1
|
Abstract
Historically, the immune system was believed to develop along a linear axis of maturity from fetal life to adulthood. Now, it is clear that distinct layers of immune cells are generated from unique waves of hematopoietic progenitors during different windows of development. This model, known as the layered immune model, has provided a useful framework for understanding why distinct lineages of B cells and γδ T cells arise in succession and display unique functions in adulthood. However, the layered immune model has not been applied to CD8+ T cells, which are still often viewed as a uniform population of cells belonging to the same lineage, with functional differences between cells arising from environmental factors encountered during infection. Recent studies have challenged this idea, demonstrating that not all CD8+ T cells are created equally and that the functions of individual CD8+ T cells in adults are linked to when they were created in the host. In this review, we discuss the accumulating evidence suggesting there are distinct ontogenetic subpopulations of CD8+ T cells and propose that the layered immune model be extended to the CD8+ T cell compartment.
Collapse
Affiliation(s)
- Cybelle Tabilas
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY 14853, USA
- Co-first author
| | - Norah L. Smith
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY 14853, USA
- Co-first author
| | - Brian D. Rudd
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY 14853, USA
| |
Collapse
|
2
|
Ng MSF, Roth TL, Mendoza VF, Marson A, Burt TD. Helios enhances the preferential differentiation of human fetal CD4 + naïve T cells into regulatory T cells. Sci Immunol 2020; 4:4/41/eaav5947. [PMID: 31757834 DOI: 10.1126/sciimmunol.aav5947] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Accepted: 10/24/2019] [Indexed: 12/14/2022]
Abstract
T cell receptor (TCR) stimulation and cytokine cues drive the differentiation of CD4+ naïve T cells into effector T cell populations with distinct proinflammatory or regulatory functions. Unlike adult naïve T cells, human fetal naïve CD4+ T cells preferentially differentiate into FOXP3+ regulatory T (Treg) cells upon TCR activation independent of exogenous cytokine signaling. This cell-intrinsic predisposition for Treg differentiation is implicated in the generation of tolerance in utero; however, the underlying mechanisms remain largely unknown. Here, we identify epigenetic and transcriptional programs shared between fetal naïve T and committed Treg cells that are inactive in adult naïve T cells and show that fetal-derived induced Treg (iTreg) cells retain this transcriptional program. We show that a subset of Treg-specific enhancers is accessible in fetal naïve T cells, including two active superenhancers at Helios Helios is expressed in fetal naïve T cells but not in adult naïve T cells, and fetal iTreg cells maintain Helios expression. CRISPR-Cas9 ablation of Helios in fetal naïve T cells impaired their differentiation into iTreg cells upon TCR stimulation, reduced expression of immunosuppressive genes in fetal iTreg cells such as IL10, and increased expression of proinflammatory genes including IFNG Consequently, Helios knockout fetal iTreg cells had reduced IL-10 and increased IFN-γ cytokine production. Together, our results reveal important roles for Helios in enhancing preferential fetal Treg differentiation and fine-tuning eventual Treg function. The Treg-biased programs identified within fetal naïve T cells could potentially be used to engineer enhanced iTreg populations for adoptive cellular therapies.
Collapse
Affiliation(s)
- Melissa S F Ng
- Biomedical Sciences Graduate Program, University of California, San Francisco (UCSF), San Francisco, CA 94143, USA.,Singapore Immunology Network, Agency for Science, Technology and Research, Biopolis, Singapore 138648, Singapore
| | - Theodore L Roth
- Biomedical Sciences Graduate Program, University of California, San Francisco (UCSF), San Francisco, CA 94143, USA.,Department of Microbiology and Immunology, UCSF, San Francisco, CA 94143, USA.,Diabetes Center, UCSF, San Francisco, CA 94143, USA
| | - Ventura F Mendoza
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, UCSF, San Francisco, CA 94143, USA
| | - Alexander Marson
- Department of Microbiology and Immunology, UCSF, San Francisco, CA 94143, USA.,Diabetes Center, UCSF, San Francisco, CA 94143, USA.,Innovative Genomics Institute, University of California, Berkeley, CA 94720, USA.,Department of Medicine, UCSF, San Francisco, CA 94143, USA.,Chan Zuckerberg Biohub, San Francisco, CA 94158, USA.,UCSF Helen Diller Family Comprehensive Cancer Center, UCSF, San Francisco, CA 94158, USA.,Parker Institute for Cancer Immunotherapy, San Francisco, CA 94129, USA
| | - Trevor D Burt
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, UCSF, San Francisco, CA 94143, USA. .,Department of Pediatrics, Division of Neonatology, UCSF, San Francisco, CA 94110, USA
| |
Collapse
|
3
|
Khosravi-Maharlooei M, Hoelzl M, Li HW, Madley RC, Waffarn EE, Danzl NM, Sykes M. Rapid thymectomy of NSG mice to analyze the role of native and grafted thymi in humanized mice. Eur J Immunol 2019; 50:138-141. [PMID: 31583677 DOI: 10.1002/eji.201948205] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Revised: 07/30/2019] [Accepted: 10/03/2019] [Indexed: 01/08/2023]
Abstract
We developed a rapid method to remove the native mouse thymus from NSG mice, which allowed us to compare the behavior of human immune cells in the presence or absence of human T cells in human immune system mice. Removing the native mouse thymus is critical for studies of human thymopiesis in grafted thymic tissue in humanized mice.
Collapse
Affiliation(s)
- Mohsen Khosravi-Maharlooei
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Medical Center, New York, NY, USA
| | - Markus Hoelzl
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Medical Center, New York, NY, USA
| | - Hao Wei Li
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Medical Center, New York, NY, USA
| | - Rachel C Madley
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Medical Center, New York, NY, USA.,Department of Microbiology & Immunology, Columbia University Medical Center, Columbia University, New York, NY, USA
| | - Elizabeth E Waffarn
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Medical Center, New York, NY, USA
| | - Nichole M Danzl
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Medical Center, New York, NY, USA
| | - Megan Sykes
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Medical Center, New York, NY, USA.,Department of Microbiology & Immunology, Columbia University Medical Center, Columbia University, New York, NY, USA.,Department of Surgery, Columbia University Medical Center, Columbia University, New York, NY, USA
| |
Collapse
|
4
|
Dong M, Artusa P, Kelly SA, Fournier M, Baldwin TA, Mandl JN, Melichar HJ. Alterations in the Thymic Selection Threshold Skew the Self-Reactivity of the TCR Repertoire in Neonates. THE JOURNAL OF IMMUNOLOGY 2017; 199:965-973. [PMID: 28659353 DOI: 10.4049/jimmunol.1602137] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Accepted: 06/02/2017] [Indexed: 12/19/2022]
Abstract
Neonatal and adult T cells differ in their effector functions. Although it is known that cell-intrinsic differences in mature T cells contribute to this phenomenon, the factors involved remain unclear. Given emerging evidence that the binding strength of a TCR for self-peptide presented by MHC (self-pMHC) impacts T cell function, we sought to determine whether altered thymic selection influences the self-reactivity of the TCR repertoire during ontogeny. We found that conventional and regulatory T cell subsets in the thymus of neonates and young mice expressed higher levels of cell surface CD5, a surrogate marker for TCR avidity for self-pMHC, as compared with their adult counterparts, and this difference in self-reactivity was independent of the germline bias of the neonatal TCR repertoire. The increased binding strength of the TCR repertoire for self-pMHC in neonates was not solely due to reported defects in clonal deletion. Rather, our data suggest that thymic selection is altered in young mice such that thymocytes bearing TCRs with low affinity for self-peptide are not efficiently selected into the neonatal repertoire, and stronger TCR signals accompany both conventional and regulatory T cell selection. Importantly, the distinct levels of T cell self-reactivity reflect physiologically relevant differences based on the preferential expansion of T cells from young mice to fill a lymphopenic environment. Therefore, differences in thymic selection in young versus adult mice skew the TCR repertoire, and the relatively higher self-reactivity of the T cell pool may contribute to the distinct immune responses observed in neonates.
Collapse
Affiliation(s)
- Mengqi Dong
- Maisonneuve-Rosemont Hospital Research Center, Montreal, Quebec H1T 2M4, Canada.,Department of Microbiology, Infectious Diseases, and Immunology, University of Montreal, Montreal, Quebec H3T 1J4, Canada
| | - Patricio Artusa
- Department of Physiology and McGill Research Centre for Complex Traits, McGill University, Montreal, Quebec H3G 1Y6, Canada
| | - Stephanie A Kelly
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, Alberta T6G 2E1, Canada; and
| | - Marilaine Fournier
- Maisonneuve-Rosemont Hospital Research Center, Montreal, Quebec H1T 2M4, Canada
| | - Troy A Baldwin
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, Alberta T6G 2E1, Canada; and
| | - Judith N Mandl
- Department of Physiology and McGill Research Centre for Complex Traits, McGill University, Montreal, Quebec H3G 1Y6, Canada
| | - Heather J Melichar
- Maisonneuve-Rosemont Hospital Research Center, Montreal, Quebec H1T 2M4, Canada; .,Department of Medicine, University of Montreal, Montreal, Quebec H3T 1J4, Canada
| |
Collapse
|
5
|
Petersen F, Yue X, Riemekasten G, Yu X. Dysregulated homeostasis of target tissues or autoantigens - A novel principle in autoimmunity. Autoimmun Rev 2017; 16:602-611. [PMID: 28411168 DOI: 10.1016/j.autrev.2017.04.006] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2017] [Accepted: 03/11/2017] [Indexed: 01/22/2023]
Abstract
Monogenic autoimmune disorders provide a powerful tool for our understanding of the principles of autoimmunity due to the obvious impact of a single gene on the disease. So far, approximately 100 single gene defects causing murine monogenic autoimmune disorders have been reported and the functional characterization of these genes will provide significant progress in understanding the nature of autoimmunity. According to their function, genes leading to monogenic autoimmune disorders can be categorized into two groups. An expectable first group contains genes involved in the homeostasis of the immune system, including homeostasis of immune organs and immune cells. Intriguingly, the second group consists of genes functionally involved in the homeostasis of target tissues or autoantigens. According to our novel hypothesis, we propose that autoimmunity represents a consequence of a dysregulated homeostasis of the immune system and/or its targets including autoantigens and target tissues. In this review we refer to both aspects of homeostasis in autoimmunity with a highlight on the role of the homeostasis of target tissues and autoantigens.
Collapse
Affiliation(s)
- Frank Petersen
- Priority Area Asthma & Allergy, Research Center Borstel, Airway Research Center North (ARCN), Members of the German Center for Lung Research (DZL), 23845 Borstel, Germany
| | - Xiaoyang Yue
- Priority Area Asthma & Allergy, Research Center Borstel, Airway Research Center North (ARCN), Members of the German Center for Lung Research (DZL), 23845 Borstel, Germany
| | - Gabriela Riemekasten
- Priority Area Asthma & Allergy, Research Center Borstel, Airway Research Center North (ARCN), Members of the German Center for Lung Research (DZL), 23845 Borstel, Germany; Department of Rheumatology, University of Lübeck, 23538 Lübeck, Germany
| | - Xinhua Yu
- Priority Area Asthma & Allergy, Research Center Borstel, Airway Research Center North (ARCN), Members of the German Center for Lung Research (DZL), 23845 Borstel, Germany; Xiamen-Borstel Joint Laboratory of Autoimmunity, Medical College of Xiamen University, Xiamen 361102, China.
| |
Collapse
|
6
|
O'Hagan KL, Choi J, Pryshchep O, Chernoff J, Phee H. Pak2 Links TCR Signaling Strength to the Development of Regulatory T Cells and Maintains Peripheral Tolerance. THE JOURNAL OF IMMUNOLOGY 2015; 195:1564-77. [PMID: 26157175 DOI: 10.4049/jimmunol.1500843] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Accepted: 06/10/2015] [Indexed: 01/01/2023]
Abstract
Although significant effort has been devoted to understanding the thymic development of Foxp3(+) regulatory T cells (Tregs), the precise signaling pathways that govern their lineage commitment still remain enigmatic. Our findings show a novel role for the actin cytoskeletal remodeling protein, p21-activated kinase 2 (Pak2), in Treg development and homeostasis. The absence of Pak2 in T cells resulted in a marked reduction in both thymus- and peripherally derived Tregs, accompanied by the development of spontaneous colitis in Pak2-deficient mice. Additionally, Pak2 was required for the proper differentiation of in vitro-induced Tregs as well as maintenance of Tregs. Interestingly, Pak2 was necessary for generating the high-affinity TCR- and IL-2-mediated signals that are required by developing Tregs for their lineage commitment. These findings provide novel insight into how developing thymocytes translate lineage-specific high-affinity TCR signals to adopt the Treg fate, and they further posit Pak2 as an essential regulator for this process.
Collapse
Affiliation(s)
- Kyle Leonard O'Hagan
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611; and
| | - Jinyong Choi
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611; and
| | - Olga Pryshchep
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611; and
| | - Jonathan Chernoff
- Cancer Biology Program, Fox Chase Cancer Center, Philadelphia, PA 19111
| | - Hyewon Phee
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611; and
| |
Collapse
|
7
|
Santori FR. The immune system as a self-centered network of lymphocytes. Immunol Lett 2015; 166:109-16. [PMID: 26092524 DOI: 10.1016/j.imlet.2015.06.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2015] [Accepted: 06/04/2015] [Indexed: 12/13/2022]
Abstract
This essay makes a brief historical and comparative review of selective and network theories of the immune system which is presented as a chemical sensory system with immune and non-immune functions. The ontogeny of immune networks is the result of both positive and negative selection of lymphocytes to self-epitopes that serve as a "template" for the recognition of foreign antigens. The development of immune networks progresses from single individual clones in early ontogeny into complex "information processing networks" in which lymphocytes are linked to inhibitory and stimulatory immune cells. The results of these regulatory interactions modulate immune responses and tolerance.
Collapse
Affiliation(s)
- Fabio R Santori
- The Kimmel Center for Biology and Medicine of the Skirball Institute, New York University School of Medicine, New York, NY 10016, USA
| |
Collapse
|
8
|
Zheng P, Chang X, Lu Q, Liu Y. Cytopenia and autoimmune diseases: a vicious cycle fueled by mTOR dysregulation in hematopoietic stem cells. J Autoimmun 2013; 41:182-7. [PMID: 23375848 DOI: 10.1016/j.jaut.2012.12.011] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2012] [Accepted: 12/13/2012] [Indexed: 12/24/2022]
Abstract
A long-standing but poorly understood defect in autoimmune diseases is dysfunction of the hematopoietic cells. Leukopenia is often associated with systemic lupus erythematous (SLE) and other autoimmune diseases. In addition, homeostatic proliferation of T cells, which is a host response to T-cell lymphopenia, has been implicated as potential cause of rheumatoid arthritis (RA) in human and experimental models of autoimmune diabetes in the NOD mice and the BB rats. Conversely, successful treatments of aplastic anemia by immune suppression suggest that the hematologic abnormality may have a root in autoimmune diseases. Traditionally, the link between autoimmune diseases and defects in hematopoietic cells has been viewed from the prism of antibody-mediated hemolytic cytopenia. While autoimmune destruction may well be part of pathogenesis of defects in hematopoietic system, it is worth considering the hypothesis that either leukopenia or pancytopenia may also result directly from defective hematopoietic stem cells (HSC). We have recently tested this hypothesis in the autoimmune Scurfy mice which has mutation Foxp3, the master regulator of regulatory T cells. Our data demonstrated that due to hyperactivation of mTOR, the HSC in the Scurfy mice are extremely poor in hematopoiesis. Moreover, rapamycin, an mTOR inhibitor rescued HSC defects and prolonged survival of the Scurfy mice. Our data raised the intriguing possibility that targeting mTOR dysregulation in the HSC may help to break the vicious cycle between cytopenia and autoimmune diseases.
Collapse
Affiliation(s)
- Pan Zheng
- Department of Surgery, University of Michigan, School of Medicine, Ann Arbor, MI 48109, USA.
| | | | | | | |
Collapse
|
9
|
del Rio R, Sun Y, Alard P, Tung KSK, Teuscher C. H2 control of natural T regulatory cell frequency in the lymph node correlates with susceptibility to day 3 thymectomy-induced autoimmune disease. THE JOURNAL OF IMMUNOLOGY 2010; 186:382-9. [PMID: 21135167 DOI: 10.4049/jimmunol.1002110] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Day 3 thymectomy (D3Tx) results in a loss of peripheral tolerance mediated by natural regulatory T cells (nTregs) and development of autoimmune ovarian dysgenesis (AOD) and autoimmune dacryoadenitis (ADA) in A/J and (C57BL/6J × A/J) F(1) hybrids (B6A), but not in C57BL/6J (B6) mice. Previously, using quantitative trait locus (QTL) linkage analysis, we showed that D3Tx-AOD is controlled by five unlinked QTL (Aod1-Aod5) and H2. In this study, using D3Tx B6-Chr(A/J)/NaJ chromosome (Chr) substitution strains, we confirm that QTL on Chr16 (Aod1a/Aod1b), Chr3 (Aod2), Chr1 (Aod3), Chr2 (Aod4), Chr7 (Aod5), and Chr17 (H2) control D3Tx-AOD susceptibility. In addition, we also present data mapping QTL controlling D3Tx-ADA to Chr17 (Ada1/H2), Chr1 (Ada2), and Chr3 (Ada3). Importantly, B6-ChrX(A/J) mice were as resistant to D3Tx-AOD and D3Tx-ADA as B6 mice, thereby excluding Foxp3 as a susceptibility gene in these models. Moreover, we report quantitative differences in the frequency of nTregs in the lymph nodes (LNs), but not spleen or thymus, of AOD/ADA-resistant B6 and AOD/ADA-susceptible A/J, B6A, and B6-Chr17(A/J) mice. Similar results correlating with experimental allergic encephalomyelitis and orchitis susceptibility were seen with B10.S and SJL/J mice. Using H2-congenic mice, we show that the observed difference in frequency of LN nTregs is controlled by Ada1/H2. These data support the existence of an LN-specific, H2-controlled mechanism regulating the prevalence of nTregs in autoimmune disease susceptibility.
Collapse
Affiliation(s)
- Roxana del Rio
- Department of Medicine, University of Vermont, Burlington, VT 05405, USA
| | | | | | | | | |
Collapse
|
10
|
Regulatory T cells: The suppressor arm of the immune system. Autoimmun Rev 2010; 10:112-5. [DOI: 10.1016/j.autrev.2010.08.013] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2010] [Accepted: 08/19/2010] [Indexed: 12/20/2022]
|
11
|
Case LK, Del Rio R, Bonney EA, Zachary JF, Blankenhorn EP, Tung KSK, Teuscher C. The postnatal maternal environment affects autoimmune disease susceptibility in A/J mice. Cell Immunol 2010; 260:119-27. [PMID: 19914609 DOI: 10.1016/j.cellimm.2009.10.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2009] [Revised: 10/06/2009] [Accepted: 10/07/2009] [Indexed: 01/25/2023]
Abstract
The postnatal maternal environment is known to increase susceptibility to a number of autoimmune diseases. Here we asked whether the postnatal maternal environment could influence autoimmune disease development to day 3 thymectomy (d3tx)-induced autoimmune ovarian disease (AOD) and experimental allergic encephalomyelitis (EAE) in cross-fostered A/J and B6 mice. A/J pups foster-nursed by B6 mothers exhibit an increase in autoimmune disease development while cross-fostering B6 pups on A/J mothers did not alter their susceptibility. The increase in AOD incidence seen in foster-nursed d3tx A/J mice correlated with a decrease in the total number of CD4(+) T cells in the lymph nodes of these animals. Analysis of the cellular composition in the milk revealed that B6 mice shed significantly more maternally derived lymphocytes into their milk compared to A/J mothers. These data suggest that there are maternally derived postnatal factors that influence the development of autoimmune disease in A/J mice.
Collapse
Affiliation(s)
- Laure K Case
- Department of Medicine, University of Vermont, Burlington, VT 05405, USA
| | | | | | | | | | | | | |
Collapse
|
12
|
Gagnerault MC, Lanvin O, Pasquier V, Garcia C, Damotte D, Lucas B, Lepault F. Autoimmunity during Thymectomy-Induced Lymphopenia: Role of Thymus Ablation and Initial Effector T Cell Activation Timing in Nonobese Diabetic Mice. THE JOURNAL OF IMMUNOLOGY 2009; 183:4913-20. [DOI: 10.4049/jimmunol.0901954] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
13
|
Abstract
BACKGROUND Ovarian cancer is frequently diagnosed at an advanced stage, and although initially responsive to surgery and chemotherapy, has a high rate of recurrence and mortality. Cellular immunotherapy may offer the prospect of treatment to prevent or delay recurrent metastatic disease. OBJECTIVE To provide an overview of current innovations in cellular immunotherapy for ovarian cancer, with an emphasis on dendritic cell vaccination and adoptive T-cell immunotherapy. METHODS Three key areas are explored in this review: first, an appraisal of the current state of the art of cellular immunotherapy for treatment of ovarian cancer; second, a discussion of the immunological defenses erected by ovarian cancer to prevent immunological attack, with an emphasis on the role of tumor-associated regulatory T cells; and third, an exploration of innovative techniques that may enhance the ability of cellular immunotherapy to overcome ovarian tumor-associated immune suppression. RESULTS/CONCLUSION Ovarian cancer is recognized as a paradigm for tumor-associated immune suppression. Innovative approaches for antagonism of tumor-associated regulatory T-cell infiltration and redirection of self antigen-driven regulatory T-cell activation may provide the key to development of future strategies for cellular immunotherapy against ovarian cancer.
Collapse
Affiliation(s)
- Martin J Cannon
- University of Arkansas for Medical Sciences, Department of Microbiology and Immunology, 4301 West Markham, Little Rock, AR 72205, USA.
| | | |
Collapse
|
14
|
Ahmed M, Lanzer KG, Yager EJ, Adams PS, Johnson LL, Blackman MA. Clonal expansions and loss of receptor diversity in the naive CD8 T cell repertoire of aged mice. THE JOURNAL OF IMMUNOLOGY 2009; 182:784-92. [PMID: 19124721 DOI: 10.4049/jimmunol.182.2.784] [Citation(s) in RCA: 87] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
There are well-characterized age-related changes in the peripheral repertoire of CD8 T cells characterized by reductions in the ratio of naive:memory T cells and the development of large clonal expansions in the memory pool. In addition, the TCR repertoire of naive T cells is reduced with aging. Because a diverse repertoire of naive T cells is essential for a vigorous response to new infections and vaccinations, there is much interest in understanding the mechanisms responsible for declining repertoire diversity. It has been proposed that one reason for declining repertoire diversity in the naive T cell pool is an increasing dependence on homeostatic proliferation in the absence of new thymic emigrants for maintenance of the naive peripheral pool. In this study, we have analyzed the naive CD8 T cell repertoire in young and aged mice by DNA spectratype and sequence analysis. Our data show that naive T cells from aged mice have perturbed spectratype profiles compared with the normally Gaussian spectratype profiles characteristic of naive CD8 T cells from young mice. In addition, DNA sequence analysis formally demonstrated a loss of diversity associated with skewed spectratype profiles. Unexpectedly, we found multiple repeats of the same sequence in naive T cells from aged but not young mice, consistent with clonal expansions previously described only in the memory T cell pool. Clonal expansions among naive T cells suggests dysregulation in the normal homeostatic proliferative mechanisms that operate in young mice to maintain diversity in the naive T cell repertoire.
Collapse
|
15
|
Monteiro JP, Farache J, Mercadante AC, Mignaco JA, Bonamino M, Bonomo A. Pathogenic effector T cell enrichment overcomes regulatory T cell control and generates autoimmune gastritis. THE JOURNAL OF IMMUNOLOGY 2009; 181:5895-903. [PMID: 18941178 DOI: 10.4049/jimmunol.181.9.5895] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Regulatory T cells (Treg) deficiency leads to a severe, systemic, and lethal disease, as showed in immune dysregulation, polyendocrinopathy, enteropathy, X-linked syndrome patients, and scurfy mouse. Postneonatal thymectomy autoimmune gastritis has also been attributed to the absence of Tregs. In this case however, disease is mild, organ-specific, and, more important, it is not an obligatory outcome. We addressed this paradox comparing T cell compartments in gastritis-susceptible and resistant animals. We found that neonatal thymectomy-induced gastritis is not caused by the absence of Tregs. Instead of this, it is the presence of gastritogenic T cell clones that determines susceptibility to disease. The expansion of such clones under lymphopenic conditions results in a reduced Treg:effector T cell ratio that is not enough to control gastritis development. Finally, the presence of gastritogenic clones is determined by the amount of gastric Ag expressed in the neonatal thymus, emphasizing the importance of effector repertoire variability, present even in genetically identical subjects, to organ-specific autoimmune disease susceptibility.
Collapse
Affiliation(s)
- João P Monteiro
- Divisão de Medicina Experimental, Coordenação de Pesquisa, Instituto Nacional de Câncer, Rio de Janeiro, Brazil
| | | | | | | | | | | |
Collapse
|
16
|
Samy ET, Wheeler KM, Roper RJ, Teuscher C, Tung KSK. Cutting edge: Autoimmune disease in day 3 thymectomized mice is actively controlled by endogenous disease-specific regulatory T cells. THE JOURNAL OF IMMUNOLOGY 2008; 180:4366-70. [PMID: 18354156 DOI: 10.4049/jimmunol.180.7.4366] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Female B6AF1 mice thymectomized on day 3 (d3tx) develop autoimmune ovarian disease (AOD) and dacryoadenitis. It has been hypothesized that d3tx breaks tolerance by depleting late ontogeny regulatory T cells (Treg). We now report that Treg greatly expand over effector T cells in d3tx mice and adoptively suppress autoimmune disease in d3tx recipients. In the d3tx donors, Treg from ovarian lymph nodes (LN) preferentially suppress AOD and Treg from lacrimal gland LN preferentially suppress dacryoadenitis, suggesting they are strategically positioned for disease control. Indeed, the autologous disease in d3tx mice is dramatically enhanced by in vivo depletion of endogenous Treg. Moreover, normal 3-day-old mice possess Treg that suppress AOD and autoimmune gastritis as efficiently as adult cells. Thus, d3tx mice possess disease-relevant Treg of presumed neonatal origin. They accumulate in the regional LN and actively inhibit concurrent autoimmune disease; however, they cannot fully prevent autoimmune disease development.
Collapse
Affiliation(s)
- Eileen T Samy
- Department of Pathology, University of Virginia, Charlottesville, VA 22908, USA
| | | | | | | | | |
Collapse
|
17
|
|
18
|
Adkins B. Heterogeneity in the CD4 T Cell Compartment and the Variability of Neonatal Immune Responsiveness. ACTA ACUST UNITED AC 2007; 3:151-159. [PMID: 19122799 DOI: 10.2174/157339507781483496] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Over the past decade, it has become clear that T cell immune responses in both murine and human neonates are very heterogeneous, running the gamut from poor or deviant responsiveness to mature, adult-like inflammatory function. How this variability arises is not well understood but there is now a great deal of information suggesting that differences in the T cell compartments in neonates vs adults play important roles. A number of cell types or processes are qualitatively or quantitatively different in the neonate. These include (a) alternate epigenetic programs at the Th2 cytokine locus, (b) enhanced homeostatic proliferation, (c) a relative abundance of fetal-origin cells, (d) a greater representation of recent thymic emigrants, (e) high proportions of potentially self-reactive cells, (f) a developmental delay in the production of regulatory T cells, and (g) cells bearing TCR with limited N region diversity. Different conditions of antigen exposure may lead to different environmental signals that promote the selective responsiveness of one or more of these populations. Therefore, the variability of neonatal responses may be a function of the heterogeneous nature of the responding T cell population. In this review, we will describe these various subpopulations in detail and speculate as to the manner in which they could contribute to the heterogeneity of neonatal immune responses.
Collapse
Affiliation(s)
- Becky Adkins
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, Florida, USA
| |
Collapse
|
19
|
Liu Y, Zheng P. CD24: a genetic checkpoint in T cell homeostasis and autoimmune diseases. Trends Immunol 2007; 28:315-20. [PMID: 17531534 DOI: 10.1016/j.it.2007.05.001] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2007] [Revised: 04/17/2007] [Accepted: 05/09/2007] [Indexed: 10/23/2022]
Abstract
CD24 is widely used as a marker for differentiation of multiple lineages of cells and can provide costimulation for T cells, especially in non-lymphoid target organs. Recent studies demonstrate that CD24 controls an important genetic checkpoint for homeostasis and autoimmune diseases in both mice and humans. Understanding the molecular and cellular basis of CD24 function could provide important insights into T cell biology and autoimmunity.
Collapse
Affiliation(s)
- Yang Liu
- Division of Immunotherapy, Department of Surgery, The University of Michigan, Ann Arbor, MI 48109, USA.
| | | |
Collapse
|
20
|
Michaëlsson J, Mold JE, McCune JM, Nixon DF. Regulation of T cell responses in the developing human fetus. THE JOURNAL OF IMMUNOLOGY 2006; 176:5741-8. [PMID: 16670279 DOI: 10.4049/jimmunol.176.10.5741] [Citation(s) in RCA: 181] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Although human T cells enter the peripheral lymphoid tissues early during fetal development, the adaptive immune system in the fetus has largely been regarded as functionally immature and unresponsive to stimulation. In this study, we show that depletion of fetal CD4+CD25(high) T regulatory (T(Reg)) cells, which are present at high frequency in fetal lymphoid tissues, results in vigorous T cell proliferation and cytokine production in vitro, even in the absence of exogenous stimulation. Analysis of CD4+ and CD8(+) T cell populations revealed a large subset of cells that expressed the early activation Ag, CD69. We show that this population represents a subset of highly reactive fetal T cells actively suppressed by fetal CD4+CD25(high) T(Reg) cells during development. These findings indicate that fetal T cells are, in the absence of CD4+CD25(high) T(Reg) cells, highly responsive to stimulation and provide evidence for an important role for CD4+CD25(high) T(Reg) cells in controlling T cell responses in utero.
Collapse
Affiliation(s)
- Jakob Michaëlsson
- Gladstone Institute of Virology and Immunology, University of California-San Francisco, 1001 Potrero Avenue, San Francisco, CA 94158, USA
| | | | | | | |
Collapse
|
21
|
Jiang Q, Su H, Knudsen G, Helms W, Su L. Delayed functional maturation of natural regulatory T cells in the medulla of postnatal thymus: role of TSLP. BMC Immunol 2006; 7:6. [PMID: 16579866 PMCID: PMC1450317 DOI: 10.1186/1471-2172-7-6] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2005] [Accepted: 04/03/2006] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Generation of functional (CD4+)(CD8-)CD25+ regulatory T cells (Treg) in the murine thymus depends on FoxP3. Removal of the thymus from neonatal mice has been shown to result in a multiple organ autoimmune disease phenotype that can be prevented by introducing the FoxP3+ Treg population to the animal. It has therefore, been proposed that functional FoxP3+ Treg cells are not made in the neonatal thymus; however, it remains unclear when and where functional (FoxP3+)(CD4+)(CD8-)CD25+ thymocytes are generated in postnatal thymus. RESULTS We report that neither FoxP3 mRNA nor protein is expressed in (CD4+)(CD8-)CD25+, or (CD4+)(CD8-)CD25- thymocytes until 3-4 days post birth, despite the presence of mature (CD4+)(CD8-)CD25+/- thymocytes in the thymus by 1-2 days after birth. (FoxP3-)(CD4+)(CD8-)CD25+ thymocytes from day 2 newborn mice show no Treg activity. Interestingly, we are able to detect low numbers of FoxP3+ thymocytes dispersed throughout the medullary region of the thymus as early as 3-4 days post birth. Expression of FoxP3 is induced in embryonic day 17 fetal thymus organ culture (FTOC) after 4-6 days of in vitro culture. Treatment of FTOCs with thymic stromal derived lymphopoietin (TSLP) enhanced expression of FoxP3, and blocking the TSLP receptor reduces FoxP3 expression in FTOC. Furthermore, TSLP stimulates FoxP3 expression in purified (CD4+)CD8- thymocytes, but not in (CD4+)CD8+, (CD4-)CD8+ and (CD4-)CD8- thymocytes. CONCLUSION Expression of FoxP3 or Treg maturation is ontogenically distinct and kinetically delayed from the generation of (CD4+)(CD8-)CD25+ or (CD4+)(CD8-)CD25- thymocytes in the postnatal thymus. TSLP produced from medullary thymic epithelia cells (mTEC) contributes to the expression of FoxP3 and the maturation of natural regulatory T cells. Overall, these results suggest that the development of Treg cells requires paracrine signaling during late stages of thymocyte maturation that is distinct from signaling during positive or negative selection.
Collapse
Affiliation(s)
- Qi Jiang
- Lineberger Comprehensive Cancer Center, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Microbiology and Immunology, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Hua Su
- Lineberger Comprehensive Cancer Center, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Microbiology and Immunology, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Geoffry Knudsen
- Lineberger Comprehensive Cancer Center, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Microbiology and Immunology, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Whitney Helms
- Lineberger Comprehensive Cancer Center, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Microbiology and Immunology, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Lishan Su
- Lineberger Comprehensive Cancer Center, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Microbiology and Immunology, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Curriculum in Genetics and Molecular Biology, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
22
|
Almeida ARM, Rocha B, Freitas AA, Tanchot C. Homeostasis of T cell numbers: from thymus production to peripheral compartmentalization and the indexation of regulatory T cells. Semin Immunol 2005; 17:239-49. [PMID: 15826829 DOI: 10.1016/j.smim.2005.02.002] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
A system under homeostatic control tends to maintain its structure and functions by establishing dynamic equilibriums controlled by multiple regulatory mechanisms. We have shown that this is the case for immune system. Several different mechanisms seem to participate in the homeostatic control of T cell numbers and population distribution. In other words, besides a quantitative dimension, there is also a qualitative dimension in T cell homeostasis. This is achieved through competition by driving the specialization of sub-populations of lymphocytes to occupy specific niches in the peripheral pool and by developing independent homeostatic mechanisms for each particular cell sub-set. Thus, the sizes of the naïve and memory T cell compartments are governed by independent homeostatic mechanisms, which preserve the capacity to deal with any novel infection (conferred by the presence of naïve T cells) whilst ensuring the efficacy of memory responses when dealing with recurring antigens. Peripheral T cell homeostasis also depends on the integrity of sub-population structure and the presence of regulatory CD4+ CD25+ T cells. The indexation of regulatory CD4+ CD25+ T cell numbers to the numbers of peripheral activated CD4+ T cells is another mechanism of homeostasis that has major advantages in the control of immune responses. It ensures continuous regulation of T cell numbers throughout immune responses, allowing for increases in cell numbers as long as the proportion of CD4+ CD25+ regulatory T cells is kept.
Collapse
Affiliation(s)
- Afonso R M Almeida
- Lymphocyte Population Biology Unit, URA CNRS 1961, Institut Pasteur, 28 Rue du Dr. Roux, 75015 Paris, France
| | | | | | | |
Collapse
|
23
|
Davidson TS, Longnecker DS, Hickey WF. An experimental model of autoimmune pancreatitis in the rat. THE AMERICAN JOURNAL OF PATHOLOGY 2005; 166:729-36. [PMID: 15743785 PMCID: PMC1602363 DOI: 10.1016/s0002-9440(10)62294-8] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Autoimmune pancreatitis (AIP), a recently defined disease of unknown etiology, is characterized by inflammatory infiltrates in the pancreas with conspicuous involvement of the ducts. The disease clinically manifests in humans as epigastric pain, weight loss, and jaundice. This report describes the development of a novel animal model of this disease in the rat, which we have termed experimental autoimmune pancreatitis. Adoptive transfer of amylase-specific CD4(+) T cells was able to confer pancreatitis to naive syngeneic recipient animals. No treatments before the adoptive transfer of T cells were necessary for disease to ensue, and the severity of disease was proportional to the number of T cells administered. The pancreatic lesions of rats with experimental autoimmune pancreatitis were characterized histologically as overwhelmingly lymphocytic with occasional plasma cells, neutrophils, and mast cells. Acinar tissue destruction and ductular inflammation were common features, with less frequent involvement of larger ducts. Immunohistochemical analysis revealed the presence of CD4(+) T cells in large numbers as well as CD8(+) T cells, macrophages, and dendritic cells. Expression of MHC I and MHC II also increased at the site of the lesion. Clinically, the disease manifested as either failure to gain weight at a rate concomitant with control animals or as outright weight loss. Thus, administration of activated CD4(+) T cells specific for the pancreatic enzyme amylase can induce pancreatitis in the rat in a manner that is reminiscent of human AIP.
Collapse
Affiliation(s)
- Todd S Davidson
- Department of Pathology, Dartmouth Medical School, Dartmouth Hitchcock Medical Center, Lebanon, NH 03756, USA
| | | | | |
Collapse
|
24
|
Abstract
Regulatory T cells (TR cells) play a major role in controlling immune self reactivity. However, little is known about their occurrence and functions in early developmental stages. In this issue of the European Journal of Immunology, Cupedo et al. report the presence of functional CD4+CD25+ TR cells in the human fetus. In contrast to previous studies, the analysis is performed on fetal thymus, spleen and lymph node samples in addition to cord blood cells. Interestingly, TR cells are present in all these organs from 14 weeks of gestation, along with FoxP3 (forkhead box protein 3) RNA, a marker for naturally arising TR cells. The fetal TR cells show, however, phenotypic differences depending on their location, possibly because of variations in their activation state. The emergence of TR cells so early in fetal development raises a number of questions about the mechanisms of self reactivity and tolerance in the prenatal stages, which may have important implications for our understanding of childhood pathologies.
Collapse
Affiliation(s)
- Ana Izcue
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| | | |
Collapse
|
25
|
Ishimoto Y, Tomiyama-Miyaji C, Watanabe H, Yokoyama H, Ebe K, Tsubata S, Aoyagi Y, Abo T. Age-dependent variation in the proportion and number of intestinal lymphocyte subsets, especially natural killer T cells, double-positive CD4+ CD8+ cells and B220+ T cells, in mice. Immunology 2004; 113:371-7. [PMID: 15500624 PMCID: PMC1782573 DOI: 10.1111/j.1365-2567.2004.01961.x] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
The age-dependent variation in the proportion and number of lymphocyte subsets was examined at various extrathymic sites, including the liver, small intestine, colon and appendix in mice. In comparison with young mice (4 weeks of age), the number of total lymphocytes yielded by all tested organs was greater in adult (9 weeks) and old (40 weeks) mice. The major lymphocyte subset that expanded with age was interleukin-2 receptor (IL-2R) beta+ CD3int cells (50% of them expressed NK1.1) in the liver, whereas it was CD3+ IL-2Rbeta- NK1.1- cells at all intraepithelial sites in the intestine. Although NK1.1+ CD3+ cells were present at intraepithelial sites in the intestine, the proportion of this subset was rather low. The ratio of CD4 to CD8 tended to decrease among natural killer T (NKT) cells and T cells at all intraepithelial sites in the intestine with age. A unique population of double-positive CD4+ CD8+ cells in the small intestine increased in old mice. B220+ T cells were found mainly in the appendix and colon, and the proportion of these T cells decreased in old mice. Conventional NKT cells were very few in Jalpha281-/- and CD1d-/- mice in the liver, while NKT cells which existed in the appendix remained unchanged even in these mice. This was because unconventional CD8+ NKT cells were present in the intestine. The present results suggest that despite the fact that both the liver and intraepithelial sites in the intestine carry many extrathymic T cells, the distribution of lymphocyte subsets and their age-associated variation are site-specific.
Collapse
Affiliation(s)
- Yuiko Ishimoto
- Department of Immunology, Niigata University School of Medicine, Japan
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Shanker A. Is thymus redundant after adulthood? Immunol Lett 2004; 91:79-86. [PMID: 15019273 DOI: 10.1016/j.imlet.2003.12.012] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2003] [Revised: 12/20/2003] [Accepted: 12/28/2003] [Indexed: 11/29/2022]
Abstract
Thymus is considered to involute with age with a decline in thymic function. However, this generality is not universally and incontrovertibly true. Many studies performed in animals and men have proved to the contrary that thymic activity and function appear to be well maintained in the old age and may be indispensable for T cell reconstitution in different immunological settings. During some clinical situations where T cell pool needs to be regenerated, renewal of thymic activity and mass has been observed in an otherwise dormant thymic remnant. New studies have revealed a dynamic interplay between postnatal thymus output and peripheral T cell pool. Moreover, age-related loss of thymic function appears to be only quantitative and not qualitative. This review, thus, focuses on the different conditions that lead to thymic involution and attempts to bring about the emerging notion and the clinical relevance of continuous thymic activity well beyond the adulthood to optimise the function of the immune system in the context of cancer and infectious diseases.
Collapse
Affiliation(s)
- Anil Shanker
- Centre d'Immunologie de Marseille-Luminy, Institut National de la Santé et de la Recherche Médicale, Université de la Méditerranée, 163 Avenue de Luminy, Case 906, Marseille Cedex 09, 13288 France.
| |
Collapse
|
27
|
Abstract
The thymus is a complex epithelial organ in which thymocyte development is dependent upon the sequential contribution of morphologically and phenotypically distinct stromal cell compartments. It is these microenvironments that provide the unique combination of cellular interactions, cytokines, and chemokines to induce thymocyte precursors to undergo a differentiation program that leads to the generation of functional T cells. Despite the indispensable role of thymic epithelium in the generation of T cells, the mediators of this process and the differentiation pathway undertaken by the primordial thymic epithelial cells are not well defined. There is a lack of lineage-specific cell-surface-associated markers, which are needed to characterize putative thymic epithelial stem cell populations. This review explores the role of thymic stromal cells in T-cell development and thymic organogenesis, as well as the molecular signals that contribute to the growth and expansion of primordial thymic epithelial cells. It highlights recent advances in these areas, which have allowed for a lineage relationship amongst thymic epithelial cell subsets to be proposed. While many fundamental questions remain to be addressed, collectively these works have broadened our understanding of how the thymic epithelium becomes specialized in the ability to support thymocyte differentiation. They should also facilitate the development of novel, rationally based therapeutic strategies for the regeneration and manipulation of thymic function in the treatment of many clinical conditions in which defective T cells have an important etiological role.
Collapse
Affiliation(s)
- Jason Gill
- Department of Pathology and Immunology, Monash University, Faculty of Medicine, Nursing and Health Sciences, Alfred Medical Research and Education Precinct, Prahran, Australia.
| | | | | | | | | | | |
Collapse
|
28
|
Roper RJ, McAllister RD, Biggins JE, Michael SD, Min SH, Tung KSK, Call SB, Gao J, Teuscher C. Aod1 controlling day 3 thymectomy-induced autoimmune ovarian dysgenesis in mice encompasses two linked quantitative trait loci with opposing allelic effects on disease susceptibility. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2003; 170:5886-91. [PMID: 12794114 DOI: 10.4049/jimmunol.170.12.5886] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Day 3 thymectomy (D3Tx) leads to a paucity of CD4(+)CD25(+) suppressor T cells, a loss of peripheral tolerance, and the development of organ-specific autoimmune disease in adult mice. Importantly, D3Tx does not lead to autoimmune disease in all mouse strains, indicating that this process is genetically controlled. Previously, we reported linkage of D3Tx-induced autoimmune ovarian dysgenesis (AOD) and its intermediate phenotypes, antiovarian autoantibody responsiveness, oophoritis, and atrophy, to five quantitative trait loci (QTL), designated Aod1 through Aod5. We also showed interaction between these QTL and H2 as well as Gasa2, a QTL controlling susceptibility to D3Tx-induced autoimmune gastritis. To physically map Aod1, interval-specific bidirectional recombinant congenic strains of mice were generated and studied for susceptibility to D3Tx-induced AOD. Congenic mapping studies revealed that Aod1 controls susceptibility to oophoritis and comprises two linked QTL with opposing allelic effects. Aod1a resides between D16Mit211 (23.3 cM) and D16Mit51 (66.75 cM) on chromosome 16. Aod1b maps proximal of Aod1a between D16Mit89 (20.9 cM) and D16Mit211 (23.3 cM) and includes the candidate genes stefin A1, A2, and A3 (Stfa1-Stfa3), inhibitors of cathepsin S, a cysteine protease required for autoantigen presentation, and the development of autoimmune disease of the salivary and lacrimal glands following D3Tx. cDNA sequencing revealed the existence of structural polymorphisms for both Stfa1 and Stfa2. Given the roles of cathepsins in Ag processing and presentation, Stfa1 and Stfa2 alleles have the potential to control susceptibility to autoimmune disease at the level of both CD4(+)CD25(+) suppressor and CD4(+)CD25(-) effector T cells.
Collapse
Affiliation(s)
- Randall J Roper
- Department of Veterinary Pathobiology, University of Illinois, Urbana, IL 61802, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Chapman JC, Min S, Kunaporn S, Tung K, Shah S, Michael SD. The administration of cortisone to female B6A mice during their immune adaptive period causes anovulation and the formation of ovarian cysts. Am J Reprod Immunol 2002; 48:184-9. [PMID: 12443030 DOI: 10.1034/j.1600-0897.2002.01081.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
PROBLEM Female mice that are injected with estradiol-17beta (E2) and testosterone during the 7-day immune adaptive period are infertile at adulthood. To determine whether the resultant infertility can be caused by steroids other than estrogens/ androgens, this study examined the effect of injecting cortisone, alone, and in combination with E2 and testosterone, on reproductive function. METHOD OF STUDY Neonatal (C57BL/6J x A/J)F1 B6A female mice were injected from 3 to 6 days of age with sesame oil:ethanol (9:1; v:v), alone, or containing 20 microgg cortisone acetate, 20 microg E2, or 20 microg testosterone. Two additional groups were given 20 microg cortisone acetate in combination with 20 microg E2 or 20 microg testosterone. At adulthood the animals were killed, the stage of vaginal estrus determined, the ovaries examined for the presence of corpora lutea and follicular cysts, and circulating levels of progesterone, E2, and testosterone were measured by radioimmunoassay (RIA). RESULTS It was found that injections of cortisone seriously compromise reproductive development. For example, 11% of cortisone-injected animals had ovaries that lacked corpora lutea. In addition, 39% of cortisone-injected females had ovaries with follicular cysts. Cortisone-injected females also had low levels of circulating progesterone (18 ng/mL versus 30 ng/mL for the sesame oil-injected females). CONCLUSION It is concluded that the deleterious effect of steroids on reproductive function, when administered during the immune adaptive period, is not restricted to estrogens and androgens. It is proposed that injections of cortisone alter T-lymphocyte subsets, which contributes to anovulation and the production of follicular cysts.
Collapse
Affiliation(s)
- John C Chapman
- Department of Biological Sciences, Binghamton University, NY 13902-6000, USA
| | | | | | | | | | | |
Collapse
|
30
|
Burt RK, Slavin S, Burns WH, Marmont AM. Induction of tolerance in autoimmune diseases by hematopoietic stem cell transplantation: getting closer to a cure? Int J Hematol 2002; 76 Suppl 1:226-47. [PMID: 12430858 DOI: 10.1007/bf03165251] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Hematopoietic stem cells (HSCs) are the earliest cells of the immune system, giving rise to B and T lymphocytes, monocytes, tissue macrophages, and dendritic cells. In animal models, adoptive transfer of HSCs, depending on circumstances, may cause, prevent, or cure autoimmune diseases. Clinical trials have reported early remission of otherwise refractory autoimmune disorders after either autologous or allogeneic hematopoietic stem cell transplantation (HSCT). By percentage of transplantations performed, autoimmune diseases are the most rapidly expanding indication for stem cell transplantation. Although numerous editorials or commentaries have been previously published, no prior review has focused on the immunology of transplantation tolerance or development of phase 3 autoimmune HSCT trials. Results from current trials suggest that mobilization of HSCs, conditioning regimen, eligibility and exclusion criteria, toxicity, outcome, source of stem cells, and posttransplantation follow-up need to be disease specific. HSCT-induced remission of an autoimmune disease allows for a prospective analysis of events involved in immune tolerance not available in cross-sectional studies.
Collapse
Affiliation(s)
- Richard K Burt
- Northwestern University Medical Center, Division of Immune Therapy and Autoimmune Disease, Chicago, IL, USA
| | | | | | | |
Collapse
|
31
|
Morón G, Maletto B, Rópolo A, Pistoresi-Palencia MC. Changes in the development of experimental autoimmune prostatitis (EAP) by castration in aged rats. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2000; 24:673-682. [PMID: 10831801 DOI: 10.1016/s0145-305x(99)00088-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
During Experimental Autoimmune Prostatitis (EAP), 12-month-old rats show a higher cellular autoimmune response and lower humoral autoimmune response against prostatic components than 3-month-old rats subjected to the same antigen stimulus. We analyzed if thymus recovery by orchidectomy could affect the development of EAP in 12-month-old rats. Thirty days after gonadectomy, 12-month-old rats showed an increment in the thymic mass and in the thymocytes absolute number, with percentages of the four main cell subpopulations (defined by CD4-CD8 molecules expression) similar to the 3-month-old rats. The DTH response of castrated 12-month-old with EAP were diminished in comparison with sham-castrated 12-month-old rats with EAP, resembling the values observed in 3-month-old rats with EAP. The prostates of castrated 12-month-old rats with EAP did not show inflammatory mononuclear cell infiltration, as did control 3- and 12-month-old rats with EAP. Castration seems to modulate negatively EAP in 12-month-old rats, possibly through the regeneration of thymus after testosterone deprivation.
Collapse
Affiliation(s)
- G Morón
- Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Ciudad Universitaria, 5000, Córdoba, Argentina
| | | | | | | |
Collapse
|
32
|
Abstract
Recent evidence supports the idea that T cells in neonatal animals are developmentally mature in their capacity to mount protective helper and cytotoxic responses. Nonetheless, neonates fall prey to infections which have little effect on adults and they often fail to mount mature responses to environmental, experimental, or vaccine antigens. To reconcile these observations, it may be important to consider the potential role of apoptosis in neonatal immune responses. In adults, apoptosis is well established as a centrally important process in the homeostasis of cellular immune responses. Activated T cells deprived of IL-2 undergo cytokine withdrawal-induced apoptosis. Previously activated T cells can also be triggered by secondary stimulation to undergo activation induced apoptosis. This review summarizes our current state of knowledge of apoptosis of murine neonatal T cells and discusses the possible impact(s) of this apoptosis on neonatal immune responses in vivo.
Collapse
Affiliation(s)
- B Adkins
- Department of Microbiology and Immunology R138, University of Miami Medical School, FL 33101, USA.
| | | |
Collapse
|
33
|
Colson YL, Abou El-Ezz AY, Gaines BA, Ildstad ST. Positive and negative selection of alphabetaTCR+ T cells in thymectomized adult radiation bone marrow chimeras. Transplantation 1999; 68:403-10. [PMID: 10459545 DOI: 10.1097/00007890-199908150-00013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND The mature T-cell repertoire is characterized by the negative selection of potentially autoreactive T cells and the positive selection of T cells restricted to antigen-recognition in the context of self-MHC molecules. It is currently believed that the thymus is critical for these selection events. Although alpha(beta)T cell receptor (TCR)+ T cells have been reported in thymectomized recipients, whether this represents clonal expansion of residual T cells or de novo generation of new T cells in the absence of a thymus has not been definitively evaluated. METHODS In the current study, development of the T cell repertoire was evaluated in adult radiation bone marrow chimeras prepared after complete surgical thymectomy. RESULTS CD4+ and CD8+ T cells were present and exhibited donor-specific TCR-Vbeta expression and self-tolerance, indicative of negative selection. Positive selection was confirmed with the demonstration of host MHC restriction and the presence of donor-derived CD8+ T cells after the transplantation of marrow from Class I deficient donors into normal recipients. CONCLUSIONS These data provide evidence, for the first time, that the development of a functional T-cell repertoire can occur in adult recipients without the thymic microenvironment.
Collapse
Affiliation(s)
- Y L Colson
- The Department of Surgery, University of Pittsburgh, Pennsylvania 15261, USA
| | | | | | | |
Collapse
|
34
|
Morse SS, Sakaguchi N, Sakaguchi S. Virus and Autoimmunity: Induction of Autoimmune Disease in Mice by Mouse T Lymphotropic Virus (MTLV) Destroying CD4+ T Cells. THE JOURNAL OF IMMUNOLOGY 1999. [DOI: 10.4049/jimmunol.162.9.5309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Abstract
Neonatal infection of the mouse T lymphotropic virus (MTLV), a member of herpes viridae, causes various organ-specific autoimmune diseases, such as autoimmune gastritis, in selected strains of normal mice. The infection selectively depletes CD4+ T cells in the thymus and periphery for 2–3 wk from 1 wk after infection. Thymectomy 3 wk after neonatal MTLV infection enhances the autoimmune responses and produces autoimmune diseases at higher incidences and in a wider spectrum of organs than MTLV infection alone. On the other hand, inoculation of peripheral CD4+ cells from syngeneic noninfected adult mice prevents the autoimmune development. These autoimmune diseases can be adoptively transferred to syngeneic athymic nude mice by CD4+ T cells. The virus is not detected by bioassay in the organs/tissues damaged by the autoimmune responses. Furthermore, similar autoimmune diseases can be induced in normal mice by manipulating the neonatal thymus/T cells (e.g., by neonatal thymectomy) without virus infection. These results taken together indicate that neonatal MTLV infection elicits autoimmune disease by primarily affecting thymocytes/T cells, not self Ags. It may provoke or enhance thymic production of CD4+ pathogenic self-reactive T cells by altering the thymic clonal deletion mechanism, or reduce the production of CD4+ regulatory T cells controlling self-reactive T cells, or both. The possibility is discussed that other T cell-tropic viruses may cause autoimmunity in humans and animals by affecting the T cell immune system, not the self Ags to be targeted by the autoimmunity.
Collapse
Affiliation(s)
| | - Noriko Sakaguchi
- †Department of Immunopathology, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan
| | - Shimon Sakaguchi
- †Department of Immunopathology, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan
| |
Collapse
|
35
|
Schneider MK, Grönvik KO. CD4-CD8-C.B-17 SCID thymocytes enter the CD4+CD8+ stage in the presence of neonatally grafted T cells. Scand J Immunol 1998; 47:466-74. [PMID: 9627131 DOI: 10.1046/j.1365-3083.1998.00331.x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The aim of this work was to study the selection of donor T cells and their influence on thymic development in C.B-17 scid/scid (severe combined immunodeficient; SCID) mice during chronic graft-versus-host disease (GVHD). Recipient SCID mice (H-2d), neonatally grafted with allogeneic peripheral T cells from CBA/J strain (H-2k) of mice, only developed a mild acute GVHD, and were, at the chronic stage, devoid of pathological symptoms. Thymic cell numbers of injected mice differed from 10(5) to 1.2 x 10(7) at 2-3 weeks post-injection (p.i.), and from 4 x 10(5) to 8.5 x 10(7) at 2 months p.i. In these mice, the thymus size was correlated to the CD4-CD8- (double negative; DN) to CD4+CD8+ (double positive; DP) cell ratio, where at 2 months p.i., 8 out of 16 treated SCID mice contained 5 x 10(6) cells or more and also possessed the highest frequencies of endogenous DP cells (25-95%). In contrast to previous findings, peripheral donor T cells from allogeneic and syngeneic mice, infiltrating the host thymus, had a positive effect on the development of endogenous DP thymocytes. Furthermore, these thymocytes were developmentally blocked at the DP stage, occasionally in combination with the expression of CD25, CD44 and CD117 but in the absence of T-cell receptor (TCR) expression. Also, at this time-point, the CBA/J donor TCR Vbeta repertoire was equal to that of normal CBA/J mice, but purified responding donor cells were proliferatively inhibited against H-2d stimulators in ex vivo mixed lymphocyte cultures. In contrast, the same responders showed a pronounced proliferation against syngeneic H-2Kk stimulators, suggesting either a reversion from anergy of autoreactive CBA/J T cells or a vast expansion of multiple self-reactive T-cell clones, when parked in a milieu with a lower concentration of self-antigens.
Collapse
MESH Headings
- Age Factors
- Animals
- Animals, Newborn
- CD2 Antigens/analysis
- CD3 Complex/analysis
- CD4 Antigens/analysis
- CD4-Positive T-Lymphocytes/cytology
- CD4-Positive T-Lymphocytes/immunology
- CD8 Antigens/analysis
- CD8-Positive T-Lymphocytes/cytology
- CD8-Positive T-Lymphocytes/immunology
- Cell Count
- Cell Differentiation
- Chronic Disease
- Graft vs Host Disease/pathology
- Lymphocyte Activation/immunology
- Major Histocompatibility Complex/immunology
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Inbred CBA
- Mice, SCID
- Proto-Oncogene Proteins c-kit/analysis
- Receptors, Antigen, T-Cell, alpha-beta/analysis
- Receptors, Interleukin-2/analysis
- Severe Combined Immunodeficiency/immunology
- Severe Combined Immunodeficiency/pathology
- T-Lymphocytes/immunology
- T-Lymphocytes/transplantation
- Thymus Gland/cytology
- Thymus Gland/growth & development
- Time Factors
- Transplantation, Homologous
Collapse
Affiliation(s)
- M K Schneider
- Department of Vaccine Research, National Veterinary Institute, Uppsala University, Sweden
| | | |
Collapse
|
36
|
Maity R, Nair SR, Caspi RR, Nelson LM. Post-thymectomy murine experimental autoimmune oophoritis is associated with reduced natural killer cell activity. Am J Reprod Immunol 1997; 38:360-5. [PMID: 9352028 DOI: 10.1111/j.1600-0897.1997.tb00312.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
PROBLEM Natural killer (NK) cells can influence the immune response by secreting potent lymphokines. It has been suggested that NK cells have a suppressive action on B cells, and that impaired NK cell activity may play a role in some types of autoimmunity. NK cell abnormalities have been reported in women with premature ovarian failure. We therefore examined NK cell activity during the development of murine experimental autoimmune oophoritis, which serves as a model for autoimmune ovarian failure in women. METHOD OF STUDY Neonatally thymectomized and sham-operated C57B1/6 x A/J (B6A) mice were prepared and sacrificed at 4, 6, 8, and 10 weeks after surgery. Splenic NK cell activity was determined in groups of five or more mice by measuring the percent specific lysis of target YAC-1 lymphoma cells using a standard 4-hr chromium release cytotoxicity assay. The number of splenic NK cells in neonatally thymectomized and sham-operated animals was also compared using flow cytometry. In a subsequent experiment, interleukin 12 (IL-12; NK cell-stimulating factor) was administered to neonatal mice before neonatal thymectomy. RESULTS Neonatally thymectomized mice with associated autoimmune oophoritis had a 75% reduction in the number of splenic NK cells, and 50% or greater reduction in splenic NK cell activity at 4, 6, and 8 weeks after surgery. IL-12 treatment before neonatal thymectomy maintained NK cell activity and was shown to ameliorate the associated autoimmune oophoritis. CONCLUSION Murine post-thymectomy autoimmune oophoritis is associated with reduced NK cell number and impaired NK cell activity, and in these respects the model is similar to premature ovarian failure in women. Research to define the relationship between NK cell abnormalities and the mechanism of ovarian failure in this model might lend insight into the pathogenesis of premature ovarian failure in women.
Collapse
Affiliation(s)
- R Maity
- Section on Women's Health, Developmental Endocrinology Branch, National Institute of Child Health and Human Development, Bethesda, Maryland 20892-1862, USA
| | | | | | | |
Collapse
|
37
|
Abstract
We investigated the role of continuous thymus output in the shaping of mature T cell repertoires by studying in vivo the survival of a single clone of mature Rag2-deficient T cell receptor (TCR) transgenic cells at different stages of activation in the absence or presence of thymus export. In the absence of thymus export, TCR-transgenic lymphocytes survived indefinitely in the peripheral pools. When new lymphocytes were produced in the thymus and migrated to the periphery, resident memory T cells were maintained in constant numbers, whereas naive and self-reactive T cells were replaced by recent thymus migrants. This T cell renewal ensured both the efficiency of recall responses to antigens as memory T cells persisted independently of thymus output, and the capacity of the immune system to respond to new antigen stimulation as the naive T cell pool was continuously renewed. Our results also indicate that thymus export is required to control the number of self-reactive peripheral T cells that may invade the peripheral pools if thymus output fails.
Collapse
Affiliation(s)
- C Tanchot
- U345 Institut National de la Sante et de la Recherche Medicale, Institut Necker, 156, 75730 Paris Cedex 15, France
| | | |
Collapse
|
38
|
Davis CM, McLaughlin TM, Watson TJ, Buckley RH, Schiff SE, Hale LP, Haynes BF, Markert ML. Normalization of the peripheral blood T cell receptor V beta repertoire after cultured postnatal human thymic transplantation in DiGeorge syndrome. J Clin Immunol 1997; 17:167-75. [PMID: 9083893 DOI: 10.1023/a:1027382600143] [Citation(s) in RCA: 24] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Complete DiGeorge syndrome is an immunodeficiency disease characterized by thymic aplasia and the absence of functioning peripheral T cells. A patient with this syndrome was transplanted with cultured postnatal human thymic tissue. Within 5 weeks of transplantation, flow cytometry, T cell receptor V beta sequence analysis, and cell function studies showed the presence of oligoclonal populations of nonfunctional clonally expanded peripheral T cells that were derived from pretransplantation T cells present in the skin. However, at 3 months posttransplantation, a biopsy of the transplanted thymus showed normal intrathymic T cell maturation of host T cells with normal TCR V beta expression on thymocytes. By 9 months postransplantation, peripheral T cell function was restored and the TCR V beta repertoire became polyclonal, coincident with the appearance of normal T cell function. These data suggest that the transplanted thymus was responsible for the establishment of a new T cell repertoire via thymopoiesis in the chimeric thymic graft.
Collapse
Affiliation(s)
- C M Davis
- Department of Pediatrics, Duke University Medical Center, Durham, North Carolina 27710, USA
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Abstract
Premature ovarian failure (POF) is defined as a syndrome characterized by menopause before the age of 40 yr. The patients suffer from anovulation and hypoestrogenism. Approximately 1% of women will experience menopause before the age of 40 yr. POF is a heterogeneous disorder with a multicausal pathogenesis involving chromosomal, genetic, enzymatic, infectious, and iatrogenic causes. There remains, however, a group of POF patients without a known etiology, the so-called "idiopathic" form. An autoimmune etiology is hypothesized for the POF cases with a concomitant Addison's disease and/or oöphoritis. It is concluded in this review that POF in association with adrenal autoimmunity and/or Addison's disease (2-10% of the idiopathic POF patients) is indeed an autoimmune disease. The following evidence warrants this view: 1) The presence of autoantibodies to steroid-producing cells in these patients; 2) The characterization of shared autoantigens between adrenal and ovarian steroid-producing cells; 3) The histological picture of the ovaries of such cases (lymphoplasmacellular infiltrate around steroid-producing cells); 4) The existence of various autoimmune animal models for this syndrome, which underlines the autoimmune nature of the disease. There is some circumstantial evidence for an autoimmune pathogenesis in idiopathic POF patients in the absence of adrenal autoimmunity or Addison's disease. Arguments in support of this are: 1) The presence of cellular immune abnormalities in this POF patient group reminiscent of endocrine autoimmune diseases such as IDDM, Graves' disease, and Addison's disease; 2) The more than normal association with IDDM and myasthenia gravis. Data on the presence of various ovarian autoantibodies and anti-receptor antibodies in these patients are, however, inconclusive and need further evaluation. A strong argument against an autoimmune pathogenesis of POF in these patients is the nearly absent histological confirmation (the presence of an oöphoritis) in these cases (< 3%). However, in animal models using ZP immunization, similar follicular depletion and fibrosis (as in the POF women) can be detected. Accepting the concept that POF is a heterogenous disorder in which some of the idiopathic forms are based on an abnormal self-recognition by the immune system will lead to new approaches in the treatment of infertility of these patients. There are already a few reports on a successful ovulation-inducing treatment of selected POF patients (those with other autoimmune phenomena) with immunomodulating therapies, such as high dosages of corticosteroids (288-292).
Collapse
Affiliation(s)
- A Hoek
- Department of Immunology, Erasmus University, Rotterdam, The Netherlands
| | | | | |
Collapse
|
40
|
Thiele HG, Haag F, Koch-Nolte F. Molecular cloning and characterization of the T-cell mono(ADP-ribosyl)transferase RT6. Relationships to other mADPRTs and possible functions. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 1997; 419:109-20. [PMID: 9193643 DOI: 10.1007/978-1-4419-8632-0_13] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Affiliation(s)
- H G Thiele
- Department of Immunology, University Hospital Hamburg-Eppendorf, Germany
| | | | | |
Collapse
|
41
|
Garza KM, Griggs ND, Tung KS. Neonatal injection of an ovarian peptide induces autoimmune ovarian disease in female mice: requirement of endogenous neonatal ovaries. Immunity 1997; 6:89-96. [PMID: 9052840 DOI: 10.1016/s1074-7613(00)80245-9] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Neonatal female mice injected with the self ZP3 peptide are not tolerant to the peptide; they develop autoimmune ovarian disease (AOD) and autoantibody response 5 weeks later. ZP3 challenge leads to severe AOD and ZP3-specific T cell and antibody responses. In contrast, neonatal tolerance to foreign ZP3 peptide is established in male mice: ZP3 peptide-specific T cell proliferative response is reduced and AOD is absent in ovarian grafts. Tolerance is associated with a Th2-dominant T cell cytokine and antibody isotype profiles. As controls, neonatal tolerance to foreign peptides, with Th2 deviation, was induced in both male and female mice. Endogenous ZP3 is important for the gender difference. Ablation of ovaries in female mice on days 2 and 5, but not on day 7 or 14, switches the ZP3 autoimmune response to a tolerogenic response with a concomitant change in cytokine profile. Thus, neonatal self ZP3 peptide, supported by endogenous ovaries within a neonatal time window, evokes a pathogenic autoimmune response.
Collapse
Affiliation(s)
- K M Garza
- Department of Microbiology, University of Virginia, Charlottesville 22908, USA
| | | | | |
Collapse
|
42
|
Mouse Mammary Tumor Virus: Immunological Interplays between Virus and Host **This article was accepted for publication on 1 October 1996. Adv Immunol 1997. [DOI: 10.1016/s0065-2776(08)60743-9] [Citation(s) in RCA: 49] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
43
|
Moroda T, Iiai T, Kawachi Y, Kawamura T, Hatakeyama K, Abo T. Restricted appearance of self-reactive clones into T cell receptor intermediate cells in neonatally thymectomized mice with autoimmune disease. Eur J Immunol 1996; 26:3084-91. [PMID: 8977308 DOI: 10.1002/eji.1830261239] [Citation(s) in RCA: 24] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Neonatally thymectomized (NTx) mice fall victim to such autoimmune diseases as gastritis and pancreatitis with aging. Self-reactive T cell clones are known to be consistently generated through TCR intermediate (i.e. TCRint) cell differentiation in normal mice (i.e. via the extrathymic pathways and an alternative intrathymic pathway). It was investigated whether the generation of such clones in NTx mice follows this rule or whether they are generated by default via mainstream T cell differentiation in the thymus. The majority of T cells generated in NTx mice were TCRint cells in all organs tested. In contrast to athymic mice, which carry only TCRint cells with aging, a leaky appearance of high TCR (i.e. TCRhi) cells emerged in the lymph nodes and other organs of NTx mice. Self-reactive clones estimated by anti-Vbeta monoclonal antibodies in conjunction with the Mls system were confined to TCRint cells in all tested organs, including a target organ, the stomach, in NTx mice. A leaky population of TCRhi cells did not contain a significant number of self-reactive clones. Moreover, such self-reactive clones among TCRint cells in NTx mice with autoimmune disease were shown to be nonanergic in the proliferation assay. The present results suggest that the generation of self-reactive clones is totally due to TCRint cell differentiation, although it is still undetermined whether the expanding TCRint cell population is generated via the extrathymic pathway or an alternative intrathymic pathway. It is shown here not to be due to a failure of the TCRhi cell-differentiation pathway in NTx mice.
Collapse
Affiliation(s)
- T Moroda
- Department of Immunology, Niigata University School of Medicine, Japan
| | | | | | | | | | | |
Collapse
|
44
|
Asano M, Toda M, Sakaguchi N, Sakaguchi S. Autoimmune disease as a consequence of developmental abnormality of a T cell subpopulation. J Exp Med 1996; 184:387-96. [PMID: 8760792 PMCID: PMC2192701 DOI: 10.1084/jem.184.2.387] [Citation(s) in RCA: 975] [Impact Index Per Article: 33.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Neonatal thymectomy (NTx), especially around day 3 after birth, causes various organ-specific autoimmune diseases in mice. This report shows that: (a) T cells expressing the interleukin 2 receptor alpha chains (CD25) ontogenically begin to appear in the normal periphery immediately after day 3, rapidly increasing within 2 wk to nearly adult levels (approximately 10% of CD3+ cells, especially of CD4+ cells); (b) NTx on day 3 eliminates CD25+ T cells from the periphery for several days; inoculation immediately after NTx of CD25+ splenic T cells from syngeneic non-Tx adult mice prevents autoimmune development, whereas inoculation of CD25- T cells even at a larger dose does not; and furthermore, (c) similar autoimmune diseases can be produced in adult athymic nu/nu mice by inoculating either spleen cell suspensions from 3-d-old euthymic nu/+ mice or CD25+ cell-depleted spleen cell suspensions from older, even 1-yr-old, nu/+ mice. The CD25- populations from neonates or adults are also similar in the profile of cytokine formation. These results, taken together, indicate that one aspect of peripheral self-tolerance is maintained by CD25+ T cells that sustain potentially pathogenic self-reactive T cells in a CD25- dormant state; the thymic production of the former is developmentally programmed to begin on day 3 after birth in mice. Thus, NTx on day 3 can, at least transiently, eliminate/reduce the autoimmune-preventive CD25+ T cells, thereby leading to activation of the self-reactive T cells that have been produced before NTx.
Collapse
Affiliation(s)
- M Asano
- Department of Immunopathology, Tokyo Metropolitan Institute of Geromology, Japan
| | | | | | | |
Collapse
|
45
|
Modigliani Y, Coutinho A, Pereira P, Le Douarin N, Thomas-Vaslin V, Burlen-Defranoux O, Salaün J, Bandeira A. Establishment of tissue-specific tolerance is driven by regulatory T cells selected by thymic epithelium. Eur J Immunol 1996; 26:1807-15. [PMID: 8765025 DOI: 10.1002/eji.1830260822] [Citation(s) in RCA: 90] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Grafts of thymic epithelium (TE) rudiments restore T cell development and function in allogeneic athymic mice. These TE chimeras are specifically tolerant to grafts of peripheral tissues (e.g. skin and heart) from the TE donor strain, although they harbor peripheral immunocompetent T cells capable of rejecting those grafts. Initial analysis has shown that TE chimeras also harbor TE-selected CD4 T lymphocytes that inhibit graft rejection by tissue-reactive T cells in immunocompetent recipients. Peripheral tolerance in TE chimeras is thus maintained by dominant mechanisms dependent on regulatory CD4 T lymphocytes. Here we show that TE-selected regulatory T cells recruit nontolerant tissue-reactive CD4 and CD8 T cells to express similar regulatory functions. Only recent thymic emigrants, but not peripheral resident mature T cells are susceptible to this process of functional education, which also requires exposure to specific antigens and occurs entirely in the periphery. We propose that these mechanisms play a major role in establishing and maintaining natural self tolerance to tissue-specific antigens.
Collapse
Affiliation(s)
- Y Modigliani
- Unité d'Immunobiologie, CNRS URA 1961, Institut Pasteur, Paris, France
| | | | | | | | | | | | | | | |
Collapse
|
46
|
George AJ, Ritter MA. Thymic involution with ageing: obsolescence or good housekeeping? IMMUNOLOGY TODAY 1996; 17:267-72. [PMID: 8962629 DOI: 10.1016/0167-5699(96)80543-3] [Citation(s) in RCA: 248] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Affiliation(s)
- A J George
- Dept of Immunology, Royal Postgraduate Medical School, London, UK.
| | | |
Collapse
|
47
|
Modigliani Y, Bandeira A, Coutinho A. A model for developmentally acquired thymus-dependent tolerance to central and peripheral antigens. Immunol Rev 1996; 149:155-20. [PMID: 9005213 DOI: 10.1111/j.1600-065x.1996.tb00903.x] [Citation(s) in RCA: 93] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Current models of tolerance to peripheral, tissue-specific antigens contain some major caveats. First, they consider peripheral tolerance independently from intrathymic T cell selection, a dichotomy that is challenged by observations on TE-induced tolerance. Second, they do not account for the fact that vertebrates are more readily tolerised in development than in adult life. Third, they do not explain the fact that embryonic/neonatal tolerance to foreign tissues can only be induced by HC or TE. A model of thymic selection and peripheral tolerance is developed here that resolves those problems, by assuming two classes of T cell effector functions, one being regulatory and the other aggressive. Three postulates are required: (1) both epithelial and hemopoietic cellular compartments of the thymic stroma can support both positive and negative selection of T cells, but with vastly different avidity requirements and efficiency; (2) positively selected T cells with the highest avidity that escape deletion are activated intrathymically and irreversibly committed for regulatory effector functions; (3) the functional phenotype of all other thymic emigrants is determined in the periphery upon encounter with antigen. Functional commitment in the periphery depends on the maturity stage (RTE or PMR) of the immunocompetent cell, on the nature of the antigen-presenting cells, and on the effector classes of other T lymphocytes interacting on the same presenting cell. This model explains a number of observations on experimental autoimmune disease and transplantation tolerance, and it contains several readily testable predictions.
Collapse
Affiliation(s)
- Y Modigliani
- Unité d'Immunobiologie, CNRS URA, Institut Pasteur, Paris, France
| | | | | |
Collapse
|
48
|
Affiliation(s)
- P A Gleeson
- Department of Pathology and Immunology, Monash University Medical School, Victoria, Australia.
| | | | | |
Collapse
|
49
|
Cameron ER, Campbell M, Blyth K, Argyle SA, Keanie L, Neil JC, Onions DE. Apparent bypass of negative selection in CD8+ tumours in CD2-myc transgenic mice. Br J Cancer 1996; 73:13-7. [PMID: 8554976 PMCID: PMC2074291 DOI: 10.1038/bjc.1996.3] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
A role for antigen stimulation in lymphoid neoplasia has been postulated and is supported by indirect evidence that suggests that the interaction of antigen with both T cells and B cells may constitute an epigenetic event that can contribute to tumour induction or tumour progression. Using myc-bearing transgenic mice that develop mainly clonal T-cell lymphomas we have investigated the possibility that endogenous antigen-mediated clonal deletion might be overridden in tumorigenesis. CD2-myc transgenic mice were backcrossed on to a CBA/Ca background to ensure Mtv-mediated deletion of V beta 11-expressing T cells in the resultant offspring. Lymphomas arising from these mice were subsequently screened for V beta 11 expression. There was a clear correlation between the age at which mice developed neoplasia and the tumour phenotype. Mice with CD4- CD8+ tumours succumbed to thymic lymphoma at a significantly younger age than mice developing CD4+ CD8+ tumours. A small number of tumours consisted of the 'forbidden' V beta 11 phenotype, showing that cells vulnerable to transformation could escape negative selection. The majority of the V beta 11-positive tumours were CD4- CD8+ and were only observed in mice showing clinical evidence of tumour development at a relatively young age. The phenotype of these cells and the age at which tumours arose suggests that T cells escaping tolerance may be susceptible to transformation.
Collapse
MESH Headings
- Animals
- CD2 Antigens/genetics
- CD4-CD8 Ratio
- CD4-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/pathology
- Cell Transformation, Neoplastic/immunology
- Genes, myc
- Lymphoma, T-Cell/genetics
- Lymphoma, T-Cell/immunology
- Lymphoma, T-Cell/pathology
- Mice
- Mice, Inbred C57BL
- Mice, Inbred CBA
- Mice, Transgenic
- Phenotype
- Receptors, Antigen, T-Cell, alpha-beta/genetics
- Receptors, Antigen, T-Cell, alpha-beta/immunology
- Superantigens/immunology
Collapse
Affiliation(s)
- E R Cameron
- Department of Veterinary Pathology, University of Glasgow Veterinary School, UK
| | | | | | | | | | | | | |
Collapse
|
50
|
Modigliani Y, Pereira P, Thomas-Vaslin V, Salaün J, Burlen-Defranoux O, Coutinho A, Le Douarin N, Bandeira A. Regulatory T cells in thymic epithelium-induced tolerance. I. Suppression of mature peripheral non-tolerant T cells. Eur J Immunol 1995; 25:2563-71. [PMID: 7589127 DOI: 10.1002/eji.1830250924] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Athymic mice grafted at birth with allogeneic thymic epithelium (TE) display life-long tolerance to tissue grafts of the TE donor strain, in spite of harboring peripheral T cells capable of rejecting those grafts. Tolerance is maintained in these chimeras by TE-specific regulatory CD4 T cells. We presently address the quantification and the mechanisms of this dominant tolerance process. C57BL/6 mice containing variable but defined numbers of peripheral, resident T cells received cell transfers of graded numbers of peripheral T cells from B6(BALB E10) chimeras (C57BL/6 nude mice grafted with TE from 10-day-old BALB/c embryos), resulting in a series of animals containing a wide range of donor (tolerant) versus host (non-tolerant) T cell chimerism. Increasing the relative representation of donor T cells results in a progressive delay in the rejection of BALB/c skin grafts, life-long tolerance being achieved at a ratio of tolerant and non-tolerant T cell populations of 1. In recipients displaying full tolerance, graft-reactive non-tolerant T cells were not deleted, anergized or committed to noninflammatory functions. Thus, sorted host T cells from tolerant recipients readily rejected BALB/c skin grafts upon transfer to immunodeficient animals. Finally, measurements of "helper" and inflammatory activities, as well as interleukin-4 and interferon-gamma production, failed to discriminate between T cell populations from tolerant and non-tolerant animals after specific in vitro stimulation. We conclude that: (a) TE-selected regulatory T cells can suppress, in a quantitative manner, in vivo T cell responses against major and minor histocompatibility antigens expressed by the TE and, (b) this suppressive activity neither inactivates mature non-tolerant T cells, nor does it seem to drive their differentiation along noninflammatory pathways.
Collapse
Affiliation(s)
- Y Modigliani
- Unité d'Immunobiologie, CNRS URA 1961, Institut Pasteur, Paris, France
| | | | | | | | | | | | | | | |
Collapse
|