1
|
Biswas M, So K, Bertolini TB, Krishnan P, Rana J, Muñoz-Melero M, Syed F, Kumar SRP, Gao H, Xuei X, Terhorst C, Daniell H, Cao S, Herzog RW. Distinct functions and transcriptional signatures in orally induced regulatory T cell populations. Front Immunol 2023; 14:1278184. [PMID: 37954612 PMCID: PMC10637621 DOI: 10.3389/fimmu.2023.1278184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Accepted: 10/16/2023] [Indexed: 11/14/2023] Open
Abstract
Oral administration of antigen induces regulatory T cells (Treg) that can not only control local immune responses in the small intestine, but also traffic to the central immune system to deliver systemic suppression. Employing murine models of the inherited bleeding disorder hemophilia, we find that oral antigen administration induces three CD4+ Treg subsets, namely FoxP3+LAP-, FoxP3+LAP+, and FoxP3-LAP+. These T cells act in concert to suppress systemic antibody production induced by therapeutic protein administration. Whilst both FoxP3+LAP+ and FoxP3-LAP+ CD4+ T cells express membrane-bound TGF-β (latency associated peptide, LAP), phenotypic, functional, and single cell transcriptomic analyses reveal distinct characteristics in the two subsets. As judged by an increase in IL-2Rα and TCR signaling, elevated expression of co-inhibitory receptor molecules and upregulation of the TGFβ and IL-10 signaling pathways, FoxP3+LAP+ cells are an activated form of FoxP3+LAP- Treg. Whereas FoxP3-LAP+ cells express low levels of genes involved in TCR signaling or co-stimulation, engagement of the AP-1 complex members Jun/Fos and Atf3 is most prominent, consistent with potent IL-10 production. Single cell transcriptomic analysis further reveals that engagement of the Jun/Fos transcription factors is requisite for mediating TGFβ expression. This can occur via an Il2ra dependent or independent process in FoxP3+LAP+ or FoxP3-LAP+ cells respectively. Surprisingly, both FoxP3+LAP+ and FoxP3-LAP+ cells potently suppress and induce FoxP3 expression in CD4+ conventional T cells. In this process, FoxP3-LAP+ cells may themselves convert to FoxP3+ Treg. We conclude that orally induced suppression is dependent on multiple regulatory cell types with complementary and interconnected roles.
Collapse
Affiliation(s)
- Moanaro Biswas
- Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Kaman So
- Department of Biostatistics and Health Data Science and Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Thais B. Bertolini
- Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Preethi Krishnan
- Department of Chemical and Biological Engineering, University of British Columbia, Vancouver, BC, Canada
| | - Jyoti Rana
- Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Maite Muñoz-Melero
- Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Farooq Syed
- Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Sandeep R. P. Kumar
- Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Hongyu Gao
- Center for Medical Genomics, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Xiaoling Xuei
- Center for Medical Genomics, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Cox Terhorst
- Division of Immunology, Beth Israel Deaconess Medical Center (BIDMC), Harvard Medical School, Boston, MA, United States
| | - Henry Daniell
- Department of Basic and Translational Sciences, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Sha Cao
- Department of Biostatistics and Health Data Science and Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Roland W. Herzog
- Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, United States
| |
Collapse
|
2
|
Takasaka N, Seed RI, Cormier A, Bondesson AJ, Lou J, Elattma A, Ito S, Yanagisawa H, Hashimoto M, Ma R, Levine MD, Publicover J, Potts R, Jespersen JM, Campbell MG, Conrad F, Marks JD, Cheng Y, Baron JL, Nishimura SL. Integrin αvβ8-expressing tumor cells evade host immunity by regulating TGF-β activation in immune cells. JCI Insight 2018; 3:122591. [PMID: 30333313 DOI: 10.1172/jci.insight.122591] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Accepted: 08/30/2018] [Indexed: 12/27/2022] Open
Abstract
TGF-β is a promising immunotherapeutic target. It is expressed ubiquitously in a latent form that must be activated to function. Determination of where and how latent TGF-β (L-TGF-β) is activated in the tumor microenvironment could facilitate cell- and mechanism-specific approaches to immunotherapeutically target TGF-β. Binding of L-TGF-β to integrin αvβ8 results in activation of TGF-β. We engineered and used αvβ8 antibodies optimized for blocking or detection, which - respectively - inhibit tumor growth in syngeneic tumor models or sensitively and specifically detect β8 in human tumors. Inhibition of αvβ8 potentiates cytotoxic T cell responses and recruitment of immune cells to tumor centers - effects that are independent of PD-1/PD-L1. β8 is expressed on the cell surface at high levels by tumor cells, not immune cells, while the reverse is true of L-TGF-β, suggesting that tumor cell αvβ8 serves as a platform for activating cell-surface L-TGF-β presented by immune cells. Transcriptome analysis of tumor-associated lymphoid cells reveals macrophages as a key cell type responsive to β8 inhibition with major increases in chemokine and tumor-eliminating genes. High β8 expression in tumor cells is seen in 20%-80% of various cancers, which rarely coincides with high PD-L1 expression. These data suggest tumor cell αvβ8 is a PD-1/PD-L1-independent immunotherapeutic target.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Yifan Cheng
- Department of Biochemistry and Biophysics, and.,Howard Hughes Medical Institute, UCSF, San Francisco, California, USA
| | | | | |
Collapse
|
3
|
Butler NS, Harris TH, Blader IJ. Regulation of immunopathogenesis during Plasmodium and Toxoplasma infections: more parallels than distinctions? Trends Parasitol 2013; 29:593-602. [PMID: 24184186 DOI: 10.1016/j.pt.2013.10.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2013] [Revised: 10/03/2013] [Accepted: 10/04/2013] [Indexed: 01/08/2023]
Abstract
Toxoplasma and Plasmodium parasites exact a significant toll on public health. Host immunity required for efficient control of infection by these Apicomplexans involves the induction of potent T cell responses, which sometimes results in immunopathological damage. Thus, protective immune responses must be balanced by regulatory networks that limit immunopathology. We review several key cellular and molecular immunoregulatory networks operational during Toxoplasma and Plasmodium infections. Accumulating data show that despite differences in how the immune response controls these parasites, many host immunoregulatory pathways and cellular networks are common to both. Thus, understanding the cellular and molecular circuits that prevent or regulate immunopathological responses against one parasite is likely to inform our understanding of the host response to the other parasite.
Collapse
Affiliation(s)
- Noah S Butler
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA.
| | | | | |
Collapse
|
4
|
Lebastchi AH, Khan SF, Qin L, Li W, Zhou J, Hibino N, Yi T, Rao DA, Pober JS, Tellides G. Transforming growth factor beta expression by human vascular cells inhibits interferon gamma production and arterial media injury by alloreactive memory T cells. Am J Transplant 2011; 11:2332-41. [PMID: 21812925 PMCID: PMC3203343 DOI: 10.1111/j.1600-6143.2011.03676.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Arteriosclerosis is characterized by the local activation of effector T cells leading to production of proinflammatory cytokines, such as IFN (interferon)-γ and IL-17, within the vessel wall. Conversely, the production of antiinflammatory cytokines, for example, TGF-β, by regulatory lymphocytes is known to inhibit both the differentiation of naïve T cells into effector T cells and the development of arteriosclerosis in murine models. We investigated the role of TGF-β on the alloreactivity of human effector memory T cells (Tem). Quiescent vascular cells, but not Tem, expressed TGF-β. Blockade of TGF-β activity in cocultures of CD4(+) Tem with allogeneic endothelial cells significantly increased IFN-γ, but not IL-17, secretion. Additionally, serologic neutralization of TGF-β in immunodeficient mouse hosts of human coronary artery grafts into which allogeneic human T cells were adoptively transferred resulted in heavier medial infiltration by Tem, greater loss of medial smooth muscle cells and increased IFN-γ production within the grafts without significantly reducing either intimal injury or IL-17 production. Protective effects of TGF-β may be limited by fewer TGF-β-expressing vascular cells within the intimal compartment, by a reduction in the expression of TGF-β by vascular cells in rejecting grafts, or possibly to less effective suppression of Tem than naïve T cells.
Collapse
Affiliation(s)
- Amir H. Lebastchi
- Department of Surgery, Interdepartmental Program in Vascular Biology and Therapeutics, Yale University School of Medicine, New Haven, Connecticut 06510
| | - Salman F. Khan
- Department of Surgery, Interdepartmental Program in Vascular Biology and Therapeutics, Yale University School of Medicine, New Haven, Connecticut 06510
| | - Lingfeng Qin
- Department of Surgery, Interdepartmental Program in Vascular Biology and Therapeutics, Yale University School of Medicine, New Haven, Connecticut 06510
| | - Wei Li
- Department of Surgery, Interdepartmental Program in Vascular Biology and Therapeutics, Yale University School of Medicine, New Haven, Connecticut 06510
| | - Jing Zhou
- Department of Surgery, Interdepartmental Program in Vascular Biology and Therapeutics, Yale University School of Medicine, New Haven, Connecticut 06510
| | - Narutoshi Hibino
- Department of Surgery, Interdepartmental Program in Vascular Biology and Therapeutics, Yale University School of Medicine, New Haven, Connecticut 06510
| | - Tai Yi
- Department of Immunobiology, Interdepartmental Program in Vascular Biology and Therapeutics, Yale University School of Medicine, New Haven, Connecticut 06510
| | - Deepak A. Rao
- Department of Immunobiology, Interdepartmental Program in Vascular Biology and Therapeutics, Yale University School of Medicine, New Haven, Connecticut 06510
| | - Jordan S. Pober
- Department of Immunobiology, Interdepartmental Program in Vascular Biology and Therapeutics, Yale University School of Medicine, New Haven, Connecticut 06510
| | - George Tellides
- Department of Surgery, Interdepartmental Program in Vascular Biology and Therapeutics, Yale University School of Medicine, New Haven, Connecticut 06510
,Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut 06516
| |
Collapse
|
5
|
Azhar M, Yin M, Bommireddy R, Duffy JJ, Yang J, Pawlowski SA, Boivin GP, Engle SJ, Sanford LP, Grisham C, Singh RR, Babcock GF, Doetschman T. Generation of mice with a conditional allele for transforming growth factor beta 1 gene. Genesis 2009; 47:423-31. [PMID: 19415629 DOI: 10.1002/dvg.20516] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Transforming growth factor beta1 (TGFbeta1) is a multifunctional growth factor involved in wound healing, tissue fibrosis, and in the pathogenesis of many syndromic diseases (e.g., Marfan syndrome, Camurati-Engelmann disease) and muscular, neurological, ophthalmic, cardiovascular and immunological disorders, and cancer. Since the generation of Tgfb1 knockout mice, there has been extraordinary progress in understanding its physiological and pathophysiological function. Here, we report the generation of a conditional knockout allele for Tgfb1 in which its exon 6 is flanked with LoxP sites. As proof of principle, we crossed these mice to LckCre transgenic mice and specifically disrupted Tgfb1 in T cells. The results indicate that T-cell-produced TGFbeta1 is required for normal in vivo regulation of peripheral T-cell activation, maintenance of T-cell homeostasis, and suppression of autoimmunity.
Collapse
Affiliation(s)
- Mohamad Azhar
- BIO5 Institute, University of Arizona, Tucson, Arizona, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
6
|
Carambia A, Herkel J. CD4 T cells in hepatic immune tolerance. J Autoimmun 2009; 34:23-8. [PMID: 19720498 DOI: 10.1016/j.jaut.2009.08.006] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2009] [Accepted: 08/13/2009] [Indexed: 12/20/2022]
Abstract
The liver features a unique immune microenvironment, which seems to favour immune tolerance, both locally and systemically. The hepatic microenvironment is formed by the unique anatomical structure of the liver sinusoids, a peculiar composition of antigen presenting cells and the relative abundance of anti-inflammatory cytokines. The outcome of T cell stimulation within the hepatic microenvironment is often tolerance. This is illustrated by the observations that antigen delivered to the portal vein, or allografts co-transplanted with allogeneic liver are not attacked by the immune system. Moreover, the tolerogenic properties of the liver seem to be part of the cause for the frequent persistence of hepatitis virus infections. This review summarizes some of the mechanisms of tolerance induction in the liver with a focus on CD4 T cells. Hepatic CD4 T cell tolerance seems to emerge from various tolerogenic mechanisms, including immune deviation from inflammatory to non-inflammatory effector function, a relative preponderance of negative co-stimulation notably through PD-1, generation and expansion of regulatory T cells, or the relative abundance of immunoinhibitory cytokines, such as inteleukin-10 and TGF-beta. Understanding the mechanisms of hepatic tolerance induction may teach us how to develop or improve therapies for inflammatory diseases of the liver and other organs. Indeed, novel therapeutic options that utilize hepatic tolerance mechanisms are beginning to emerge, such as the generation of Treg in the liver for therapy of autoimmune disease or the blockade of PD-1 for the therapy of chronic viral hepatitis.
Collapse
Affiliation(s)
- Antonella Carambia
- Department of Medicine I, University Medical Centre Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg, Germany.
| | | |
Collapse
|
7
|
Tinoco R, Alcalde V, Yang Y, Sauer K, Zuniga EI. Cell-intrinsic transforming growth factor-beta signaling mediates virus-specific CD8+ T cell deletion and viral persistence in vivo. Immunity 2009; 31:145-57. [PMID: 19604493 DOI: 10.1016/j.immuni.2009.06.015] [Citation(s) in RCA: 202] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2008] [Revised: 06/15/2009] [Accepted: 06/25/2009] [Indexed: 01/13/2023]
Abstract
Although deficient CD8(+) T cell responses have long been associated with chronic viral infections, the underlying mechanisms are still unclear. Here we report that sustained transforming growth factor-beta (TGF-beta) expression and phosphorylation of its signaling mediator, Smad-2, were distinctive features of virus-specific CD8(+) T cells during chronic versus acute viral infections in vivo. The result was TGF-beta-dependent apoptosis of virus-specific CD8(+) T cells that related to upregulation of the proapoptotic protein Bim during chronic infection. Moreover, selective attenuation of TGF-beta signaling in T cells increased the numbers and multiple functions of antiviral CD8(+) T cells and enabled rapid eradication of the persistence-prone virus and memory generation. Finally, we found that cell-intrinsic TGF-beta signaling was responsible for virus-specific-CD8(+) T cell apoptosis and decreased numbers but was not necessary for their functional exhaustion. Our findings reveal persisting TGF-beta-Smad signaling as a hallmark and key regulator of CD8(+) T cell responses during chronic viral infections in vivo.
Collapse
Affiliation(s)
- Roberto Tinoco
- Division of Biological Sciences, University of California San Diego, La Jolla, CA 92093-0322, USA
| | | | | | | | | |
Collapse
|
8
|
Perruche S, Zhang P, Maruyama T, Bluestone JA, Saas P, Chen W. Lethal effect of CD3-specific antibody in mice deficient in TGF-beta1 by uncontrolled flu-like syndrome. THE JOURNAL OF IMMUNOLOGY 2009; 183:953-61. [PMID: 19561097 DOI: 10.4049/jimmunol.0804076] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
CD3-specific Ab therapy results in a transient, self-limiting, cytokine-associated, flu-like syndrome in experimental animals and in patients, but the underlying mechanism for this spontaneous resolution remains elusive. By using an in vivo model of CD3-specific Ab-induced flu-like syndrome, we show in this paper that a single injection of sublethal dose of the Ab killed all TGF-beta1(-/-) mice. The death of TGF-beta1(-/-) mice was associated with occurrence of this uncontrolled flu-like syndrome, as demonstrated by a sustained storm of systemic inflammatory TNF and IFN-gamma cytokines. We present evidence that deficiency of professional phagocytes to produce TGF-beta1 after apoptotic T cell clearance may be responsible, together with hypersensitivity of T cells to both activation and apoptosis, for the uncontrolled inflammation. These findings indicate a key role for TGF-beta1 and phagocytes in protecting the recipients from lethal inflammation and resolving the flu-like syndrome after CD3-specific Ab treatment. The study may also provide a novel molecular mechanism explaining the early death in TGF-beta1(-/-) mice.
Collapse
Affiliation(s)
- Sylvain Perruche
- Mucosal Immunology Unit, Oral Infection and Immunity Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | | | | | |
Collapse
|
9
|
Ingman WV, Robertson SA. The essential roles of TGFB1 in reproduction. Cytokine Growth Factor Rev 2009; 20:233-9. [PMID: 19497778 DOI: 10.1016/j.cytogfr.2009.05.003] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Transforming growth factor beta 1 (TGFB1) is implicated as a key regulator of the development and cyclic remodelling characteristic of reproductive tissues. The physiological significance of TGFB1 in reproductive biology and fertility has been extensively examined in Tgfb1 null mutant mice. Genetic deficiency in TGFB1 causes perturbed functioning of the hypothalamic-pituitary-gonadal axis, inhibiting luteinising hormone (LH) synthesis and leading to downstream effects on testosterone production in males and estrous cycle abnormalities in females. Oocyte developmental incompetence, accompanied by early embryo arrest as well as altered pubertal mammary gland morphogenesis are observed. In addition to LH and testosterone deficiency, male Tgfb1 null mice demonstrate complete inability to mate with females, associated with failure to initiate and/or sustain successful penile intromission or ejaculation. These studies demonstrate the profound significance of TGFB1 in male and female reproductive physiology, and provide a foundation for exploring the significance of this cytokine in human infertility and sexual dysfunction.
Collapse
Affiliation(s)
- Wendy V Ingman
- Discipline Obstetrics and Gynaecology and Research Centre for Reproductive Health, University of Adelaide, South Australia 5005, Australia.
| | | |
Collapse
|
10
|
Stavnezer J, Kang J. The surprising discovery that TGF beta specifically induces the IgA class switch. THE JOURNAL OF IMMUNOLOGY 2009; 182:5-7. [PMID: 19109126 DOI: 10.4049/jimmunol.182.1.5] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Affiliation(s)
- Janet Stavnezer
- Department of Molecular Genetics and Microbiology, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | | |
Collapse
|
11
|
McGrath LJ, Ingman WV, Robker RL, Robertson SA. Exogenous transforming growth factor beta1 replacement and fertility in male Tgfb1 null mutant mice. Reprod Fertil Dev 2009; 21:561-70. [DOI: 10.1071/rd08294] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2008] [Accepted: 02/02/2009] [Indexed: 11/23/2022] Open
Abstract
Analysis of Tgfb1 null mutant mice has demonstrated that the cytokine transforming growth factor β1 (TGFB1) has essential non-redundant roles in fertility. The present study attempted to alleviate the infertility phenotype of Tgfb1 null mutant male mice by administration of exogenous TGFB1, either orally by colostrum feeding or subcutaneously by delivery of recombinant human latent TGFB1 (rhLTGFB1) via osmotic mini-pumps. Bovine colostrum and fresh unpasteurised bovine milk were found to be rich sources of TGFB1 and TGFB2; however, feeding Tgfb1 null mutant mice colostrum for 2 days failed to raise serum levels of TGFB1. Administration of rhLTGFB1 (~150 μg in total) over 14 days to Tgfb1 null mutant mice resulted in detectable TGFB1 in serum; however, mean levels remained 10-fold less than in Tgfb1 heterozygous mice. After 7 days and 14 days of rhLTGFB1 administration, serum testosterone, spontaneous non-contact erections and mating behaviour were assessed. Despite the increased serum TGFB1, administration of rhLTGFB1 to Tgfb1 null mutant mice failed to improve these fertility parameters. It is concluded that sustained restoration of circulating latent TGFB1 to levels approaching the normal physiological range does not rescue the infertility phenotype caused by TGFB1 deficiency. Reproductive function in male Tgfb1 null mutant mice may not respond to systemic TGFB1 supplementation due to a requirement for local sources of TGFB1 at the site of action in the reproductive tract, or perturbed development during the neonatal period or puberty such that adult reproductive function is permanently impaired.
Collapse
|
12
|
Abstract
A functional adaptive immune system depends on a diverse and self-tolerant population of T lymphocytes that are generated in the thymus and maintained in the peripheral lymphoid organs. Recent studies have defined the cytokine transforming growth factor-beta (TGF-beta) as a critical regulator of thymic T cell development as well as a crucial player in peripheral T cell homeostasis, tolerance to self antigens, and T cell differentiation during the immune response. The unique mechanism of TGF-beta activation and the plasticity of TGF-beta signaling create a stage for TGF-beta to integrate signals from multiple cell types and environmental cues to regulate T cells.
Collapse
|
13
|
Li MO, Flavell RA. Contextual regulation of inflammation: a duet by transforming growth factor-beta and interleukin-10. Immunity 2008; 28:468-76. [PMID: 18400189 DOI: 10.1016/j.immuni.2008.03.003] [Citation(s) in RCA: 369] [Impact Index Per Article: 23.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Transforming growth factor-beta (TGF-beta) and interleukin-10 (IL-10) are regulatory cytokines with pleiotropic roles in the immune system. The prominent function of TGF-beta is to maintain T cell tolerance to self or innocuous environmental antigens via its direct effects on the differentiation and homeostasis of effector and regulatory T cells. A critical route for the regulation of T cells by TGF-beta is via activation of a T cell-produced latent form of TGF-beta1 by dendritic cell-expressed avbeta8 integrin. IL-10 operates primarily as a feedback inhibitor of exuberant T cell responses to microbial antigens. T cells are also the principal producers of IL-10, the expression of which is regulated by IL-27, IL-6, and TGF-beta. The collective activity of TGF-beta and IL-10 ensures a controlled inflammatory response specifically targeting pathogens without evoking excessive immunopathology to self-tissues.
Collapse
Affiliation(s)
- Ming O Li
- Immunology Program, Memorial Sloan-Kettering Cancer Center, New York, NY 10065, USA.
| | | |
Collapse
|
14
|
Li MO, Wan YY, Flavell RA. T cell-produced transforming growth factor-beta1 controls T cell tolerance and regulates Th1- and Th17-cell differentiation. Immunity 2007; 26:579-91. [PMID: 17481928 DOI: 10.1016/j.immuni.2007.03.014] [Citation(s) in RCA: 543] [Impact Index Per Article: 31.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2007] [Revised: 03/05/2007] [Accepted: 03/19/2007] [Indexed: 12/21/2022]
Abstract
TGF-beta1 is a regulatory cytokine with a pleiotropic role in immune responses. TGF-beta1 is widely expressed in leukocytes and stromal cells. However, the functions of TGF-beta1 expressed by specific lineages of cells remain unknown in vivo. Here, we show that mice with a T cell-specific deletion of the Tgfb1 gene developed lethal immunopathology in multiple organs, and this development was associated with enhanced T cell proliferation, activation, and CD4+ T cell differentiation into T helper 1 (Th1) and Th2 cells. TGF-beta1 produced by Foxp3-expressing regulatory T cells was required to inhibit Th1-cell differentiation and inflammatory-bowel disease in a transfer model. In addition, T cell-produced TGF-beta1 promoted Th17-cell differentiation and was indispensable for the induction of experimental autoimmune encephalomyelitis. These findings reveal essential roles for T cell-produced TGF-beta1 in controlling differentiation of T helper cells and controlling inflammatory diseases.
Collapse
MESH Headings
- Animals
- Cell Differentiation/immunology
- Cells, Cultured
- Colitis/genetics
- Colitis/immunology
- Colitis/metabolism
- Colitis/pathology
- Encephalomyelitis, Autoimmune, Experimental/genetics
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/metabolism
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Forkhead Transcription Factors/metabolism
- Gene Deletion
- Gene Expression Regulation
- Homeostasis
- Immune Tolerance/immunology
- Lymphocyte Activation/immunology
- Mice
- Mice, Knockout
- T-Lymphocytes/cytology
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- Transforming Growth Factor beta1/biosynthesis
- Transforming Growth Factor beta1/genetics
Collapse
Affiliation(s)
- Ming O Li
- Section of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | | | | |
Collapse
|
15
|
Brionne TC, Tesseur I, Masliah E, Wyss-Coray T. Loss of TGF-beta 1 leads to increased neuronal cell death and microgliosis in mouse brain. Neuron 2004; 40:1133-45. [PMID: 14687548 DOI: 10.1016/s0896-6273(03)00766-9] [Citation(s) in RCA: 278] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
TGF-beta1 is a key regulator of diverse biological processes in many tissues and cell types, but its exact function in the developing and adult mammalian CNS is still unknown. We report that lack of TGF-beta1 expression in neonatal Tgfb1(-/-) mice results in a widespread increase in degenerating neurons accompanied by reduced expression of synaptophysin and laminin and a prominent microgliosis. Lack of TGF-beta1 also strongly reduces survival of primary neurons cultured from Tgfb1(-/-) mice. TGF-beta1 deficiency in adult Tgfb1(-/+) mice results in increased neuronal susceptibility to excitotoxic injury, whereas astroglial overexpression of TGF-beta1 protects adult mice against neurodegeneration in acute, excitotoxic and chronic injury paradigms. This study reveals a nonredundant function for TGF-beta1 in maintaining neuronal integrity and survival of CNS neurons and in regulating microglial activation. Because individual TGF-beta1 expression levels in the brain vary considerably between humans, this finding could have important implications for susceptibility to neurodegeneration.
Collapse
Affiliation(s)
- Thomas C Brionne
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA
| | | | | | | |
Collapse
|
16
|
Ruzek MC, Hawes M, Pratt B, McPherson J, Ledbetter S, Richards SM, Garman RD. Minimal effects on immune parameters following chronic anti-TGF-beta monoclonal antibody administration to normal mice. Immunopharmacol Immunotoxicol 2003; 25:235-57. [PMID: 12784916 DOI: 10.1081/iph-120020473] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Mice genetically deficient in TGF-beta1 or TGF-beta signaling capacity in T or B cells demonstrate profound immune dysregulation, as evidenced by increased lymph node size, expression of markers of memory/activation on T cells, inflammation in a variety of tissues and development of autoantibodies. However, this constant and complete lack of TGF-beta1 or TGF-betaR signaling may not reflect effects of TGF-beta neutralization using antibodies in mature animals. Thus, the present studies were designed to determine if administration of an anti-TGF-beta monoclonal antibody (neutralizes TGF-beta1, 2 and 3) to mature, normal mice results in evidence of immune dysregulation or immune-mediated pathology. An initial study examined daily administration of 0.25, 0.75 and 2.5 mg/kg of anti-TGF-beta to mice for three weeks, achieving blood levels of as high as 9 mg/ml. Comprehensive hematological and histopathological evaluation showed no evidence of pathology. A second study was designed to extend the antibody treatment period and further examine the functional status of the immune system. Mice were injected with 1 mg/mouse (approximately 50 mg/kg) of anti-TGF-beta (1D11) three times per week achieving circulating blood levels of 1-2 mg/ml. Many parameters of immune status were assessed, including natural killer (NK) cell activity, lymphocyte proliferative responses, phagocytic activity, phenotypic assessment of leukocyte subsets, and serum measurements of proinflammatory cytokines, autoantibodies and immunoglobulin isotypes. In addition, histopathological assessment of heart, lungs, liver, kidney, salivary glands, skin, spleen and lymph nodes was also performed. Very few of the multiple immune parameters examined showed detectable changes in anti-TGF-beta-treated mice. Changes that were observed were primarily restricted to the spleen and included increased spleen cell recoveries, increased percentages of macrophages, decreased percentages of NK cells, decreased phagocytic activity, decreased proliferative responses to mitogens and slight increases in T and B cells displaying an activated phenotype. Many of these same parameters examined in the lymph nodes were not altered by the anti-TGF-beta treatment. The thymus was decreased in size, but altered only slightly in one population of developing T cells. Most of the changes observed were modest and returned to control levels after discontinuation of treatments. The only serological finding was an increase in IgA levels in anti-TGF-beta-treated mice, but not in any other isotype. Finally, there was no evidence of increased inflammation in any of the peripheral tissues examined in the anti-TGF-beta-treated mice. In conclusion, although there were changes in some of the immunological parameters examined in these studies, they were few and typically reversed following discontinuation of treatment. The modest nature of the changes observed in these studies is particularly evident when compared to published data of those same parameters examined in mice genetically deficient in TGF-beta1 or mice having TGF-beta unresponsive T or B cells. Thus, there does not appear to be any significant immune dysregulation detectable after long-term antibody-mediated neutralization of TGF-beta in normal mice.
Collapse
Affiliation(s)
- Melanie C Ruzek
- Cell and Protein Therapeutics R&D, Genzyme Corporation, Framingham, Massachusetts 01701-9322, USA.
| | | | | | | | | | | | | |
Collapse
|