HIV patients,82 although additionally it has been reported that CD31 is expressed on human thymocytes.83 CD31 has been shown to have the ability to modulate the TCR activation threshold via activity of tyrosine phosphatases and may therefore have a role in the actual T-cell selection process84,85 Additionally, the characterization of CD31 expression on thymocytes is ongoing and applies to the field of thymocyte egress and the characterization of receptors and ligands involved in human that CD31 may additionally be expressed in mature thymocytes prepared for egress (and, thus, its expression may correlate with that of other factors responsible for egress). Perhaps, CD31 may function synergistically with other chemotaxis receptors to allow egress of mature thymocytes from thymus to periphery; it will be intriguing to follow the emergence of new data in this field in the coming years.
VI. MURINE MODELS AND THEIR RELEVANCE FOR HUMAN IMMUNOLOGY
The adaption of mouse models to the study of mechanisms of immune function was a scientific milestone and a key step in achieving a greater understanding of the human immune system. According to H.C. Morse, a researcher who was at the forefront of the development of murine models for research, “Our current realization of the potential of the inbred mouse has developed over a period of more than 70 years and reflects the dedicated research of many skilled and imaginative scientists.”86 The majority of the facts discussed above, including the characterization of T-cell development in the thymus, the various facets of the lymphocyte repertoire, and the necessity for cytokines such as S1P for T-cell egress from the thymus, were made possible by the use of mice in research.45,48,87,88 Murine models began to be used in 1929 with the establishment of the Jackson Laboratory and the development of the first inbred mouse strain,89,90 which eventually led to the development of various models of inbred mice designed to gain a better understanding of human diseases.86,91 With a repertoire of model systems in which we may manipulate various aspects, such as, for example, knocking out the expression of a specific gene, researchers are able to observe the resultant phenotype and thus infer the function of a particular facet of the immune system. Additionally, murine models of a wide range of diseases and autoimmune conditions have been developed. Moreover, mice closely resemble humans in regard to many aspects of the immune system (however, there are indeed many differences, described below), are relatively simple to work with, and have a short reproductive time frame; thus; they serve as invaluable modern investigative tools.
Murine work is an adequate model of the immune system of humans in many ways. Like humans, mice develop T, B, NK and dendritic cells; macrophages; monocytes; and other important immune cells. Many of the human cytokines studied in immunology, or analogous cytokines, are also produced in mice, although the effects of cytokines can vary.92,93 Recent reports indicate that only ~300 genes differ between mice and humans.94 However, there exist several discrepancies between the mouse and human immune system, evidenced in both adaptive and innate branches of the immune system.95 These differences may influence the interpretation of results of immunological research, in the aim of understanding the basics of immune system development, activation, and response to challenge. Some of the key areas in which differences are observed between mouse and human include balance of lymphocyte subsets; B- and T-cell signaling pathways; T-helper 1 and 2 (Th1 and Th2) cells; certain cytokines, chemokines, and their receptors; toll-like receptors; antigen presentation by endothelial cells; and expression and function of costimulatory molecules.96 A recent report also demonstrated that genes that are up- or down-regulated in response to various immunological challenges (i.e., infectious agents/exotoxins, burn trauma, etc.) differ significantly in mice and humans; the authors concluded that genomic responses in mice “poorly mimic response in humans.”97
In an attempt to address the above issues, humanized mouse models have been developed. Notably, the majority of humanized mouse models evolved from HIV research laboratories, because researchers needed a way to examine the effect of HIV on humans and found traditional murine systems inadequate. The severe combined immunodeficient (SCID) mouse was the first such model to be used.98,99 Today, several more advanced humanized mouse models exist, and the underlying shared feature of all models is a reconstitution in the mouse of a “human-like” immune system. In general, genetic manipulation renders certain aspects of the murine immune system ineffective; components such as murine T, B, and NK cells do not develop, and human lymphocyte precursors are normally introduced that develop into human immune cells in the mouse.100 The immunodeficient nonobese diabetic (NOD), severe combined immunodeficient, common γ-chain knockout (γc−/−) (NSG) thy/liv model is generated by (1) a NOD genetic background that reduces innate immunity in the mouse, resulting in macrophages and dendritic cells that are not functional, (2) a SCID mutation (loss-of-function mutation in the Prkdc gene, which is responsible for repairing the double-stranded DNA break that occurs during v-d-j recombination in T- and B-cell development), resulting in compromised adaptive immunity (does not develop) in T and B cells, and (3) a common γ-chain mutation (the IL2rγ gene) that compromises a common key component of the receptor for six interleukins (IL-2, 4, 7, 9, 15, and 21), resulting in murine NK cells that do not develop. These mice may be implanted with human fetal thymus and liver tissue and injected with human fetal CD34+ hematopoietic progenitor cells, which develop in the thymus into functional “human-like” T lymphocytes (this type of mouse is commonly known as a BLT mouse.) B and NK cells of human origin also develop in the animal, allowing a strikingly accurate reconstruction of a humanoid immune system within the mouse.101,102 For a view of the generation of the NSG thy/liv mouse, see Figure 4.
FIG. 4.
Schematic diagram of the generation of humanized bone marrow/liver/thymus (BLT) mice used in HIV research.
Thus, the NSG thy/liv mouse, as well as other humanized mouse models, have come into favor for modeling HIV infection and are beginning to be used to model other human diseases as well, because they appear to more accurately represent the human immune response to challenge. For example, in HIV infection of the BLT mouse, the cellular response observed, as well as the ability of the virus to escape the immune response mounted by the host, is similar to that observed in humans.103–105 It is crucial to continue to expand the use of humanized mouse models to research of a wide variety of disease states, considering that several therapies that have been demonstrated to work in mouse models have failed to have the desired effect in human trials.106–111 Additionally, multiple differences exist between murine and human thymus immunology, which must be taken into consideration when using animal models to investigate human thymocyte development and egress. The human response to HIV infection, particularly in the thymus, is discussed below.
VII. HIV INFECTION AND INFECTION OF THE THYMUS
A. Human Immunodeficiency Virus
The prelude to what became known as acquired immunodeficiency syndrome (AIDS) was first described in the early 1980s as a mysterious cluster of symptoms, including Kaposi’s sarcoma, that appeared to disproportionately target homosexual men, injection drug users, hemophiliacs, and Haitians.112–118 The disease remained an enigma during the early 1980s and caused considerable fear among the public and controversy over extremist sentiments regarding the groups of individuals affected.119,120 The infectious agent responsible for AIDS, a virus with similar characteristics to known lentiviruses, was reported simultaneously in 1983 by French and American research groups led by Luc Montagnier and Robert Gallo,121,122 who would later vie for credit for the discovery. Following its initial characterization, virologists and immunologists raced to learn more about the infectious agent. It was soon discovered that the HIV targets lymphocytes, or cells of the immune system, especially T cells, within which it replicates,123 and that the virus uses CD4124 as well as one of two coreceptors, CXCR4 or CCR5125 for infection of T cells. Further characterization of the virus itself revealed it to be a retrovirus, more specifically, a lentivirus (a subset of retroviruses) comprised of two copies of single-stranded RNA capable of reverse transcription (creation of DNA from RNA) and integration into the host genome.125 The virus wreaks havoc on the immune system in many ways, but most significantly by depleting CD4+ T cells, which impairs cell-mediated immunity and eventually allows opportunistic infections that the host would normally be able to clear to cause irreversible damage.126–128
Transmission of HIV most commonly occurs via sexual contact, resulting in exposure to either cell-associated or free infectious virus particles.129 This route of infection includes male-to-female, female-to-male, or male-to-male sexual transmission across mucosal (epithelial cell) surfaces.130 Less commonly, HIV is transmitted via intravenous drug use or the sharing of needles for other purposes,131 intrauterine or mother-to-child transmission during birth,132 or contact with infected bodily fluids including blood,133 semen,134 breast milk,135,136 or vaginal fluid.137 The basic mechanisms of HIV infection, epidemiology of the disease, and the immune response to infection are reviewed in several manuscripts;129,138–144 the subject of this review is the potential effect of established HIV-1 infection on the specific requisite thymocyte egress-mediating cell-surface receptor S1P-R1.
Perturbations in lymphocyte count, cell populations, and cytokines during HIV-1 infection all have the potential to affect expression of cell-surface receptors. This has not been examined as it pertains to thymocyte egress receptors but has been considered in other systems.145,146 Cytokine perturbations during HIV infection have mostly been characterized in the periphery and not within the thymus proper. For example, it has been well documented that many proinflammatory cytokines peak several days after an initial infection event, and each cytokine has its own corresponding time point postinfection at which it peaks in concentration in the periphery.147
B. Typical Infection of the Human Thymus
HIV infection spreads to various organs of the body, including the thymus.148–151 CD4 is expressed in a large proportion of thymocytes including double-positive and single (CD4)- positive cells, CXCR4 is expressed at high levels within the cortical subset (mainly double-positive cells), and CCR5 is expressed within the medullary subset (mainly mature SP cells). Thus, there are ample targets for both CXCR4 and CCR5-tropic HIV infection within the thymus.152 Interestingly, we have observed that CD25+FoxP3+ thymic Tregs are preferentially infected by CCR5-tropic HIV-1, whereas CXCR4-tropic virus does not readily infect this thymic subset (unpublished data).153 During the course of infection, susceptible subsets are depleted (Resop R, unpubl.),148,154 leaving immature double-negative thymocytes and some SP cells (especially CD8 SP cells) remaining. Moreover, CD4 is down-regulated on infected T cells by gp120, an HIV accessory protein155,156 contributing not only to a depleted thymus but also a significantly altered profile of the remaining cells therein and a skewed peripheral T-cell profile as well.157 However, the thymus continues to contribute to reconstitution. The extent to which it may perform this duty depends on the success of highly active antiretroviral therapy (HAART) treatment within the individual, the stage of HIV infection, and the age of the infected individual, among other factors.154
Because these important changes in the human thymus during HIV infection needed to be examined, a model system was necessary. Fortunately, humanized mice are well suited for the modeling of HIV infection of the thymus. In SCID and recombinant activating gene (RAG−/−) thy/liv mice infected with CCR5 (R5)-tropic or CXCR4 (X4)-tropic HIV-1 (the two tropisms of HIV, using the CCR5 or CXCR4 coreceptor) intraperitoneally or intrathymically, the thymus becomes robustly infected after several weeks.158 We have observed that cytokines, including interferon-α (IFN-α) and TNF-α are elevated in intrathymically infected human fetal thymic implants in immunodeficient mice by 5 weeks postinfection (Resop et al., manuscript in preparation). Intriguingly, a transient boost in thymopoiesis following an initial infection event has been observed in models of simian immunodeficiency virus (SIV),159–161 but the mechanisms involved or the duration of this effect of infection have not yet been elucidated.
VIII. CONCLUDING REMARKS
The role of S1P and its receptors plays a part in the egress of mature human thymocytes from the thymus to the periphery, and the only work on this subject to date had been performed in mice. Seminal work by Matloubian et al., showing that S1P1−/− hematopoietic cells remained sequestered in the thymus after development into mature CD4+ and CD8+ T cells and could not egress to the periphery, was performed in a traditional (nonhumanized) mouse model that could not be assumed to reflect the function of the S1P/S1P receptor system in humans.48 Thus, it is imperative to examine the function of the S1P/S1P receptor system in the human thymus and to identify and characterize the populations expressing S1P receptors and those that respond to S1P.
Because HIV infection results in extensive T-cell depletion, there is an urgent need to examine approaches for reconstitution of the T-cell “repertoire” in HIV-infected individuals. Reconstitution of a peripheral repertoire depends largely on functional and enhanced thymopoiesis or generation of mature naïve T cells in the thymus. The egress of mature T cells following development is critical to reconstitution and had thus far only been examined superficially; Dion et al. and other investigators noted a transient increase in T -ell output in the acute (earliest) stage of HIV infection,162–164 but little follow-up work has been performed to address whether this mechanism is maintained for a considerable duration or whether T-cell output is quickly impaired following a short-lived initial burst. Other researchers have noted that the thymus, as it is infected and the cells therein depleted, eventually declines in function in HIV patients.165,166 Many HIV-infected individuals, compliance with HAART notwithstanding, do not experience ample reconstitution of the T-cell/TCR repertoire.167 The function of S1P in the egress of mature thymocytes during HIV infection has not been examined. S1P and its receptors, if found to be present and functional during HIV infection, could represent a tantalizing new possibility for HIV therapy if the system were able to be modulated to promote enhanced thymic output and controlled so that only mature T cells would egress and repopulate the periphery.
Acknowledgments
This work would not have been possible without Dr. John L. Fahey’s outstanding mentorship to Christel Uittenbogaart when she switched her career from clinical work to basic science. Dr. Fahey’s foresight of the importance of unraveling the immune responses in cancer and infections such as HIV stimulated the type of research essential for the development of breakthrough therapies to combat these diseases. His contributions to science and to the education of new generations of basic and clinical researchers are unequaled. Research support for the present work was made possible by from the National Institutes of Health (NIH) grants AI069218, AI080564, and AI102771 to C.U., an Idea Award (ID08-LA-053) to C.U., and a dissertation fellowship (D13-LA-394) to R.R. from the University of California HIV/AIDS Research Program. The research could not have been accomplished without the excellent assistance of all current and past members of the Uittenbogaart laboratory and the technical support of the UCLA Jonsson Comprehensive Cancer Center (JCCC)/Center for AIDS Research Flow Cytometry Core Facility (supported by NIH grants P30 CA016042 and 5P30 AI028697) and the UCLA CFAR Virology Core (supported by NIH grant 5P30 AI028697).
ABBREVIATIONS
- BLT
Bone marrow/liver/thymus
- HIV
human immunodeficiency virus
- HSC
hematopoietic stem cell
- KLF2
Kruppel-like factor 2
- NK
natural killer
- RTE
recent thymic emigrants
- SCID
severe combined immunodeficiency
- S1P-R1
sphingosine-1-phosphate receptor 1
- TCR
T-cell receptor
- thy/liv
thymus/liver
References
- 1.Murphy KJ, Charles A, Jr, Travers P, Walport M. Immunobiology. 8. Garland Science; 2012. [Google Scholar]
- 2.Medzhitov R, Janeway CA., Jr Innate immunity: The virtues of a nonclonal system of recognition. Cell. 1997;91:295–8. doi: 10.1016/s0092-8674(00)80412-2. [DOI] [PubMed] [Google Scholar]
- 3.Hazenberg MD, Spits H. Human innate lymphoid cells. Blood. 2014;124:700–9. doi: 10.1182/blood-2013-11-427781. [DOI] [PubMed] [Google Scholar]
- 4.Grimaldi CM, Jeganathan V, Diamond B. Hormonal regulation of B cell development: 17 β-estradiol impairs negative selection of high-affinity DNA-reactive B cells at more than one developmental checkpoint. J Immunol. 2006;176:2703–10. doi: 10.4049/jimmunol.176.5.2703. [DOI] [PubMed] [Google Scholar]
- 5.Carsetti R, Kohler G, Lamers MC. Transitional B cells are the target of negative selection in the B cell compartment. J Exp Med. 1995;181:2129–40. doi: 10.1084/jem.181.6.2129. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Giltiay NV, Chappell CP, Clark EA. B-cell selection and the development of autoantibodies. Arthritis Res Ther. 2012;14(Suppl 4):S1. doi: 10.1186/ar3918. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Chappell CP, Jacob J. Germinal-center-derived B-cell memory. Adv Exp Med Biol. 2007;590:139–48. doi: 10.1007/978-0-387-34814-8_10. [DOI] [PubMed] [Google Scholar]
- 8.Goodnow CC, Vinuesa CG, Randall KL, Mackay F, Brink R. Control systems and decision making for antibody production. Nat Immunol. 2010;11:681–8. doi: 10.1038/ni.1900. [DOI] [PubMed] [Google Scholar]
- 9.Schwarz BA, Bhandoola A. Trafficking from the bone marrow to the thymus: A prerequisite for thymopoiesis. Immunol Rev. 2006;209:47–57. doi: 10.1111/j.0105-2896.2006.00350.x. [DOI] [PubMed] [Google Scholar]
- 10.Owen JA, Punt CJ, Stranford SA. Kuby immunology. 7. New York: W.H. Freeman; 2007. [Google Scholar]
- 11.Egerton M, Scollay R, Shortman K. Kinetics of mature T-cell development in the thymus. Proc Natl Acad Sci U S A. 1990;87:2579–82. doi: 10.1073/pnas.87.7.2579. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.McDuffie M, Roehm N, Born W, Marrack P, Kappler JW. T cell receptor/MHC interactions in the thymus and the shaping of the T cell repertoire. Transplant Proc. 1987;19:111–6. [PubMed] [Google Scholar]
- 13.Marrack P, Kappler J. The T cell receptor. Science. 1987;238:1073–9. doi: 10.1126/science.3317824. [DOI] [PubMed] [Google Scholar]
- 14.Kwan J, Killeen N. CCR7 directs the migration of thymocytes into the thymic medulla. J Immunol. 2004;172:3999–4007. doi: 10.4049/jimmunol.172.7.3999. [DOI] [PubMed] [Google Scholar]
- 15.Ueno T, Saito F, Gray DH, Kuse S, Hieshima K, Nakano H, Kaikiuchi T, Lipp M, Boyd RL, Takahama Y. CCR7 signals are essential for cortex-medulla migration of developing thymocytes. J Exp Med. 2004;200:493–505. doi: 10.1084/jem.20040643. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Takahama Y. Journey through the thymus: Stromal guides for T-cell development and selection. Nat Rev Immunol. 2006;6:127–35. doi: 10.1038/nri1781. [DOI] [PubMed] [Google Scholar]
- 17.Kappler JW, Roehm N, Marrack P. T cell tolerance by clonal elimination in the thymus. Cell. 1987;49:273–80. doi: 10.1016/0092-8674(87)90568-x. [DOI] [PubMed] [Google Scholar]
- 18.Marrack P, Lo D, Brinster R, Palmiter R, Burkly L, Flavell RH, Kappler J. The effect of thymus environment on T cell development and tolerance. Cell. 1988;53:627–34. doi: 10.1016/0092-8674(88)90578-8. [DOI] [PubMed] [Google Scholar]
- 19.Surh CD, Sprent J. T-cell apoptosis detected in situ during positive and negative selection in the thymus. Nature. 1994;372:100–3. doi: 10.1038/372100a0. [DOI] [PubMed] [Google Scholar]
- 20.Li QJ, Chau J, Ebert PJ, Sylvester G, Min H, Liu G, Braich R, Manoharan NM, Soutschek J, Scare P, Klein LO, Davis MM, Chen CZ. miR-181a is an intrinsic modulator of T cell sensitivity and selection. Cell. 2007;129:147–61. doi: 10.1016/j.cell.2007.03.008. [DOI] [PubMed] [Google Scholar]
- 21.Colantonio AD. The innate immune response to human immunodeficicency virus-1 infection of the human thymus [dissertation] Los Angeles (CA): University of California, Los Angeles; 2008. [Google Scholar]
- 22.Lind EF, Prockop SE, Porritt HE, Petrie HT. Mapping precursor movement through the postnatal thymus reveals specific microenvironments supporting defined stages of early lymphoid development. J Exp Med. 2001;194:127–34. doi: 10.1084/jem.194.2.127. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Fujii Y, Okumura M, Inada K, Nakahara K, Matsuda H. CD45 isoform expression during T cell development in the thymus. Eur J Immunol. 1992;22:1843–50. doi: 10.1002/eji.1830220725. [DOI] [PubMed] [Google Scholar]
- 24.Vanhecke D, Leclercq G, Plum J, Vandekerckhove B. Characterization of distinct stages during the differentiation of human CD69+CD3+ thymocytes and identification of thymic emigrants. J Immunol. 1995;155:1862–72. [PubMed] [Google Scholar]
- 25.Feng C, Woodside KJ, Vance BA, El-Khoury D, Canelles M, Lee J, Gress R, Fowlkus BJ, Shores EW, Love PE. A potential role for CD69 in thymocyte emigration. Int Immunol. 2002;14:535–44. doi: 10.1093/intimm/dxf020. [DOI] [PubMed] [Google Scholar]
- 26.Bai A, Hu H, Yeung M, Chen J. Kruppel-like factor 2 controls T cell trafficking by activating L-selectin (CD62L) and sphingosine-1-phosphate receptor 1 transcription. J Immunol. 2007;178:7632–9. doi: 10.4049/jimmunol.178.12.7632. [DOI] [PubMed] [Google Scholar]
- 27.Carlson CM, Endrizzi BT, Wu J, Ding X, Weinreich MA, Walsh ER, Wani MA, Lindgrel JB, Hogquist KA, Jameson SC. Kruppel-like factor 2 regulates thymocyte and T-cell migration. Nature. 2006;442:299–302. doi: 10.1038/nature04882. [DOI] [PubMed] [Google Scholar]
- 28.Sebzda E, Zou Z, Lee JS, Wang T, Kahn ML. Transcription factor KLF2 regulates the migration of naive T cells by restricting chemokine receptor expression patterns. Nat Immunol. 2008;9:292–300. doi: 10.1038/ni1565. [DOI] [PubMed] [Google Scholar]
- 29.Gurney KB, Uittenbogaart CH. Human immunodeficiency virus persistence and production in T-cell development. Clin Vaccine Immunol. 2006;13:1237–45. doi: 10.1128/CVI.00184-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Xu X, Zhang S, Li P, Lu J, Xuan Q, Ge Q. Maturation and emigration of single-positive thymocytes. Clin Dev Immunol. 2013;2013:282870. doi: 10.1155/2013/282870. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Berkley AM, Hendricks DW, Simmons KB, Fink PJ. Recent thymic emigrants and mature naive T cells exhibit differential DNA methylation at key cytokine loci. J Immunol. 2013;190:6180–6. doi: 10.4049/jimmunol.1300181. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.McCaughtry TM, Wilken MS, Hogquist KA. Thymic emigration revisited. J Exp Med. 2007;204:2513–20. doi: 10.1084/jem.20070601. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Bleul CC, Fuhlbrigge RC, Casasnovas JM, Aiuti A, Springer TA. A highly efficacious lymphocyte chemoattractant, stromal cell-derived factor 1 (SDF-1) J Exp Med. 1996;184:1101–9. doi: 10.1084/jem.184.3.1101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Fink PJ. The biology of recent thymic emigrants. Annu Rev Immunol. 2013;31:31–50. doi: 10.1146/annurev-immunol-032712-100010. [DOI] [PubMed] [Google Scholar]
- 35.Schnell FJ, Kersh GJ. Control of recent thymic emigrant survival by positive selection signals and early growth response gene 1. J Immunol. 2005;175:2270–7. doi: 10.4049/jimmunol.175.4.2270. [DOI] [PubMed] [Google Scholar]
- 36.Cotta-de-Almeida V, Villa-Verde DM, Lepault F, Pleau JM, Dardenne M, Savino W. Impaired migration of NOD mouse thymocytes: A fibronectin receptor-related defect. Eur J Immunol. 2004;34:1578–87. doi: 10.1002/eji.200324765. [DOI] [PubMed] [Google Scholar]
- 37.Li R, Maminishkis A, Zahn G, Vossmeyer D, Miller SS. Integrin α5β1 mediates attachment, migration, and proliferation in human retinal pigment epithelium: Relevance for proliferative retinal disease. Invest Ophthalmol Vis Sci. 2009;50:5988–96. doi: 10.1167/iovs.09-3591. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Temchura VV, Frericks M, Nacken W, Esser C. Role of the aryl hydrocarbon receptor in thymocyte emigration in vivo. Eur J Immunol. 2005;35:2738–47. doi: 10.1002/eji.200425641. [DOI] [PubMed] [Google Scholar]
- 39.Hankinson O. The aryl hydrocarbon receptor complex. Annu Rev Pharmacol Toxicol. 1995;35:307–40. doi: 10.1146/annurev.pa.35.040195.001515. [DOI] [PubMed] [Google Scholar]
- 40.Vivinus-Nebot M, Rousselle P, Breittmayer JP, Cenciarini C, Berrih-Aknin S, Spong S, Nokeilainen P, Cottrez F, Marinkovich MP, Bernard A. Mature human thymocytes migrate on laminin-5 with activation of metalloproteinase-14 and cleavage of CD44. J Immunol. 2004;172:1397–1406. doi: 10.4049/jimmunol.172.3.1397. [DOI] [PubMed] [Google Scholar]
- 41.Kurobe H, Liu C, Ueno T, Saito F, Ohigashi I, Seach N, Arakaki R, Hiyashi Y, Kitagawa T, Lipp M, Boyd RL, Takahama Y. CCR7-dependent cortex-to-medulla migration of positively selected thymocytes is essential for establishing central tolerance. Immunity. 2006;24:165–77. doi: 10.1016/j.immuni.2005.12.011. [DOI] [PubMed] [Google Scholar]
- 42.Witt CM, Robey EA. The ins and outs of CCR7 in the thymus. J Exp Med. 2004;200:405–9. doi: 10.1084/jem.20041110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Sinclair LV, Finlay D, Feijoo C, Cornish GH, Gray A, Ager A, Okkenhaug K, Hagenbeek TJ, Spits H, Cantrell DA. Phosphatidylinositol-3-OH kinase and nutrient-sensing mTOR pathways control T lymphocyte trafficking. Nat Immunol. 2008;9:513–21. doi: 10.1038/ni.1603. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Barbee SD, Alberola-Ila J. Phosphatidylinositol 3-kinase regulates thymic exit. J Immunol. 2005;174:1230–8. doi: 10.4049/jimmunol.174.3.1230. [DOI] [PubMed] [Google Scholar]
- 45.Gabor MJ, Godfrey DI, Scollay R. Recent thymic emigrants are distinct from most medullary thymocytes. Eur J Immunol. 1997;27:2010–15. doi: 10.1002/eji.1830270827. [DOI] [PubMed] [Google Scholar]
- 46.Weinreich MA, Hogquist KA. Thymic emigration: When and how T cells leave home. J Immunol. 2008;181:2265–70. doi: 10.4049/jimmunol.181.4.2265. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Jin R, Wang W, Yao JY, Zhou YB, Qian XP, Zhang J, Zhang Y, Chen WF. Characterization of the in vivo dynamics of medullary CD4+CD8− thymocyte development. J Immunol. 2008;180:2256–63. doi: 10.4049/jimmunol.180.4.2256. [DOI] [PubMed] [Google Scholar]
- 48.Matloubian M, Lo CG, Cinamon G, Lesneski MJ, Xu Y, Brinkmann V, Allende ML, Proia RL, Cyster JG. Lymphocyte egress from thymus and peripheral lymphoid organs is dependent on S1P receptor 1. Nature. 2004;427:355–60. doi: 10.1038/nature02284. [DOI] [PubMed] [Google Scholar]
- 49.Shiow LR, Rosen DB, Brdickova N, Xu Y, An J, Lanier LL, Cyster JG, Matloubian M. CD69 acts downstream of interferon-α/β to inhibit S1P1 and lymphocyte egress from lymphoid organs. Nature. 2006;440:540–4. doi: 10.1038/nature04606. [DOI] [PubMed] [Google Scholar]
- 50.Olivera A, Spiegel S. Sphingosine-1-phosphate as second messenger in cell proliferation induced by PDGF and FCS mitogens. Nature. 1993;365:557–60. doi: 10.1038/365557a0. [DOI] [PubMed] [Google Scholar]
- 51.Thudichum JLW. A treatise on the chemical constitution of the brain. 1884 [Google Scholar]
- 52.Cuvillier O, Pirianov G, Kleuser B, Vanek PG, Coso OA, Gutkind S, Spiegel S. Suppression of ceramide-mediated programmed cell death by sphingosine-1-phosphate. Nature. 1996;381:800–3. doi: 10.1038/381800a0. [DOI] [PubMed] [Google Scholar]
- 53.Spiegel S, Cuvillier O, Edsall LC, Kohama T, Menzeleev R, Olah Z, Olivera A, Pirianov G, Thomas DM, Tu Z, Van Brocklyn JR, Wang F. Sphingosine-1-phosphate in cell growth and cell death. Ann N Y Acad Sci. 1998;845:11–8. doi: 10.1111/j.1749-6632.1998.tb09658.x. [DOI] [PubMed] [Google Scholar]
- 54.Spiegel S, Cuvillier O, Edsall L, Kohama T, Menzeleev R, Olivera A, Thomas D, Tu Z, Van Brocklyn JR, Wang F. Roles of sphingosine-1-phosphate in cell growth, differentiation, and death. Biochemistry (Mosc) 1998;63:69–73. [PubMed] [Google Scholar]
- 55.Maceyka M, Harikumar KB, Milstien S, Spiegel S. Sphingosine-1-phosphate signaling and its role in disease. Trends Cell Biol. 2012;22:50–60. doi: 10.1016/j.tcb.2011.09.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Oskeritzian CA. Mast cell plasticity and sphingosine-1-phosphate in immunity, inflammation and cancer. Mol Immunol. 2014;63(1):104–12. doi: 10.1016/j.molimm.2014.03.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Oskeritzian CA, Milstien S, Spiegel S. Sphingosine-1-phosphate in allergic responses, asthma and anaphylaxis. Pharmacol Ther. 2007;115:390–9. doi: 10.1016/j.pharmthera.2007.05.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Snider AJ. Sphingosine kinase and sphingosine-1-phosphate: Regulators in autoimmune and inflammatory disease. Int J Clin Rheumtol. 2013;8(4) doi: 10.2217/ijr.13.40. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Sic H, Kraus H, Madl J, Flittner KA, von Munchow AL, Pieper K, Rizzi M, Kienzler AK, Ayata K, Rauer S, Kleuser B, Salzer U, Burger M, Zirlik K, Lougaris V, Plebani A, Römer W, Loeffler C, Scaramuzza S, Villa A, Noguchi E, Grimbacher B, Eibel H. Sphingosine-1-phosphate receptors control B-cell migration through signaling components associated with primary immunodeficiencies, chronic lymphocytic leukemia, and multiple sclerosis. J Allergy Clin Immunol. 2014;134(2):420–8. doi: 10.1016/j.jaci.2014.01.037. [DOI] [PubMed] [Google Scholar]
- 60.Arnold RS, Newton AC. Inhibition of the insulin receptor tyrosine kinase by sphingosine. Biochemistry. 1991;30:7747–54. doi: 10.1021/bi00245a011. [DOI] [PubMed] [Google Scholar]
- 61.Berdyshev EV, Gorshkova I, Usatyuk P, Kalari S, Zhao Y, Pyne NJ, Pyne S, Sabbadini RA, Garcia JGN, Natarajan V. Intracellular S1P generation is essential for S1P-induced motility of human lung endothelial cells: Role of sphingosine kinase 1 and S1P lyase. PLoS One. 2011;6:e16571. doi: 10.1371/journal.pone.0016571. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Buehrer BM, Bell RM. Sphingosine kinase: Properties and cellular functions. Adv Lipid Res. 1993;26:59–67. [PubMed] [Google Scholar]
- 63.Carr JM, Mahalingam S, Bonder CS, Pitson SM. Sphingosine kinase 1 in viral infections. Rev Med Virol. 2013;23:73–84. doi: 10.1002/rmv.1718. [DOI] [PubMed] [Google Scholar]
- 64.Carr JM, Kua T, Clarke JN, Calvert JK, Zebol JR, Beard MR, Pitson SM. Reduced sphingosine kinase 1 activity in dengue virus type-2 infected cells can be mediated by the 3′ untranslated region of dengue virus type-2 RNA. J Gen Virol. 2013;94:2437–48. doi: 10.1099/vir.0.055616-0. [DOI] [PubMed] [Google Scholar]
- 65.Poti F, Simoni M, Nofer JR. Atheroprotective role of high-density lipoprotein (HDL)-associated sphingosine-1-phosphate (S1P) Cardiovasc Res. 2014;103:395–404. doi: 10.1093/cvr/cvu136. [DOI] [PubMed] [Google Scholar]
- 66.Sachinidis A, Kettenhofen R, Seewald S, Gouni-Berthold I, Schmitz U, Seul C, Ko Y, Vetter H. Evidence that lipoproteins are carriers of bioactive factors. Arterioscler Thromb Vasc Biol. 1999;19:2412–21. doi: 10.1161/01.atv.19.10.2412. [DOI] [PubMed] [Google Scholar]
- 67.Zhang B, Tomura H, Kuwabara A, Kimura T, Miura S, Noda K, Okajima F, Saku K. Correlation of high density lipoprotein (HDL)-associated sphingosine 1-phosphate with serum levels of HDL-cholesterol and apolipoproteins. Atherosclerosis. 2005;178:199–205. doi: 10.1016/j.atherosclerosis.2004.08.024. [DOI] [PubMed] [Google Scholar]
- 68.Spiegel S, Milstien S. Sphingosine-1-phosphate: An enigmatic signalling lipid. Nat Rev Mol Cell Biol. 2003;4:397–407. doi: 10.1038/nrm1103. [DOI] [PubMed] [Google Scholar]
- 69.Lima S, Spiegel S. Sphingosine kinase: A closer look at last. Structure. 2013;21:690–2. doi: 10.1016/j.str.2013.04.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Melendez AJ, Carlos-Dias E, Gosink M, Allen JM, Takacs L. Human sphingosine kinase: Molecular cloning, functional characterization and tissue distribution. Gene. 2000;251:19–26. doi: 10.1016/s0378-1119(00)00205-5. [DOI] [PubMed] [Google Scholar]
- 71.Chalfant CE, Spiegel S. Sphingosine 1-phosphate and ceramide 1-phosphate: Expanding roles in cell signaling. J Cell Sci. 2005;118:4605–12. doi: 10.1242/jcs.02637. [DOI] [PubMed] [Google Scholar]
- 72.Cyster JG, Schwab SR. Sphingosine-1-phosphate and lymphocyte egress from lymphoid organs. Annu Rev Immunol. 2012;30:69–94. doi: 10.1146/annurev-immunol-020711-075011. [DOI] [PubMed] [Google Scholar]
- 73.Spiegel S, English D, Milstien S. Sphingosine 1-phosphate signaling: Providing cells with a sense of direction. Trends Cell Biol. 2002;12:236–42. doi: 10.1016/s0962-8924(02)02277-8. [DOI] [PubMed] [Google Scholar]
- 74.Bode C, Sensken SC, Peest U, Beutel G, Thol F, Levkau B, Li Z, Bittman R, Huang T, Tölle M, van der Giet M, Gräler MH. Erythrocytes serve as a reservoir for cellular and extracellular sphingosine 1-phosphate. J Cell Biochem. 2010;109:1232–43. doi: 10.1002/jcb.22507. [DOI] [PubMed] [Google Scholar]
- 75.Hanel P, Andreani P, Graler MH. Erythrocytes store and release sphingosine 1-phosphate in blood. FASEB J. 2007;21:1202–9. doi: 10.1096/fj.06-7433com. [DOI] [PubMed] [Google Scholar]
- 76.Ulrych T, Bohm A, Polzin A, Daum G, Nusing RM, Geisslinger G, Hohlfeld T, Schrör K, Rauch BH. Release of sphingosine-1-phosphate from human platelets is dependent on thromboxane formation. J Thromb Haemost. 2011;9:790–8. doi: 10.1111/j.1538-7836.2011.04194.x. [DOI] [PubMed] [Google Scholar]
- 77.Veltkamp R, Siebing DA, Heiland S, Schoenffeldt-Varas P, Veltkamp C, Schwaninger M, Schwab S. Hyperbaric oxygen induces rapid protection against focal cerebral ischemia. Brain Res. 2005;1037:134–8. doi: 10.1016/j.brainres.2005.01.006. [DOI] [PubMed] [Google Scholar]
- 78.Mandala SM, Thornton R, Galve-Roperh I, Poulton S, Peterson C, Olivera A, Bergstrom J, Kurtz MB, Spiegel S. Molecular cloning and characterization of a lipid phosphohydrolase that degrades sphingosine-1-phosphate and induces cell death. Proc Natl Acad Sci U S A. 2000;97:7859–64. doi: 10.1073/pnas.120146897. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Spiegel S, Milstien S. The outs and the ins of sphingosine-1-phosphate in immunity. Nat Rev Immunol. 2011;11:403–15. doi: 10.1038/nri2974. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Pham TH, Okada T, Matloubian M, Lo CG, Cyster JG. S1P1 receptor signaling overrides retention mediated by Gαi-coupled receptors to promote T cell egress. Immunity. 2008;28:122–33. doi: 10.1016/j.immuni.2007.11.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Green JA, Cyster JG. S1PR2 links germinal center confinement and growth regulation. Immunol Rev. 2012;247:36–51. doi: 10.1111/j.1600-065X.2012.01114.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Tanaskovic S, Fernandez S, Price P, Lee S, French MA. CD31 (PECAM-1) is a marker of recent thymic emigrants among CD4+ T-cells, but not CD8+ T-cells or γd T-cells, in HIV patients responding to ART. Immunol Cell Biol. 2010;88:321–7. doi: 10.1038/icb.2009.108. [DOI] [PubMed] [Google Scholar]
- 83.Kohler S, Thiel A. Life after the thymus: CD31+ and CD31− human naive CD4+ T-cell subsets. Blood. 2009;113:769–74. doi: 10.1182/blood-2008-02-139154. [DOI] [PubMed] [Google Scholar]
- 84.Fornasa G, Groyer E, Clement M, Dimitrov J, Compain C, Gaston AT, Varthaman A, Khallou-Laschet J, Newman DK, Graff-Dubois S, Nicoletti A, Caligiuri G. TCR stimulation drives cleavage and shedding of the ITIM receptor CD31. J Immunol. 2010;184:5485–92. doi: 10.4049/jimmunol.0902219. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Ma L, Mauro C, Cornish GH, Chai JG, Coe D, Fu H, Patton D, Okkenhaug K, Franzoso G, Dyson J, Nourshargh S, Marelli-Berg FM. Ig gene-like molecule CD31 plays a nonredundant role in the regulation of T-cell immunity and tolerance. Proc Natl Acad Sci U S A. 2010;107:19461–6. doi: 10.1073/pnas.1011748107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Morse HC., 3rd . The origins of inbred mice. Bethesda, Maryland: National Institutes of Health; 1978. [Google Scholar]
- 87.Marodon G, Rocha B. Generation of mature T cell populations in the thymus: CD4 or CD8 down-regulation occurs at different stages of thymocyte differentiation. Eur J Immunol. 1994;24:196–204. doi: 10.1002/eji.1830240131. [DOI] [PubMed] [Google Scholar]
- 88.Gabor MJ, Scollay R, Godfrey DI. Thymic T cell export is not influenced by the peripheral T cell pool. Eur J Immunol. 1997;27:2986–93. doi: 10.1002/eji.1830271135. [DOI] [PubMed] [Google Scholar]
- 89.Green EL, editor. Biology of the laboratory mouse. 2. New York: Dover; 1966. [Google Scholar]
- 90.Keeler CE. The laboratory mouse: Origins, heredity and culture. Cambridge: Harvard University Press; 1931. [Google Scholar]
- 91.Morse HC., 3rd The laboratory mouse: A historical perspective. The Mouse in Biomedical Research. 1981 [Google Scholar]
- 92.Langlet C, Springael C, Johnson J, Thomas S, Flamand V, Leitges M, Goldman M, Aksoy E, Willems F. PKC-α controls MYD88-dependent TLR/IL-1R signaling and cytokine production in mouse and human dendritic cells. Eur J Immunol. 2010;40:505–15. doi: 10.1002/eji.200939391. [DOI] [PubMed] [Google Scholar]
- 93.Tunyogi-Csapo M, Kis-Toth K, Radacs M, Farkas B, Jacobs JJ, Finnegan A, Mikecz K, Glant TT. Cytokine-controlled RANKL and osteoprotegerin expression by human and mouse synovial fibroblasts: Fibroblast-mediated pathologic bone resorption. Arthritis Rheum. 2008;58:2397–2408. doi: 10.1002/art.23653. [DOI] [PubMed] [Google Scholar]
- 94.Mouse Genome Sequencing Consortium. Waterston RH, Lindblad-Toh K, Birney E, Rogers J, Abril JF, Agarwal P, Agarwala R, Ainscough R, Alexandersson M, An P, Antonarakis SE, Attwood J, Baertsch R, Bailey J, Barlow K, Beck S, Berry E, Birren B, Bloom T, Bork P, Botcherby M, Bray N, Brent MR, Brown DG, Brown SD, Bult C, Burton J, Butler J, Campbell RD, Carninci P, Cawley S, Chiaromonte F, Chinwalla AT, Church DM, Clamp M, Clee C, Collins FS, Cook LL, Copley RR, Coulson A, Couronne O, Cuff J, Curwen V, Cutts T, Daly M, David R, Davies J, Delehaunty KD, Deri J, Dermitzakis ET, Dewey C, Dickens NJ, Diekhans M, Dodge S, Dubchak I, Dunn DM, Eddy SR, Elnitski L, Emes RD, Eswara P, Eyras E, Felsenfeld A, Fewell GA, Flicek P, Foley K, Frankel WN, Fulton LA, Fulton RS, Furey TS, Gage D, Gibbs RA, Glusman G, Gnerre S, Goldman N, Goodstadt L, Grafham D, Graves TA, Green ED, Gregory S, Guigó R, Guyer M, Hardison RC, Haussler D, Hayashizaki Y, Hillier LW, Hinrichs A, Hlavina W, Holzer T, Hsu F, Hua A, Hubbard T, Hunt A, Jackson I, Jaffe DB, Johnson LS, Jones M, Jones TA, Joy A, Kamal M, Karlsson EK, Karolchik D, Kasprzyk A, Kawai J, Keibler E, Kells C, Kent WJ, Kirby A, Kolbe DL, Korf I, Kucherlapati RS, Kulbokas EJ, Kulp D, Landers T, Leger JP, Leonard S, Letunic I, Levine R, Li J, Li M, Lloyd C, Lucas S, Ma B, Maglott DR, Mardis ER, Matthews L, Mauceli E, Mayer JH, McCarthy M, McCombie WR, McLaren S, McLay K, McPherson JD, Meldrim J, Meredith B, Mesirov JP, Miller W, Miner TL, Mongin E, Montgomery KT, Morgan M, Mott R, Mullikin JC, Muzny DM, Nash WE, Nelson JO, Nhan MN, Nicol R, Ning Z, Nusbaum C, O’Connor MJ, Okazaki Y, Oliver K, Overton-Larty E, Pachter L, Parra G, Pepin KH, Peterson J, Pevzner P, Plumb R, Pohl CS, Poliakov A, Ponce TC, Ponting CP, Potter S, Quail M, Reymond A, Roe BA, Roskin KM, Rubin EM, Rust AG, Santos R, Sapojnikov V, Schultz B, Schultz J, Schwartz MS, Schwartz S, Scott C, Seaman S, Searle S, Sharpe T, Sheridan A, Shownkeen R, Sims S, Singer JB, Slater G, Smit A, Smith DR, Spencer B, Stabenau A, Stange-Thomann N, Sugnet C, Suyama M, Tesler G, Thompson J, Torrents D, Trevaskis E, Tromp J, Ucla C, Ureta-Vidal A, Vinson JP, Von Niederhausern AC, Wade CM, Wall M, Weber RJ, Weiss RB, Wendl MC, West AP, Wetterstrand K, Wheeler R, Whelan S, Wierzbowski J, Willey D, Williams S, Wilson RK, Winter E, Worley KC, Wyman D, Yang S, Yang SP, Zdobnov EM, Zody MC, Lander ES. Initial sequencing and comparative analysis of the mouse genome. Nature. 2002;420:520–62. doi: 10.1038/nature01262. [DOI] [PubMed] [Google Scholar]
- 95.Haley PJ. Species differences in the structure and function of the immune system. Toxicology. 2003;188:49–71. doi: 10.1016/s0300-483x(03)00043-x. [DOI] [PubMed] [Google Scholar]
- 96.Mestas J, Hughes CC. Of mice and not men: Differences between mouse and human immunology. J Immunol. 2004;172:2731–8. doi: 10.4049/jimmunol.172.5.2731. [DOI] [PubMed] [Google Scholar]
- 97.Seok J, Warren HS, Cuenca AG, Mindrinos MN, Baker HV, Xu W, Richards DR, McDonald-Smith GP, Gao H, Hennessy L, Finnerty CC, López CM, Honari S, Moore EE, Minei JP, Cuschieri J, Bankey PE, Johnson JL, Sperry J, Nathens AB, Billiar TR, West MA, Jeschke MG, Klein MB, Gamelli RL, Gibran NS, Brownstein BH, Miller-Graziano C, Calvano SE, Mason PH, Cobb JP, Rahme LG, Lowry SF, Maier RV, Moldawer LL, Herndon DN, Davis RW, Xiao W, Tompkins RG. Inflammation and Host Response to Injury, Large Scale Collaborative Research Program. Genomic responses in mouse models poorly mimic human inflammatory diseases. Proc Natl Acad Sci U S A. 2013;110:3507–12. doi: 10.1073/pnas.1222878110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Bosma MJ, Carroll AM. The SCID mouse mutant: Definition, characterization, and potential uses. Annu Rev Immunol. 1991;9:323–50. doi: 10.1146/annurev.iy.09.040191.001543. [DOI] [PubMed] [Google Scholar]
- 99.McCune JM, Kaneshima H, Lieberman M, Weissman IL, Namikawa R. The SCID-HU mouse: Current status and potential applications. Curr Top Microbiol Immunol. 1989;152:183–93. doi: 10.1007/978-3-642-74974-2_22. [DOI] [PubMed] [Google Scholar]
- 100.Denton PW, Garcia JV. Humanized mouse models of HIV infection. AIDS Rev. 2011;13:135–48. [PMC free article] [PubMed] [Google Scholar]
- 101.Akkina R. New generation humanized mice for virus research: Comparative aspects and future prospects. Virology. 2013;435:14–28. doi: 10.1016/j.virol.2012.10.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Laboratory TJ. Jackson Laboratory NSG Mouse Page. 2014. [Google Scholar]
- 103.Schughart K, Libert C, Kas MJ. Human disease: Strength to strength for mouse models. Nature. 492:41. doi: 10.1038/492041c. 201. [DOI] [PubMed] [Google Scholar]
- 104.Dudek TE, No DC, Seung E, Vrbanac VD, Fadda L, Bhoumik P, Boutwell CL, Power KA, Gladden AD, Battis L, Mellors EF, Tivey TR, Gao X, Altfeld M, Luster AD, Tager AM, Allen TM. Rapid evolution of HIV-1 to functional CD8+ T cell responses in humanized BLT mice. Sci Transl Med. 2012;4:143ra198. doi: 10.1126/scitranslmed.3003984. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Murooka TT, Deruaz M, Marangoni F, Vrbanac VD, Seung E, von Andrian UH, Tager AM, Luster AD, Mempel TR. HIV-infected T cells are migratory vehicles for viral dissemination. Nature. 2012;490:283–7. doi: 10.1038/nature11398. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Shepherd FA, Sridhar SS. Angiogenesis inhibitors under study for the treatment of lung cancer. Lung Cancer. 2003;41(suppl 1):S63–72. doi: 10.1016/s0169-5002(03)00144-2. [DOI] [PubMed] [Google Scholar]
- 107.Oehler MK, Bicknell R. The promise of anti-angiogenic cancer therapy. Br J Cancer. 2000;82:749–52. doi: 10.1054/bjoc.1999.0991. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Panitch HS, Hirsch RL, Haley AS, Johnson KP. Exacerbations of multiple sclerosis in patients treated with γ interferon. Lancet. 1987;1:893–5. doi: 10.1016/s0140-6736(87)92863-7. [DOI] [PubMed] [Google Scholar]
- 109.Sykes M. Mixed chimerism and transplant tolerance. Immunity. 2001;14:417–24. doi: 10.1016/s1074-7613(01)00122-4. [DOI] [PubMed] [Google Scholar]
- 110.Wood KJ. Passenger leukocytes and microchimerism: What role in tolerance induction? Transplantation. 2003;75:17S–20. doi: 10.1097/01.TP.0000067946.90241.2F. [DOI] [PubMed] [Google Scholar]
- 111.Monaco AP. Chimerism in organ transplantation: Conflicting experiments and clinical observations. Transplantation. 2003;75:13S–6. doi: 10.1097/01.TP.0000067945.90241.F4. [DOI] [PubMed] [Google Scholar]
- 112.Altman LK. The New York Times. 1981. Rare Cancer Seen in 41 Homosexuals. [Google Scholar]
- 113.Farmer P. AIDS and accusation: Haiti and the geography of blame. Berkeley and Los Angeles: University of California Press; 1992. [Google Scholar]
- 114.Gottlieb MS, Groopman JE, Weinstein WM, Fahey JL, Detels R. The acquired immunodeficiency syndrome. Ann Intern Med. 1983;99:208–20. doi: 10.7326/0003-4819-99-2-208. [DOI] [PubMed] [Google Scholar]
- 115.Jaffe HW, Bregman DJ, Selik RM. Acquired immune deficiency syndrome in the United States: The first 1,000 cases. J Infect Dis. 1983;148:339–45. doi: 10.1093/infdis/148.2.339. [DOI] [PubMed] [Google Scholar]
- 116.Fahey JL, Prince H, Weaver M, Groopman J, Visscher B, Schwartz K, Detels R. Quantitative changes in T helper or T suppressor/cytotoxic lymphocyte subsets that distinguish acquired immune deficiency syndrome from other immune subset disorders. Am J Med. 1984;76:95–100. doi: 10.1016/0002-9343(84)90756-3. [DOI] [PubMed] [Google Scholar]
- 117.Longo DL, Steis RG, Lane HC, Lotze MT, Rosenberg SA, Preble O, Masur H, Rook AH, Fauci AS, Jacob J. Malignancies in the AIDS patient: Natural history, treatment strategies, and preliminary results. Ann N Y Acad Sci. 1984;437:421–30. doi: 10.1111/j.1749-6632.1984.tb37163.x. [DOI] [PubMed] [Google Scholar]
- 118.Rosen FS. The acquired immunodeficiency syndrome (AIDS) J Clin Invest. 1985;75:1–3. doi: 10.1172/JCI111659. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Garrett L. The coming plague: Newly emerging diseases in a world out of balance. New York: The Penguin Group; 1994. [Google Scholar]
- 120.Update on acquired immune deficiency syndrome (AIDS): United States. MMWR Morb Mortal Wkly Rep. 1982;31:507–8. 513–4. [PubMed] [Google Scholar]
- 121.Barre-Sinoussi F, Chermann JC, Rey F, Nugeyre MT, Chamaret S, Gruest J, Dauguet C, Axler-Blin C, Vézinet-Brun F, Rouzioux C, Rozenbaum W, Montagnier L. Isolation of a T-lymphotropic retrovirus from a patient at risk for acquired immune deficiency syndrome (AIDS) Science. 1983;220:868–71. doi: 10.1126/science.6189183. [DOI] [PubMed] [Google Scholar]
- 122.Gallo RC, Sarin PS, Gelmann EP, Robert-Guroff M, Richardson E, Kalyanaraman VS, Mann D, Sidhu GD, Stahl RE, Zolla-Pazner S, Leibowitch J, Popovic M. Isolation of human T-cell leukemia virus in acquired immune deficiency syndrome (AIDS) Science. 1983;220:865–7. doi: 10.1126/science.6601823. [DOI] [PubMed] [Google Scholar]
- 123.Stevenson M, Stanwick TL, Dempsey MP, Lamonica CA. HIV-1 replication is controlled at the level of T cell activation and proviral integration. EMBO J. 1990;9:1551–60. doi: 10.1002/j.1460-2075.1990.tb08274.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Traunecker A, Luke W, Karjalainen K. Soluble CD4 molecules neutralize human immunodeficiency virus type 1. Nature. 1988;331:84–6. doi: 10.1038/331084a0. [DOI] [PubMed] [Google Scholar]
- 125.Kilby JM, Eron JJ. Novel therapies based on mechanisms of HIV-1 cell entry. N Engl J Med. 2003;348:2228–38. doi: 10.1056/NEJMra022812. [DOI] [PubMed] [Google Scholar]
- 126.Zanoni BC, Gandhi RT. Update on opportunistic infections in the era of effective antiretroviral therapy. Infect Dis Clin North Am. 2014;28:501–18. doi: 10.1016/j.idc.2014.05.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Siegal FP, Lopez C, Hammer GS, Brown AE, Kornfeld SJ, Gold J, Hassett J, Hirschman SZ, Cunningham-Rundles C, Adelsberg BR. Severe acquired immunodeficiency in male homosexuals, manifested by chronic perianal ulcerative herpes simplex lesions. N Engl J Med. 1981;305:1439–44. doi: 10.1056/NEJM198112103052403. [DOI] [PubMed] [Google Scholar]
- 128.Gottlieb MS, Schroff R, Schanker HM, Weisman JD, Fan PT, Wolf RA, Saxon A. Pneumocystis carinii pneumonia and mucosal candidiasis in previously healthy homosexual men: Evidence of a new acquired cellular immunodeficiency. N Engl J Med. 1981;305:1425–31. doi: 10.1056/NEJM198112103052401. [DOI] [PubMed] [Google Scholar]
- 129.Moir S, Chun TW, Fauci AS. Pathogenic mechanisms of HIV disease. Annu Rev Pathol. 2011;6:223–48. doi: 10.1146/annurev-pathol-011110-130254. [DOI] [PubMed] [Google Scholar]
- 130.Mastro TD, Satten GA, Nopkesorn T, Sangkharomya S, Longini IM., Jr Probability of female-to-male transmission of HIV-1 in Thailand. Lancet. 1994;343:204–7. doi: 10.1016/s0140-6736(94)90990-3. [DOI] [PubMed] [Google Scholar]
- 131.Meijerink H, van Crevel R, van der Ven AJ. Intravenous drug use and the spread of HIV; an international perspective. Ned Tijdschr Geneeskd. 2013;157:A5690. [PubMed] [Google Scholar]
- 132.Bryson YJ, Luzuriaga K, Sullivan JL, Wara DW. Proposed definitions for in utero versus intrapartum transmission of HIV-1. N Engl J Med. 1992;327:1246–7. doi: 10.1056/NEJM199210223271718. [DOI] [PubMed] [Google Scholar]
- 133.Leveton LB, Sox HC, Stoto MA, editors. HIV and the blood supply: An analysis of crisis decisionmaking. Washington D.C: Institute of Medicine; 1995. [PubMed] [Google Scholar]
- 134.Liu CM, Osborne BJ, Hungate BA, Shahabi K, Huibner S, Lester R, Dwan MG, Kovacs C, Contente-Cuomo TL, Benko E, Aziz M, Price LB, Kaul R. The semen microbiome and its relationship with local immunology and viral load in HIV infection. PLoS Pathog. 2014;10:e1004262. doi: 10.1371/journal.ppat.1004262. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Dunn DT, Newell ML, Ades AE, Peckham CS. Risk of human immunodeficiency virus type 1 transmission through breastfeeding. Lancet. 1992;340:585–8. doi: 10.1016/0140-6736(92)92115-v. [DOI] [PubMed] [Google Scholar]
- 136.Miotti PG, Taha TE, Kumwenda NI, Broadhead R, Mtimavalye LA, Van der Hoeven L, Chiphangwi JD, Liomba G, Biggar RJ. HIV transmission through breastfeeding: A study in Malawi. JAMA. 1999;282:744–9. doi: 10.1001/jama.282.8.744. [DOI] [PubMed] [Google Scholar]
- 137.Miller CJ, Shattock RJ. Target cells in vaginal HIV transmission. Microbes Infect. 2003;5:59–67. doi: 10.1016/s1286-4579(02)00056-4. [DOI] [PubMed] [Google Scholar]
- 138.Siliciano JD, Siliciano RF. Recent developments in the search for a cure for HIV-1 infection: Targeting the latent reservoir for HIV-1. J Allergy Clin Immunol. 2014;134:12–19. doi: 10.1016/j.jaci.2014.05.026. [DOI] [PubMed] [Google Scholar]
- 139.Daar ES, Moudgil T, Meyer RD, Ho DD. Transient high levels of viremia in patients with primary human immunodeficiency virus type 1 infection. N Engl J Med. 1991;324:961–4. doi: 10.1056/NEJM199104043241405. [DOI] [PubMed] [Google Scholar]
- 140.Perreau M, Levy Y, Pantaleo G. Immune response to HIV. Curr Opin HIV AIDS. 2013;8:333–40. doi: 10.1097/COH.0b013e328361faf4. [DOI] [PubMed] [Google Scholar]
- 141.Metzner KJ. Pathogenesis of HIV-1. Ther Umsch. 2014;71:443–50. doi: 10.1024/0040-5930/a000536. [DOI] [PubMed] [Google Scholar]
- 142.Grossman Z, Meier-Schellersheim M, Paul WE, Picker LJ. Pathogenesis of HIV infection: What the virus spares is as important as what it destroys. Nat Med. 2006;12:289–95. doi: 10.1038/nm1380. [DOI] [PubMed] [Google Scholar]
- 143.Murray CJ, Ortblad KF, Guinovart C, Lim SS, Wolock TM, Roberts DA, Dansereau EA, Graetz N, Barber RM, Brown JC, Wang H, Duber HC, Naghavi M, Dicker D, Dandona L, Salomon JA, Heuton KR, Foreman K, Phillips DE, Fleming TD, Flaxman AD, Phillips BK, Johnson EK, Coggeshall MS, Abd-Allah F, Abera SF, Abraham JP, Abubakar I, Abu-Raddad LJ, Abu-Rmeileh NM, Achoki T, Adeyemo AO, Adou AK, Adsuar JC, Agardh EE, Akena D, Al Kahbouri MJ, Alasfoor D, Albittar MI, Alcalá-Cerra G, Alegretti MA, Alemu ZA, Alfonso-Cristancho R, Alhabib S, Ali R, Alla F, Allen PJ, Alsharif U, Alvarez E, Alvis-Guzman N, Amankwaa AA, Amare AT, Amini H, Ammar W, Anderson BO, Antonio CA, Anwari P, Arnlöv J, Arsenijevic VS, Artaman A, Asghar RJ, Assadi R, Atkins LS, Badawi A, Balakrishnan K, Banerjee A, Basu S, Beardsley J, Bekele T, Bell ML, Bernabe E, Beyene TJ, Bhala N, Bhalla A, Bhutta ZA, Abdulhak AB, Binagwaho A, Blore JD, Basara BB, Bose D, Brainin M, Breitborde N, Castañeda-Orjuela CA, Catalá-López F, Chadha VK, Chang JC, Chiang PP, Chuang TW, Colomar M, Cooper LT, Cooper C, Courville KJ, Cowie BC, Criqui MH, Dandona R, Dayama A, De Leo D, Degenhardt L, Del Pozo-Cruz B, Deribe K, Des Jarlais DC, Dessalegn M, Dharmaratne SD, Dilmen U, Ding EL, Driscoll TR, Durrani AM, Ellenbogen RG, Ermakov SP, Esteghamati A, Faraon EJ, Farzadfar F, Fereshtehnejad SM, Fijabi DO, Forouzanfar MH, Fra Paleo U, Gaffikin L, Gamkrelidze A, Gankpé FG, Geleijnse JM, Gessner BD, Gibney KB, Ginawi IA, Glaser EL, Gona P, Goto A, Gouda HN, Gugnani HC, Gupta R, Gupta R, Hafezi-Nejad N, Hamadeh RR, Hammami M, Hankey GJ, Harb HL, Haro JM, Havmoeller R, Hay SI, Hedayati MT, Pi IB, Hoek HW, Hornberger JC, Hosgood HD, Hotez PJ, Hoy DG, Huang JJ, Iburg KM, Idrisov BT, Innos K, Jacobsen KH, Jeemon P, Jensen PN, Jha V, Jiang G, Jonas JB, Juel K, Kan H, Kankindi I, Karam NE, Karch A, Karema CK, Kaul A, Kawakami N, Kazi DS, Kemp AH, Kengne AP, Keren A, Kereselidze M, Khader YS, Khalifa SE, Khan EA, Khang YH, Khonelidze I, Kinfu Y, Kinge JM, Knibbs L, Kokubo Y, Kosen S, Defo BK, Kulkarni VS, Kulkarni C, Kumar K, Kumar RB, Kumar GA, Kwan GF, Lai T, Balaji AL, Lam H, Lan Q, Lansingh VC, Larson HJ, Larsson A, Lee JT, Leigh J, Leinsalu M, Leung R, Li Y, Li Y, De Lima GM, Lin HH, Lipshultz SE, Liu S, Liu Y, Lloyd BK, Lotufo PA, Machado VM, Maclachlan JH, Magis-Rodriguez C, Majdan M, Mapoma CC, Marcenes W, Marzan MB, Masci JR, Mashal MT, Mason-Jones AJ, Mayosi BM, Mazorodze TT, Mckay AC, Meaney PA, Mehndiratta MM, Mejia-Rodriguez F, Melaku YA, Memish ZA, Mendoza W, Miller TR, Mills EJ, Mohammad KA, Mokdad AH, Mola GL, Monasta L, Montico M, Moore AR, Mori R, Moturi WN, Mukaigawara M, Murthy KS, Naheed A, Naidoo KS, Naldi L, Nangia V, Narayan KM, Nash D, Nejjari C, Nelson RG, Neupane SP, Newton CR, Ng M, Nisar MI, Nolte S, Norheim OF, Nowaseb V, Nyakarahuka L, Oh IH, Ohkubo T, Olusanya BO, Omer SB, Opio JN, Orisakwe OE, Pandian JD, Papachristou C, Caicedo AJ, Patten SB, Paul VK, Pavlin BI, Pearce N, Pereira DM, Pervaiz A, Pesudovs K, Petzold M, Pourmalek F, Qato D, Quezada AD, Quistberg DA, Rafay A, Rahimi K, Rahimi-Movaghar V, Ur Rahman S, Raju M, Rana SM, Razavi H, Reilly RQ, Remuzzi G, Richardus JH, Ronfani L, Roy N, Sabin N, Saeedi MY, Sahraian MA, Samonte GM, Sawhney M, Schneider IJ, Schwebel DC, Seedat S, Sepanlou SG, Servan-Mori EE, Sheikhbahaei S, Shibuya K, Shin HH, Shiue I, Shivakoti R, Sigfusdottir ID, Silberberg DH, Silva AP, Simard EP, Singh JA, Skirbekk V, Sliwa K, Soneji S, Soshnikov SS, Sreeramareddy CT, Stathopoulou VK, Stroumpoulis K, Swaminathan S, Sykes BL, Tabb KM, Talongwa RT, Tenkorang EY, Terkawi AS, Thomson AJ, Thorne-Lyman AL, Towbin JA, Traebert J, Tran BX, Dimbuene ZT, Tsilimbaris M, Uchendu US, Ukwaja KN, Uzun SB, Vallely AJ, Vasankari TJ, Venketasubramanian N, Violante FS, Vlassov VV, Vollset SE, Waller S, Wallin MT, Wang L, Wang X, Wang Y, Weichenthal S, Weiderpass E, Weintraub RG, Westerman R, White RA, Wilkinson JD, Williams TN, Woldeyohannes SM, Wong JQ, Xu G, Yang YC, Yano Y, Yentur GK, Yip P, Yonemoto N, Yoon SJ, Younis M, Yu C, Jin KY, El Sayed Zaki M, Zhao Y, Zheng Y, Zhou M, Zhu J, Zou XN, Lopez AD, Vos T. Global, regional, and national incidence and mortality for HIV, tuberculosis, and malaria during 1990–2013: A systematic analysis for the Global Burden of Disease Study 2013. Lancet. 2014;384:1005–70. doi: 10.1016/S0140-6736(14)60844-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Fanales-Belasio E, Raimondo M, Suligoi B, Butto S. HIV virology and pathogenetic mechanisms of infection: A brief overview. Ann Ist Super Sanita. 2010;46:5–14. doi: 10.4415/ANN_10_01_02. [DOI] [PubMed] [Google Scholar]
- 145.Zhao CC, Gao XQ, Xue J, Cong Z, Zhang WL, Chen T, Zhao CC, Gao XQ, Xue J, Cong Z, Zhang WL, Chen T. Interleukin-21 up-regulates interleukin-21R expression and interferon γ production by CD8+ cells in SHIV-infected macaques. Exp Biol Med (Maywood) 2013;238:400–9. doi: 10.1177/1535370213477978. [DOI] [PubMed] [Google Scholar]
- 146.Sarkar R, Mitra D, Chakrabarti S. HIV-1 gp120 protein downregulates Nef induced IL-6 release in immature dentritic cells through interplay of DC-SIGN. PLoS One. 2013;8:e59073. doi: 10.1371/journal.pone.0059073. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Stacey AR, Norris PJ, Qin L, Haygreen EA, Taylor E, Heitman J, Lebedeva M, DeCamp A, Li D, Grove D, Self SG, Borrow P. Induction of a striking systemic cytokine cascade prior to peak viremia in acute human immunodeficiency virus type 1 infection, in contrast to more modest and delayed responses in acute hepatitis B and C virus infections. J Virol. 2009;83:3719–33. doi: 10.1128/JVI.01844-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Berkowitz RD, Beckerman KP, Schall TJ, McCune JM. CXCR4 and CCR5 expression delineates targets for HIV-1 disruption of T cell differentiation. J Immunol. 1998;161:3702–10. [PubMed] [Google Scholar]
- 149.Meissner EG, Duus KM, Gao F, Yu XF, Su L. Characterization of a thymus-tropic HIV-1 isolate from a rapid progressor: Role of the envelope. Virology. 2004;328:74–88. doi: 10.1016/j.virol.2004.07.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Papiernik M, Brossard Y, Mulliez N, Roume J, Brechot C, Barin F, Goudeau A, Bach JF, Griscelli C, Henrion R. Thymic abnormalities in fetuses aborted from human immunodeficiency virus type 1 seropositive women. Pediatrics. 1992;89:297–301. [PubMed] [Google Scholar]
- 151.Grody WW, Fligiel S, Naeim F. Thymus involution in the acquired immunodeficiency syndrome. Am J Clin Pathol. 1985;84:85–95. doi: 10.1093/ajcp/84.1.85. [DOI] [PubMed] [Google Scholar]
- 152.Zaitseva MB, Lee S, Rabin RL, Tiffany HL, Farber JM, Peden KW, Murphy PM, Golding H. CXCR4 and CCR5 on human thymocytes: Biological function and role in HIV-1 infection. J Immunol. 1998;161:3103–13. [PubMed] [Google Scholar]
- 153.Resop RS, Douaisi M, Craft J, Uittenbogaart CH. Preferential infection of regulatory T cells by R5 vs. X4 HIV-1. Midwinter Conference of Immunologists, Conference Abstract; Asilomar, CA. 2011. [Google Scholar]
- 154.Kolte L. Thymic function in HIV-infection. Dan Med J. 2013;60:B4622. [PubMed] [Google Scholar]
- 155.Geleziunas R, Bour S, Wainberg MA. Cell surface down-modulation of CD4 after infection by HIV-1. FASEB J. 1994;8:593–600. doi: 10.1096/fasebj.8.9.8005387. [DOI] [PubMed] [Google Scholar]
- 156.Cummins NW, Badley AD. Mechanisms of HIV-associated lymphocyte apoptosis: 2010. Cell Death Dis. 2010;1:e99. doi: 10.1038/cddis.2010.77. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157.Zhou H, Zhao H, Hao Y, Song C, Han J, Zhang J, Gao G, Han N, Yang D, Li Y, Zhang F, Zeng H. Excessive conversion and impaired thymic output contribute to disturbed regulatory T-cell homeostasis in AIDS patients with low CD4 cell counts. AIDS. 2013;27:1059–69. doi: 10.1097/QAD.0b013e32835e2b99. [DOI] [PubMed] [Google Scholar]
- 158.Gurney KB, Colantonio AD, Blom B, Spits H, Uittenbogaart CH. Endogenous IFN-α production by plasmacytoid dendritic cells exerts an antiviral effect on thymic HIV-1 infection. J Immunol. 2004;173:7269–76. doi: 10.4049/jimmunol.173.12.7269. [DOI] [PubMed] [Google Scholar]
- 159.Dutrieux J, Fabre-Mersseman V, Charmeteau-De Muylder B, Rancez M, Ponte R, Rozlan S, Figueiredo-Morgado S, Bernard A, Beq S, Couëdel-Courteille A, Cheynier R. Modified interferon-α subtypes production and chemokine networks in the thymus during acute simian immunodeficiency virus infection, impact on thymopoiesis. AIDS. 2014;28:1101–13. doi: 10.1097/QAD.0000000000000249. [DOI] [PubMed] [Google Scholar]
- 160.Beq S, Nugeyre MT, Ho Tsong Fang R, Gautier D, Legrand R, Schmitt N, Estaquier J, Barré-Sinoussi F, Hurtrel B, Cheynier R, Israël N. IL-7 induces immunological improvement in SIV-infected rhesus macaques under antiviral therapy. J Immunol. 2006;176:914–22. doi: 10.4049/jimmunol.176.2.914. [DOI] [PubMed] [Google Scholar]
- 161.Sopper S, Nierwetberg D, Halbach A, Sauer U, Scheller C, Stahl-Hennig C, Mätz-Rensing K, Schäfer F, Schneider T, ter Meulen V, Müller JG. Impact of simian immunodeficiency virus (SIV) infection on lymphocyte numbers and T-cell turnover in different organs of rhesus monkeys. Blood. 2003;101:1213–9. doi: 10.1182/blood-2002-06-1644. [DOI] [PubMed] [Google Scholar]
- 162.Ho Tsong Fang R, Colantonio AD, Uittenbogaart CH. The role of the thymus in HIV infection: A 10 year perspective. AIDS. 2008;22:171–84. doi: 10.1097/QAD.0b013e3282f2589b. [DOI] [PubMed] [Google Scholar]
- 163.Ho Tsong Fang R, Khatissian E, Monceaux V, Cumont MC, Beq S, Ameisen JC, Aubertin AM, Israel N, Estaquier J, Hurtel B. Disease progression in macaques with low SIV replication levels: On the relevance of TREC counts. AIDS. 2008;22:171–84. doi: 10.1097/01.aids.0000166089.93574.5a. [DOI] [PubMed] [Google Scholar]
- 164.Dion ML, Poulin JF, Bordi R, Sylvestre M, Corsini R, Kettaf N, Dalloul A, Boulassel MR, Debré P, Routy JP, Grossman Z, Sékaly RP, Cheynier R. HIV infection rapidly induces and maintains a substantial suppression of thymocyte proliferation. Immunity. 2004;21:757–68. doi: 10.1016/j.immuni.2004.10.013. [DOI] [PubMed] [Google Scholar]
- 165.Douek DC, McFarland RD, Keiser PH, Gage EA, Massey JM, Haynes BF, Polis MA, Haase AT, Feinberg MB, Sullivan JL, Jamieson BD, Zack JA, Picker LJ, Koup RA. Changes in thymic function with age and during the treatment of HIV infection. Nature. 1998;396:690–5. doi: 10.1038/25374. [DOI] [PubMed] [Google Scholar]
- 166.Douek DC, Betts MR, Hill BJ, Little SJ, Lempicki R, Metcalf JA, Casazza J, Yoder C, Adelsberger JW, Stevens RA, Baseler MW, Keiser P, Richman DD, Davey RT, Koup RA. Evidence for increased T cell turnover and decreased thymic output in HIV infection. J Immunol. 2001;167:6663–8. doi: 10.4049/jimmunol.167.11.6663. [DOI] [PubMed] [Google Scholar]
- 167.Aiuti F, Mezzaroma I. Failure to reconstitute CD4+ T-cells despite suppression of HIV replication under HAART. AIDS Rev. 2006;8:88–97. [PubMed] [Google Scholar]




