1
|
Nyanyo DD, Mikamoto M, Galbiati F, Remba-Shapiro I, Bode K, Schoenfeld S, Jones PS, Swearingen B, Nachtigall LB. Autoimmune Disorders Associated With Surgical Remission of Cushing's Disease : A Cohort Study. Ann Intern Med 2024; 177:315-323. [PMID: 38373302 DOI: 10.7326/m23-2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/21/2024] Open
Abstract
BACKGROUND Glucocorticoids suppress inflammation. Autoimmune disease may occur after remission of Cushing's disease (CD). However, the development of autoimmune disease in this context is not well described. OBJECTIVE To determine 1) the incidence of autoimmune disease in patients with CD after surgical remission compared with patients with nonfunctioning pituitary adenomas (NFPAs) and 2) the clinical presentation of and risk factors for development of autoimmune disease in CD after remission. DESIGN Retrospective matched cohort analysis. SETTING Academic medical center/pituitary center. PATIENTS Patients with CD with surgical remission and surgically treated NFPA. MEASUREMENTS Cumulative incidence of new-onset autoimmune disease at 3 years after surgery. Assessment for hypercortisolemia included late-night salivary cortisol levels, 24-hour urine free cortisol (UFC) ratio (UFC value divided by the upper limit of the normal range for the assay), and dexamethasone suppression tests. RESULTS Cumulative incidence of new-onset autoimmune disease at 3 years after surgery was higher in patients with CD (10.4% [95% CI, 5.7% to 15.1%]) than in those with NFPAs (1.6% [CI, 0% to 4.6%]) (hazard ratio, 7.80 [CI, 2.88 to 21.10]). Patients with CD showed higher prevalence of postoperative adrenal insufficiency (93.8% vs. 16.5%) and lower postoperative nadir serum cortisol levels (63.8 vs. 282.3 nmol/L) than patients with NFPAs. Compared with patients with CD without autoimmune disease, those who developed autoimmune disease had a lower preoperative 24-hour UFC ratio (2.7 vs. 6.3) and a higher prevalence of family history of autoimmune disease (41.2% vs. 20.9%). LIMITATION The small sample of patients with autoimmune disease limited identification of independent risk factors. CONCLUSION Patients achieving surgical remission of CD have higher incidence of autoimmune disease than age- and sex-matched patients with NFPAs. Family history of autoimmune disease is a potential risk factor. Adrenal insufficiency may be a trigger. PRIMARY FUNDING SOURCE Recordati Rare Diseases Inc.
Collapse
Affiliation(s)
- Dennis Delasi Nyanyo
- The Neuroendocrine Unit, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts (D.D.N., F.G., I.R., K.B., L.B.N.)
| | - Masaaki Mikamoto
- The Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts (M.M., P.S.J., B.S.)
| | - Francesca Galbiati
- The Neuroendocrine Unit, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts (D.D.N., F.G., I.R., K.B., L.B.N.)
| | - Ilan Remba-Shapiro
- The Neuroendocrine Unit, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts (D.D.N., F.G., I.R., K.B., L.B.N.)
| | - Kevin Bode
- The Neuroendocrine Unit, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts (D.D.N., F.G., I.R., K.B., L.B.N.)
| | - Sara Schoenfeld
- The Division of Rheumatology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts (S.S.)
| | - Pamela S Jones
- The Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts (M.M., P.S.J., B.S.)
| | - Brooke Swearingen
- The Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts (M.M., P.S.J., B.S.)
| | - Lisa B Nachtigall
- The Neuroendocrine Unit, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts (D.D.N., F.G., I.R., K.B., L.B.N.)
| |
Collapse
|
2
|
Riedel JH, Robben L, Paust HJ, Zhao Y, Asada N, Song N, Peters A, Kaffke A, Borchers A, Tiegs G, Seifert L, Tomas NM, Hoxha E, Wenzel UO, Huber TB, Wiech T, Turner JE, Krebs CF, Panzer U. Glucocorticoids target the CXCL9/CXCL10-CXCR3 axis and confer protection against immune-mediated kidney injury. JCI Insight 2023; 8:160251. [PMID: 36355429 PMCID: PMC9870076 DOI: 10.1172/jci.insight.160251] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 11/09/2022] [Indexed: 11/12/2022] Open
Abstract
Glucocorticoids remain a cornerstone of therapeutic regimes for autoimmune and chronic inflammatory diseases - for example, in different forms of crescentic glomerulonephritis - because of their rapid antiinflammatory effects, low cost, and wide availability. Despite their routine use for decades, the underlying cellular mechanisms by which steroids exert their therapeutic effects need to be fully elucidated. Here, we demonstrate that high-dose steroid treatment rapidly reduced the number of proinflammatory CXCR3+CD4+ T cells in the kidney by combining high-dimensional single-cell and morphological analyses of kidney biopsies from patients with antineutrophil cytoplasmic antibody-associated (ANCA-associated) crescentic glomerulonephritis. Using an experimental model of crescentic glomerulonephritis, we show that the steroid-induced decrease in renal CD4+ T cells is a consequence of reduced T cell recruitment, which is associated with an ameliorated disease course. Mechanistic in vivo and in vitro studies revealed that steroids act directly on renal tissue cells, such as tubular epithelial cells, but not on T cells, which resulted in an abolished renal expression of CXCL9 and CXCL10 as well as in the prevention of CXCR3+CD4+ T cell recruitment to the inflamed kidneys. Thus, we identified the CXCL9/CXCL10-CXCR3 axis as a previously unrecognized cellular and molecular target of glucocorticoids providing protection from immune-mediated pathology.
Collapse
Affiliation(s)
- Jan-Hendrik Riedel
- Division of Translational Immunology, III. Department of Medicine and,III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Lennart Robben
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | - Yu Zhao
- Division of Translational Immunology, III. Department of Medicine and,Institute of Medical Systems Biology, Center for Molecular Neurobiology Hamburg (ZMNH), Hamburg, Germany
| | - Nariaki Asada
- Division of Translational Immunology, III. Department of Medicine and
| | - Ning Song
- Division of Translational Immunology, III. Department of Medicine and
| | - Anett Peters
- Division of Translational Immunology, III. Department of Medicine and
| | - Anna Kaffke
- Division of Translational Immunology, III. Department of Medicine and
| | - Alina Borchers
- Division of Translational Immunology, III. Department of Medicine and
| | - Gisa Tiegs
- Institute of Experimental Immunology and Hepatology,,Institute of Pathology, Section of Nephropathology, and
| | - Larissa Seifert
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Nicola M. Tomas
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Institute of Pathology, Section of Nephropathology, and
| | - Elion Hoxha
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ulrich O. Wenzel
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Tobias B. Huber
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | - Jan-Eric Turner
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christian F. Krebs
- Division of Translational Immunology, III. Department of Medicine and,III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ulf Panzer
- Division of Translational Immunology, III. Department of Medicine and,III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
3
|
Salehzadeh M, Soma KK. Glucocorticoid production in the thymus and brain: Immunosteroids and neurosteroids. Brain Behav Immun Health 2021; 18:100352. [PMID: 34988497 PMCID: PMC8710407 DOI: 10.1016/j.bbih.2021.100352] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 09/05/2021] [Accepted: 09/17/2021] [Indexed: 12/23/2022] Open
Abstract
Glucocorticoids (GCs) regulate a myriad of physiological systems, such as the immune and nervous systems. Systemic GC levels in blood are often measured as an indicator of local GC levels in target organs. However, several extra-adrenal organs can produce and metabolize GCs locally. More sensitive and specific methods for GC analysis (i.e., mass spectrometry) allow measurement of local GC levels in small tissue samples with low GC concentrations. Consequently, is it now apparent that systemic GC levels often do not reflect local GC levels. Here, we review the use of systemic GC measurements in clinical and research settings, discuss instances where systemic GC levels do not reflect local GC levels, and present evidence that local GC levels provide useful insights, with a focus on local GC production in the thymus (immunosteroids) and brain (neurosteroids). Lastly, we suggest key areas for further research, such as the roles of immunosteroids and neurosteroids in neonatal programming and the potential clinical relevance of local GC modulators.
Collapse
Affiliation(s)
- Melody Salehzadeh
- Department of Zoology, University of British Columbia, Vancouver, BC, Canada
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| | - Kiran K Soma
- Department of Zoology, University of British Columbia, Vancouver, BC, Canada
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
- Department of Psychology, University of British Columbia, Vancouver, BC, Canada
- Graduate Program in Neuroscience, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
4
|
Pérez AR, de Meis J, Rodriguez-Galan MC, Savino W. The Thymus in Chagas Disease: Molecular Interactions Involved in Abnormal T-Cell Migration and Differentiation. Front Immunol 2020; 11:1838. [PMID: 32983098 PMCID: PMC7492291 DOI: 10.3389/fimmu.2020.01838] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Accepted: 07/08/2020] [Indexed: 12/24/2022] Open
Abstract
Chagas disease, caused by the protozoan parasite T. cruzi, is a prevalent parasitic disease in Latin America. Presently, it is spreading around the world by human migration, thus representing a new global health issue. Chronically infected individuals reveal a dissimilar disease progression: while nearly 60% remain without apparent disease for life, 30% develop life-threatening pathologies, such as chronic chagasic cardiomyopathy (CCC) or megaviscerae. Inflammation driven by parasite persistence seems to be involved in the pathophysiology of the disease. However, there is also evidence of the occurrence of autoimmune events, mainly caused by molecular mimicry and bystander activation. In experimental models of disease, is well-established that T. cruzi infects the thymus and causes locally profound structural and functional alterations. The hallmark is a massive loss of CD4+CD8+ double positive (DP) thymocytes, mainly triggered by increased levels of glucocorticoids, although other mechanisms seem to act simultaneously. Thymic epithelial cells (TEC) exhibited an increase in extracellular matrix deposition, which are related to thymocyte migratory alterations. Moreover, medullary TEC showed a decreased expression of AIRE and altered expression of microRNAs, which might be linked to a disrupted negative selection of the T-cell repertoire. Also, almost all stages of thymocyte development are altered, including an abnormal output of CD4−CD8− double negative (DN) and DP immature and mature cells, many of them carrying prohibited TCR-Vβ segments. Evidence has shown that DN and DP cells with an activated phenotype can be tracked in the blood of humans with chronic Chagas disease and also in the secondary lymphoid organs and heart of infected mice, raising new questions about the relevance of these populations in the pathogenesis of Chagas disease and their possible link with thymic alterations and an immunoendocrine imbalance. Here, we discuss diverse molecular mechanisms underlying thymic abnormalities occurring during T. cruzi infection and their link with CCC, which may contribute to the design of innovative strategies to control Chagas disease pathology.
Collapse
Affiliation(s)
- Ana Rosa Pérez
- Instituto de Inmunología Clínica y Experimental de Rosario, CONICET-Universidad Nacional de Rosario, Rosario, Argentina.,Centro de Investigación y Producción de Reactivos Biológicos, Facultad de Ciencias Médicas, Universidad Nacional de Rosario, Rosario, Argentina
| | - Juliana de Meis
- Laboratory on Thymus Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil.,National Institute of Science and Technology on Neuroimmunomodulation, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil.,Rio de Janeiro Research Network on Neuroinflammation, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | | | - Wilson Savino
- Laboratory on Thymus Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil.,National Institute of Science and Technology on Neuroimmunomodulation, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil.,Rio de Janeiro Research Network on Neuroinflammation, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| |
Collapse
|
5
|
Kinsella S, Dudakov JA. When the Damage Is Done: Injury and Repair in Thymus Function. Front Immunol 2020; 11:1745. [PMID: 32903477 PMCID: PMC7435010 DOI: 10.3389/fimmu.2020.01745] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 06/30/2020] [Indexed: 01/02/2023] Open
Abstract
Even though the thymus is exquisitely sensitive to acute insults like infection, shock, or common cancer therapies such as cytoreductive chemo- or radiation-therapy, it also has a remarkable capacity for repair. This phenomenon of endogenous thymic regeneration has been known for longer even than its primary function to generate T cells, however, the underlying mechanisms controlling the process have been largely unstudied. Although there is likely continual thymic involution and regeneration in response to stress and infection in otherwise healthy people, acute and profound thymic damage such as that caused by common cancer cytoreductive therapies or the conditioning regimes as part of hematopoietic cell transplantation (HCT), leads to prolonged T cell deficiency; precipitating high morbidity and mortality from opportunistic infections and may even facilitate cancer relapse. Furthermore, this capacity for regeneration declines with age as a function of thymic involution; which even at steady state leads to reduced capacity to respond to new pathogens, vaccines, and immunotherapy. Consequently, there is a real clinical need for strategies that can boost thymic function and enhance T cell immunity. One approach to the development of such therapies is to exploit the processes of endogenous thymic regeneration into novel pharmacologic strategies to boost T cell reconstitution in clinical settings of immune depletion such as HCT. In this review, we will highlight recent work that has revealed the mechanisms by which the thymus is capable of repairing itself and how this knowledge is being used to develop novel therapies to boost immune function.
Collapse
Affiliation(s)
- Sinéad Kinsella
- Program in Immunology, Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States
| | - Jarrod A. Dudakov
- Program in Immunology, Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States
- Immunotherapy Integrated Research Center, Fred Hutchinson Cancer Research Center, Seattle, WA, United States
- Department of Immunology, University of Washington, Seattle, WA, United States
| |
Collapse
|
6
|
Zheng Z, Ai J, Guo L, Ye X, Bondada S, Howatt D, Daugherty A, Li XA. SR-BI (Scavenger Receptor Class B Type 1) Is Critical in Maintaining Normal T-Cell Development and Enhancing Thymic Regeneration. Arterioscler Thromb Vasc Biol 2019; 38:2706-2717. [PMID: 30354229 DOI: 10.1161/atvbaha.118.311728] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Objective- Continuous T-cell production from thymus is essential in replenishing naïve T-cell pool and maintaining optimal T-cell functions. However, the underlying mechanisms regulating the T-cell development in thymus remains largely unknown. Approach and Results- We identified SR-BI (scavenger receptor class B type 1), an HDL (high-density lipoprotein) receptor, as a novel modulator in T-cell development. We found that SR-BI deficiency in mice led to reduced thymus size and decreased T-cell production, which was accompanied by narrowed peripheral naïve T-cell pool. Further investigation revealed that SR-BI deficiency impaired progenitor thymic homing, causing a dramatic reduction in the percentage of earliest thymic progenitors, but did not affect other downstream T-cell developmental steps inside the thymus. As a result of the impaired progenitor thymic homing, SR-BI-deficient mice displayed delayed thymic regeneration postirradiation. Using a variety of experimental approaches, we revealed that the impaired T-cell development in SR-BI-deficient mice was not caused by hematopoietic SR-BI deficiency or SR-BI deficiency-induced hypercholesterolemia, but mainly attributed to the SR-BI deficiency in adrenal glands, as adrenal-specific SR-BI-deficient mice exhibited similar defects in T-cell development and thymic regeneration with SR-BI-deficient mice. Conclusions- This study demonstrates that SR-BI deficiency impaired T-cell development and delayed thymic regeneration by affecting progenitor thymic homing in mice, elucidating a previously unrecognized link between SR-BI and adaptive immunity.
Collapse
Affiliation(s)
- Zhong Zheng
- From the Department of Pharmacology and Nutritional Sciences (Z.Z., J.A., X.-A.L.), University of Kentucky College of Medicine, Lexington.,Saha Cardiovascular Research Center (Z.Z., J.A., L.G., X.Y., D.H., A.D., X.-A.L.), University of Kentucky College of Medicine, Lexington
| | - Junting Ai
- From the Department of Pharmacology and Nutritional Sciences (Z.Z., J.A., X.-A.L.), University of Kentucky College of Medicine, Lexington.,Saha Cardiovascular Research Center (Z.Z., J.A., L.G., X.Y., D.H., A.D., X.-A.L.), University of Kentucky College of Medicine, Lexington
| | - Ling Guo
- Saha Cardiovascular Research Center (Z.Z., J.A., L.G., X.Y., D.H., A.D., X.-A.L.), University of Kentucky College of Medicine, Lexington
| | - Xiang Ye
- Saha Cardiovascular Research Center (Z.Z., J.A., L.G., X.Y., D.H., A.D., X.-A.L.), University of Kentucky College of Medicine, Lexington
| | - Subbarao Bondada
- Department of Microbiology (S.B.), University of Kentucky College of Medicine, Lexington
| | - Deborah Howatt
- Saha Cardiovascular Research Center (Z.Z., J.A., L.G., X.Y., D.H., A.D., X.-A.L.), University of Kentucky College of Medicine, Lexington
| | - Alan Daugherty
- Saha Cardiovascular Research Center (Z.Z., J.A., L.G., X.Y., D.H., A.D., X.-A.L.), University of Kentucky College of Medicine, Lexington.,Department of Physiology (A.D., X.-A.L.), University of Kentucky College of Medicine, Lexington
| | - Xiang-An Li
- From the Department of Pharmacology and Nutritional Sciences (Z.Z., J.A., X.-A.L.), University of Kentucky College of Medicine, Lexington.,Saha Cardiovascular Research Center (Z.Z., J.A., L.G., X.Y., D.H., A.D., X.-A.L.), University of Kentucky College of Medicine, Lexington.,Department of Physiology (A.D., X.-A.L.), University of Kentucky College of Medicine, Lexington
| |
Collapse
|
7
|
Bereshchenko O, Bruscoli S, Riccardi C. Glucocorticoids, Sex Hormones, and Immunity. Front Immunol 2018; 9:1332. [PMID: 29946321 PMCID: PMC6006719 DOI: 10.3389/fimmu.2018.01332] [Citation(s) in RCA: 183] [Impact Index Per Article: 26.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Accepted: 05/29/2018] [Indexed: 12/15/2022] Open
Abstract
Glucocorticoid hormones regulate essential body functions in mammals, control cell metabolism, growth, differentiation, and apoptosis. Importantly, they are potent suppressors of inflammation, and multiple immune-modulatory mechanisms involving leukocyte apoptosis, differentiation, and cytokine production have been described. Due to their potent anti-inflammatory and immune-suppressive activity, synthetic glucocorticoids (GCs) are the most prescribed drugs used for treatment of autoimmune and inflammatory diseases. It is long been noted that males and females exhibit differences in the prevalence in several autoimmune diseases (AD). This can be due to the role of sexual hormones in regulation of the immune responses, acting through their endogenous nuclear receptors to mediate gene expression and generate unique gender-specific cellular environments. Given the fact that GCs are the primary physiological anti-inflammatory hormones, and that sex hormones may also exert immune-modulatory functions, the link between GCs and sex hormones may exist. Understanding the nature of this possible crosstalk is important to unravel the reason of sexual disparity in AD and to carefully prescribe these drugs for the treatment of inflammatory diseases. In this review, we discuss similarities and differences between the effects of sex hormones and GCs on the immune system, to highlight possible axes of functional interaction.
Collapse
Affiliation(s)
- Oxana Bereshchenko
- Section of Pharmacology, Department of Medicine, University of Perugia, Perugia, Italy.,Department of Surgery and Biomedical Sciences, University of Perugia, Perugia, Italy
| | - Stefano Bruscoli
- Section of Pharmacology, Department of Medicine, University of Perugia, Perugia, Italy
| | - Carlo Riccardi
- Section of Pharmacology, Department of Medicine, University of Perugia, Perugia, Italy
| |
Collapse
|
8
|
Liu Z, Su DM, Yu ZL, Wu F, Liu RF, Luo SQ, Lv ZY, Zeng X, Sun X, Wu ZD. Soluble antigens from the neurotropic pathogen Angiostrongylus cantonensis directly induce thymus atrophy in a mouse model. Oncotarget 2018; 8:48575-48590. [PMID: 28548945 PMCID: PMC5564709 DOI: 10.18632/oncotarget.17836] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Accepted: 05/02/2017] [Indexed: 11/25/2022] Open
Abstract
The nematode Angiostrongylus cantonensis (A.C.) is a neurotropic pathogen; stage-III larva invade the human (non-permissive host) central nervous system (CNS) to cause eosinophilic meningitis or meningoencephalitis accompanied by immunosuppression. In an A.C.-infectedmouse (another non-permissive host) model, CNS damage-associated T cell immune deficiency and severe inflammation were proposed to result from activation of the hypothalamic-pituitary-adrenal (HPA) axis. However, glucocorticoids are anti-inflammatory agents. Additionally, while defects in thymic stromal/epithelial cells (TECs) are the major reason for thymic atrophy, TECs do not express the glucocorticoid receptor. Therefore, activation of the HPA axis cannot fully explain the thymic atrophy and inflammation. Using an A.C.-infected mouse model, we found that A.C.-infected mice developed severe thymic atrophy with dramatic impairments in thymocytes and TECs, particularly cortical TECs, which harbor CD4+CD8+ double-positive thymocytes. The impairments resulted from soluble antigens (sAgs) from A.C. in the thymuses of infected mice, as intrathymic injection of these sAgs into live mice and the addition of these sAgs to thymic cell culture resulted in thymic atrophy and cellular apoptosis, respectively. Therefore, in addition to an indirect effect on thymocytes through the HPA axis, our study reveals a novel mechanism by which A.C. infection in non-permissive hosts directly induces defects in both thymocytes and TECs via soluble antigens.
Collapse
Affiliation(s)
- Zhen Liu
- Department of Parasitology of Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China.,Key Laboratory of Tropical Disease Control, Ministry of Education, Guangzhou, China.,Provincial Engineering Technology Research Center for Diseases-Vectors Control, Guangzhou, China
| | - Dong-Ming Su
- Institute for Molecular Medicine, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Zi-Long Yu
- Department of Parasitology of Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China.,Key Laboratory of Tropical Disease Control, Ministry of Education, Guangzhou, China.,Provincial Engineering Technology Research Center for Diseases-Vectors Control, Guangzhou, China
| | - Feng Wu
- Department of Parasitology of Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China.,Key Laboratory of Tropical Disease Control, Ministry of Education, Guangzhou, China.,Provincial Engineering Technology Research Center for Diseases-Vectors Control, Guangzhou, China
| | - Rui-Feng Liu
- Department of Parasitology of Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Shi-Qi Luo
- Department of Parasitology of Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China.,Key Laboratory of Tropical Disease Control, Ministry of Education, Guangzhou, China.,Provincial Engineering Technology Research Center for Diseases-Vectors Control, Guangzhou, China
| | - Zhi-Yue Lv
- Department of Parasitology of Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China.,Key Laboratory of Tropical Disease Control, Ministry of Education, Guangzhou, China.,Provincial Engineering Technology Research Center for Diseases-Vectors Control, Guangzhou, China
| | - Xin Zeng
- Department of Parasitology of Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China.,Key Laboratory of Tropical Disease Control, Ministry of Education, Guangzhou, China.,Provincial Engineering Technology Research Center for Diseases-Vectors Control, Guangzhou, China
| | - Xi Sun
- Department of Parasitology of Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China.,Key Laboratory of Tropical Disease Control, Ministry of Education, Guangzhou, China.,Provincial Engineering Technology Research Center for Diseases-Vectors Control, Guangzhou, China
| | - Zhong-Dao Wu
- Department of Parasitology of Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China.,Key Laboratory of Tropical Disease Control, Ministry of Education, Guangzhou, China.,Provincial Engineering Technology Research Center for Diseases-Vectors Control, Guangzhou, China
| |
Collapse
|
9
|
Hemmerling M, Nilsson S, Edman K, Eirefelt S, Russell W, Hendrickx R, Johnsson E, Kärrman Mårdh C, Berger M, Rehwinkel H, Abrahamsson A, Dahmén J, Eriksson AR, Gabos B, Henriksson K, Hossain N, Ivanova S, Jansson AH, Jensen TJ, Jerre A, Johansson H, Klingstedt T, Lepistö M, Lindsjö M, Mile I, Nikitidis G, Steele J, Tehler U, Wissler L, Hansson T. Selective Nonsteroidal Glucocorticoid Receptor Modulators for the Inhaled Treatment of Pulmonary Diseases. J Med Chem 2017; 60:8591-8605. [PMID: 28937774 DOI: 10.1021/acs.jmedchem.7b01215] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
A class of potent, nonsteroidal, selective indazole ether-based glucocorticoid receptor modulators (SGRMs) was developed for the inhaled treatment of respiratory diseases. Starting from an orally available compound with demonstrated anti-inflammatory activity in rat, a soft-drug strategy was implemented to ensure rapid elimination of drug candidates to minimize systemic GR activation. The first clinical candidate 1b (AZD5423) displayed a potent inhibition of lung edema in a rat model of allergic airway inflammation following dry powder inhalation combined with a moderate systemic GR-effect, assessed as thymic involution. Further optimization of inhaled drug properties provided a second, equally potent, candidate, 15m (AZD7594), that demonstrated an improved therapeutic ratio over the benchmark inhaled corticosteroid 3 (fluticasone propionate) and prolonged the inhibition of lung edema, indicating potential for once-daily treatment.
Collapse
Affiliation(s)
- Martin Hemmerling
- Respiratory, Inflammation & Autoimmunity, Innovative Medicines and Early Development Biotech Unit, AstraZeneca , Pepparedsleden 1, Mölndal, SE 43183, Sweden
| | | | - Karl Edman
- Discovery Sciences, Innovative Medicines and Early Development Biotech Unit, AstraZeneca , Pepparedsleden 1, Mölndal, SE 43183, Sweden
| | - Stefan Eirefelt
- AstraZeneca R&D Lund , Scheelevägen 1, Lund, SE 22187, Sweden
| | - Wayne Russell
- AstraZeneca R&D Lund , Scheelevägen 1, Lund, SE 22187, Sweden
| | - Ramon Hendrickx
- Respiratory, Inflammation & Autoimmunity, Innovative Medicines and Early Development Biotech Unit, AstraZeneca , Pepparedsleden 1, Mölndal, SE 43183, Sweden
| | - Eskil Johnsson
- Respiratory, Inflammation & Autoimmunity, Innovative Medicines and Early Development Biotech Unit, AstraZeneca , Pepparedsleden 1, Mölndal, SE 43183, Sweden
| | - Carina Kärrman Mårdh
- Respiratory, Inflammation & Autoimmunity, Innovative Medicines and Early Development Biotech Unit, AstraZeneca , Pepparedsleden 1, Mölndal, SE 43183, Sweden
| | - Markus Berger
- Medicinal Chemistry Berlin, Drug Discovery, Pharmaceuticals, Bayer AG , Berlin 13353, Germany
| | - Hartmut Rehwinkel
- Medicinal Chemistry Berlin, Drug Discovery, Pharmaceuticals, Bayer AG , Berlin 13353, Germany
| | - Anna Abrahamsson
- Respiratory, Inflammation & Autoimmunity, Innovative Medicines and Early Development Biotech Unit, AstraZeneca , Pepparedsleden 1, Mölndal, SE 43183, Sweden
| | - Jan Dahmén
- AstraZeneca R&D Lund , Scheelevägen 1, Lund, SE 22187, Sweden
| | - Anders R Eriksson
- Respiratory, Inflammation & Autoimmunity, Innovative Medicines and Early Development Biotech Unit, AstraZeneca , Pepparedsleden 1, Mölndal, SE 43183, Sweden
| | - Balint Gabos
- AstraZeneca R&D Lund , Scheelevägen 1, Lund, SE 22187, Sweden
| | | | - Nafizal Hossain
- AstraZeneca R&D Lund , Scheelevägen 1, Lund, SE 22187, Sweden
| | | | | | - Tina J Jensen
- Respiratory, Inflammation & Autoimmunity, Innovative Medicines and Early Development Biotech Unit, AstraZeneca , Pepparedsleden 1, Mölndal, SE 43183, Sweden
| | - Anders Jerre
- AstraZeneca R&D Lund , Scheelevägen 1, Lund, SE 22187, Sweden
| | | | | | - Matti Lepistö
- Respiratory, Inflammation & Autoimmunity, Innovative Medicines and Early Development Biotech Unit, AstraZeneca , Pepparedsleden 1, Mölndal, SE 43183, Sweden
| | - Martin Lindsjö
- Pharmaceutical Sciences, Innovative Medicines and Early Development Biotech Unit, AstraZeneca , Pepparedsleden 1, Mölndal, SE 43183, Sweden
| | - Irene Mile
- AstraZeneca R&D Lund , Scheelevägen 1, Lund, SE 22187, Sweden
| | | | - John Steele
- Respiratory, Inflammation & Autoimmunity, Innovative Medicines and Early Development Biotech Unit, AstraZeneca , Pepparedsleden 1, Mölndal, SE 43183, Sweden
| | - Ulrika Tehler
- Pharmaceutical Sciences, Innovative Medicines and Early Development Biotech Unit, AstraZeneca , Pepparedsleden 1, Mölndal, SE 43183, Sweden
| | - Lisa Wissler
- Discovery Sciences, Innovative Medicines and Early Development Biotech Unit, AstraZeneca , Pepparedsleden 1, Mölndal, SE 43183, Sweden
| | - Thomas Hansson
- Respiratory, Inflammation & Autoimmunity, Innovative Medicines and Early Development Biotech Unit, AstraZeneca , Pepparedsleden 1, Mölndal, SE 43183, Sweden
| |
Collapse
|
10
|
Talaber G, Jondal M, Okret S. Local glucocorticoid production in the thymus. Steroids 2015; 103:58-63. [PMID: 26102271 DOI: 10.1016/j.steroids.2015.06.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2014] [Revised: 06/09/2015] [Accepted: 06/15/2015] [Indexed: 12/29/2022]
Abstract
Besides generating immunocompetent T lymphocytes, the thymus is an established site of de novo extra-adrenal glucocorticoid (GC) production. Among the compartments of the thymus, both stromal thymic epithelial cells (TECs) and thymocytes secrete biologically active GCs. Locally produced GCs secreted by the various thymic cellular compartments have been suggested to have different impact on thymic homeostasis. TEC-derived GCs may regulate thymocyte differentiation whereas thymocyte-derived GCs might regulate age-dependent involution. However the full biological significance of thymic-derived GCs is still not fully understood. In this review, we summarize and describe recent advances in the understanding of local GC production in the thymus and immunoregulatory steroid production by peripheral T cells and highlight the possible role of local GCs for thymus function.
Collapse
Affiliation(s)
- Gergely Talaber
- Department of Biosciences and Nutrition, Karolinska Institutet, NOVUM, Huddinge, Sweden.
| | - Mikael Jondal
- Department of Microbiology, Tumor and Cell Biology, Karolinska Insitutet, Stockholm, Sweden
| | - Sam Okret
- Department of Biosciences and Nutrition, Karolinska Institutet, NOVUM, Huddinge, Sweden
| |
Collapse
|
11
|
Abstract
Glucocorticoids (GC) are steroid hormones with important implications in the treatment of various inflammatory and autoimmune diseases. At the same time GC are known to have numerous side-effects. Endogenous GC are predominantly produced by the adrenal glands, and adrenal-derived GC serve important functions in the regulation of development, metabolism, and immune regulation. The last two decades of research have led to the identification of numerous alternative sources of extra-adrenal GC synthesis. Among other tissues the intestine and lung are capable of locally producing considerable amounts of immunoregulatory GC. This local steroidogenesis in these mucosal tissues appears to be regulated by transcription factors and mediators different from those in the adrenals, likely reflecting an adaptation to the local requirements and conditions. Here we summarize the current knowledge about the extra-adrenal GC synthesis in the mucosal tissues, with special emphasis on the intestinal epithelium, and its implication on the regulation of immune homeostasis and inflammatory processes.
Collapse
Affiliation(s)
- Feodora Kostadinova
- Biochemical Pharmacology, Department of Biology, University of Konstanz , Germany
| | | | | | | |
Collapse
|
12
|
Role of miR-181 family in regulating vascular inflammation and immunity. Trends Cardiovasc Med 2013; 24:105-12. [PMID: 24183793 DOI: 10.1016/j.tcm.2013.09.002] [Citation(s) in RCA: 141] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2013] [Revised: 08/30/2013] [Accepted: 08/30/2013] [Indexed: 01/08/2023]
Abstract
The microRNA family, miR-181, plays diverse roles in regulating key aspects of cellular growth, development, and activation. Accumulating evidence supports a central role for the miR-181 family in vascular inflammation by controlling critical signaling pathways, such as downstream NF-κB signaling, and targets relevant to endothelial cell activation and immune cell homeostasis. This review examines the current knowledge of the miR-181 family's role in key cell types that critically control cardiovascular inflammation under pathological and physiological stimuli.
Collapse
|
13
|
Sarang Z, Garabuczi É, Joós G, Kiss B, Tóth K, Rühl R, Szondy Z. Macrophages engulfing apoptotic thymocytes produce retinoids to promote selection, differentiation, removal and replacement of double positive thymocytes. Immunobiology 2013; 218:1354-60. [PMID: 23932496 DOI: 10.1016/j.imbio.2013.06.009] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2013] [Revised: 06/04/2013] [Accepted: 06/17/2013] [Indexed: 11/30/2022]
Abstract
The thymus provides the microenvironment in which thymocytes develop into mature T-cells, and interactions with thymic stromal cells are thought to provide the necessary signals for thymocyte maturation. Recognition of self-MHC by T-cells is a basic requirement for mature T-cell functions, and those thymocytes that do not recognize or respond too strongly to the peptide-loaded self-MHC molecules found in the thymus undergo apoptosis. As a result, 95% of the thymocytes produced will die and be subsequently cleared by macrophages. This review describes a complex crosstalk between developing thymocytes and engulfing macrophages which is mediated by retinoids produced by engulfing macrophages. The interaction results in the harmonization of the rate of cell death of dying double positive cells with their clearance and replacement, and in promotion of the differentiation of the selected cells in the thymus.
Collapse
Affiliation(s)
- Zsolt Sarang
- Section of Dental Biochemistry, Department of Biochemistry and Molecular Biology, Research Center of Molecular Medicine, University of Debrecen, Nagyerdei krt. 98, 4012 Debrecen, Hungary
| | | | | | | | | | | | | |
Collapse
|
14
|
Gruver-Yates AL, Cidlowski JA. Tissue-specific actions of glucocorticoids on apoptosis: a double-edged sword. Cells 2013; 2:202-23. [PMID: 24709697 PMCID: PMC3972684 DOI: 10.3390/cells2020202] [Citation(s) in RCA: 103] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2013] [Revised: 03/05/2013] [Accepted: 03/12/2013] [Indexed: 12/20/2022] Open
Abstract
First described for their metabolic and immunosuppressive effects, glucocorticoids are widely prescribed in clinical settings of inflammation. However, glucocorticoids are also potent inducers of apoptosis in many cell types and tissues. This review will focus on the established mechanisms of glucocorticoid-induced apoptosis and outline what is known about the apoptotic response in cells and tissues of the body after exposure to glucocorticoids. Glucocorticoid-induced apoptosis affects the skeletal system, muscular system, circulatory system, nervous system, endocrine system, reproductive system, and the immune system. Interestingly, several cell types have an anti-apoptotic response to glucocorticoids that is cytoprotective. Lastly, we will discuss the pro- and anti-apoptotic effects of glucocorticoids in cancers and their clinical implications.
Collapse
Affiliation(s)
- Amanda L Gruver-Yates
- Laboratory of Signal Transduction, National Institute of Environmental Health Sciences, 111 T. W. Alexander Drive, Research Triangle Park, NC 27709, USA.
| | - John A Cidlowski
- Laboratory of Signal Transduction, National Institute of Environmental Health Sciences, 111 T. W. Alexander Drive, Research Triangle Park, NC 27709, USA.
| |
Collapse
|
15
|
Everds NE, Snyder PW, Bailey KL, Bolon B, Creasy DM, Foley GL, Rosol TJ, Sellers T. Interpreting Stress Responses during Routine Toxicity Studies. Toxicol Pathol 2013; 41:560-614. [DOI: 10.1177/0192623312466452] [Citation(s) in RCA: 201] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Stress often occurs during toxicity studies. The perception of sensory stimuli as stressful primarily results in catecholamine release and activation of the hypothalamic–pituitary–adrenal (HPA) axis to increase serum glucocorticoid concentrations. Downstream effects of these neuroendocrine signals may include decreased total body weights or body weight gain; food consumption and activity; altered organ weights (e.g., thymus, spleen, adrenal); lymphocyte depletion in thymus and spleen; altered circulating leukocyte counts (e.g., increased neutrophils with decreased lymphocytes and eosinophils); and altered reproductive functions. Typically, only some of these findings occur in a given study. Stress responses should be interpreted as secondary (indirect) rather than primary (direct) test article–related findings. Determining whether effects are the result of stress requires a weight-of-evidence approach. The evaluation and interpretation of routinely collected data (standard in-life, clinical pathology, and anatomic pathology endpoints) are appropriate and generally sufficient to assess whether or not changes are secondary to stress. The impact of possible stress-induced effects on data interpretation can partially be mitigated by toxicity study designs that use appropriate control groups (e.g., cohorts treated with vehicle and subjected to the same procedures as those dosed with test article), housing that minimizes isolation and offers environmental enrichment, and experimental procedures that minimize stress and sampling and analytical bias. This article is a comprehensive overview of the biological aspects of the stress response, beginning with a Summary (Section 1) and an Introduction (Section 2) that describes the historical and conventional methods used to characterize acute and chronic stress responses. These sections are followed by reviews of the primary systems and parameters that regulate and/or are influenced by stress, with an emphasis on parameters evaluated in toxicity studies: In-life Procedures (Section 3), Nervous System (Section 4), Endocrine System (Section 5), Reproductive System (Section 6), Clinical Pathology (Section 7), and Immune System (Section 8). The paper concludes (Section 9) with a brief discussion on Minimizing Stress-Related Effects (9.1.), and a final section explaining why Parameters routinely measured are appropriate for assessing the role of stress in toxicology studies (9.2.).
Collapse
Affiliation(s)
| | | | - Keith L. Bailey
- Oklahoma Animal Disease Diagnostic Laboratory, Oklahoma State University, Stillwater, Oklahoma, USA
| | - Brad Bolon
- Department of Veterinary Biosciences and the Comparative Pathology and Mouse Phenotyping Shared Resource, The Ohio State University, Columbus, Ohio, USA
| | | | | | - Thomas J. Rosol
- Department of Veterinary Biosciences, The Ohio State University, Columbus, Ohio, USA
| | | |
Collapse
|
16
|
Szondy Z, Garabuczi É, Tóth K, Kiss B, Köröskényi K. Thymocyte death by neglect: Contribution of engulfing macrophages. Eur J Immunol 2012; 42:1662-7. [DOI: 10.1002/eji.201142338] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
17
|
Pérez AR, Berbert LR, Lepletier A, Revelli S, Bottasso O, Silva-Barbosa SD, Savino W. TNF-α is involved in the abnormal thymocyte migration during experimental Trypanosoma cruzi infection and favors the export of immature cells. PLoS One 2012; 7:e34360. [PMID: 22461911 PMCID: PMC3312912 DOI: 10.1371/journal.pone.0034360] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2011] [Accepted: 02/27/2012] [Indexed: 11/28/2022] Open
Abstract
Previous studies revealed a significant production of inflammatory cytokines together with severe thymic atrophy and thymocyte migratory disturbances during experimental Chagas disease. Migratory activity of thymocytes and mature T cells seem to be finely tuned by cytokines, chemokines and extracellular matrix (ECM) components. Systemic TNF-α is enhanced during infection and appears to be crucial in the response against the parasite. However, it also seems to be involved in disease pathology, since it is implicated in the arrival of T cells to effector sites, including the myocardium. Herein, we analyzed the role of TNF-α in the migratory activity of thymocytes in Trypanosoma cruzi (T. cruzi) acutely-infected mice. We found increased expression and deposition of TNF-α in the thymus of infected animals compared to controls, accompanied by increased co-localization of fibronectin, a cell migration-related ECM molecule, whose contents in the thymus of infected mice is also augmented. In-vivo studies showed an enhanced export of thymocytes in T. cruzi-infected mice, as ascertained by intrathymic injection of FITC alone or in combination with TNF-α. The increase of immature CD4+CD8+ T cells in secondary lymphoid organs was even more clear-cut when TNF-α was co-injected with FITC. Ex-vivo transmigration assays also revealed higher number of migrating cells when TNF-α was added onto fibronectin lattices, with higher input of all thymocyte subsets, including immature CD4+CD8+. Infected animals also exhibit enhanced levels of expression of both mRNA TNF-α receptors in the CD4+CD8+ subpopulation. Our findings suggest that in T. cruzi acute infection, when TNF-α is complexed with fibronectin, it favours the altered migration of thymocytes, promoting the release of mature and immature T cells to different compartments of the immune system. Conceptually, this work reinforces the notion that thymocyte migration is a multivectorial biological event in health and disease, and that TNF-α is a further player in the process.
Collapse
Affiliation(s)
- Ana Rosa Pérez
- Faculty of Medical Sciences, Institute of Immunology, National University of Rosario, Rosario, Argentina.
| | | | | | | | | | | | | |
Collapse
|
18
|
Tischner D, Theiss J, Karabinskaya A, van den Brandt J, Reichardt SD, Karow U, Herold MJ, Lühder F, Utermöhlen O, Reichardt HM. Acid sphingomyelinase is required for protection of effector memory T cells against glucocorticoid-induced cell death. THE JOURNAL OF IMMUNOLOGY 2011; 187:4509-16. [PMID: 21948986 DOI: 10.4049/jimmunol.1100911] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The activity of acid sphingomyelinase (aSMase) was previously reported to be involved in glucocorticoid-induced cell death (GICD) of T lymphocytes. This mechanism in turn is believed to contribute to the therapeutic efficacy of glucocorticoids (GCs) in the treatment of inflammatory diseases. In this study, we reassessed the role of aSMase in GICD by using aSMase knockout mice. The absence of aSMase largely abolished the partial protection that effector memory CD4(+) T cells in wild-type mice possess against GICD. Reduced IL-2 secretion by aSMase-deficient CD4(+) T cells suggested that a lack of this important survival factor might be the cause of these cells' enhanced susceptibility to GICD. Indeed, addition of IL-2 restored the protection against GICD, whereas neutralization of IL-2 abrogated the otherwise protective effect seen in wild-type effector memory CD4(+) T cells. The therapeutic implications of the altered sensitivity of aSMase-deficient T cells to GICD were assessed in models of inflammatory disorders; namely, experimental autoimmune encephalomyelitis and acute graft-versus-host disease. Surprisingly, GC treatment was equally efficient in both models in terms of ameliorating the diseases, regardless of the genotype of the T cells. Thus, our data reveal a hitherto unrecognized contribution of aSMase to the sensitivity of effector memory CD4(+) T cells to GICD and call into question the traditionally attributed importance of GICD of T cells to the treatment of inflammatory diseases by GCs.
Collapse
Affiliation(s)
- Denise Tischner
- Department of Cellular and Molecular Immunology, University of Göttingen Medical School, Göttingen 37073, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Hu T, Gimferrer I, Alberola-Ila J. Control of early stages in invariant natural killer T-cell development. Immunology 2011; 134:1-7. [PMID: 21718314 PMCID: PMC3173689 DOI: 10.1111/j.1365-2567.2011.03463.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2011] [Revised: 05/17/2011] [Accepted: 05/31/2011] [Indexed: 12/20/2022] Open
Abstract
Natural killer T (NKT) cells develop in the thymus from the same precursors as conventional CD4(+) and CD8(+) αβ T cells, CD4(+) CD8(+) double-positive cells. In contrast to conventional αβT cells, which are selected by MHC-peptide complexes presented by thymic epithelial cells, invariant NKT cells are selected by lipid antigens presented by the non-polymorphic, MHC I-like molecule CD1d, present on the surface of other double-positive thymocytes, and require additional signals from the signalling lymphocytic-activation molecule (SLAM) family of receptors. In this review, we provide a discussion of recent findings that have modified our understanding of the NKT cell developmental programme, with an emphasis on events that affect the early stages of this process. This includes factors that control double-positive thymocyte lifespan, and therefore the ability to generate the canonical Vα rearrangements that characterize this lineage, as well as the signal transduction pathways engaged downstream of the T-cell receptor and SLAM molecules.
Collapse
Affiliation(s)
- Taishan Hu
- Immunobiology and Cancer Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | | | | |
Collapse
|
20
|
Talabér G, Boldizsár F, Bartis D, Pálinkás L, Szabó M, Berta G, Sétáló G, Németh P, Berki T. Mitochondrial translocation of the glucocorticoid receptor in double-positive thymocytes correlates with their sensitivity to glucocorticoid-induced apoptosis. Int Immunol 2009; 21:1269-76. [PMID: 19737783 DOI: 10.1093/intimm/dxp093] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Glucocorticoid receptor (GR) signaling plays an important role in the selection and apoptosis of thymocytes. Besides nuclear translocation, mitochondrial translocation of the ligand-bound GR in lymphoid cells was also shown, which might determine glucocorticoid (GC)-induced apoptosis sensitivity. In the present work, we followed the ligand-induced GR trafficking in CD4+CD8+ double-positive (DP) thymocytes. Using confocal microscopy, we found that upon short-term in vitro GC analog [dexamethasone (DX)] treatment, the GR translocates into the mitochondria but not into the nucleus in DP cells. We also analyzed the GR redistribution in cytosolic, nuclear and mitochondrial fractions of unseparated thymocytes by western blot and confirmed that in DX-treated cells a significant fraction of the GR translocates into the mitochondria. DX reduced the mitochondrial membrane potential of DP cells within 30 min, measured by flow cytometry, which refers to a direct modulatory activity of mitochondrial GR translocation. The abundant mitochondrial GR found in DP cells well correlates with their high GC-induced apoptosis sensitivity.
Collapse
Affiliation(s)
- Gergely Talabér
- Department of Immunology and Biotechnology, Faculty of Medicine, University of Pecs, Szigeti ut. 12, Hungary
| | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Stojić-Vukanić Z, Rauski A, Kosec D, Radojević K, Pilipović I, Leposavić G. Dysregulation of T-cell development in adrenal glucocorticoid-deprived rats. Exp Biol Med (Maywood) 2009; 234:1067-74. [PMID: 19546352 DOI: 10.3181/0902-rm-63] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
A number of different experimental approaches have been used to elucidate the impact of basal levels of adrenal gland-derived glucocorticoids (GCs) on T cell development, and thereby T cell-mediated immune responses. However, the relevance of the adrenal GCs to T cell development is still far from clear. This study was undertaken to explore the relevance of basal levels of GCs to T cell differentiation/maturation. Eight days post-adrenalectomy in adult male rats the thymocyte yield, apoptotic and proliferative rate and the relationship amongst major thymocyte subsets, as defined by TCRalphabeta/CD4/CD8 expression, were examined using flow cytometry. Adrenal GC deprivation decreased thymocyte apoptosis and altered the kinetics of T cell differentiation/maturation. In the adrenalectomized rats there was increased thymic hypercellularity and an over-representation of the CD4+CD8+ double positive (DP) TCRalphabeta(low) cells entering selection, as well as increased numbers of their DP TCRalphabeta(-) immediate precursors. These changes were accompanied with under-representation of the postselected DP TCRalphabeta(high) and the most mature CD4-CD8+ and, particularly, CD4+CD8- single positive (SP) TCRalphabeta(high) cells. This data suggests that withdrawal of adrenal GCs produces alterations in the thymocyte selection processes, possibly affecting the diversity of functional T cell repertoire and generation of potentially self-reactive cells as indicated by the reduced proportion and number of CD4-CD8- double negative TCRalphabeta(high) cells. In addition, it indicates that GCs influence the post-selection maturation of thymocytes and plays a regulatory role in controlling the ratio of mature CD4+CD8-/CD4-CD8+ SP TCRalphabeta(high) cells.
Collapse
Affiliation(s)
- Zorica Stojić-Vukanić
- Department of Microbiology and Immunology, Faculty of Pharmacy, University of Belgrade, 11221 Belgrade, Serbia
| | | | | | | | | | | |
Collapse
|
22
|
Bellinger DL, Lubahn C, Lorton D. Maternal and early life stress effects on immune function: relevance to immunotoxicology. J Immunotoxicol 2009; 5:419-44. [PMID: 19404876 DOI: 10.1080/15476910802483415] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Stress is triggered by a variety of unexpected environmental stimuli, such as aggressive behavior, fear, forced physical activity, sudden environmental changes, social isolation or pathological conditions. Stressful experiences during very early life (particularly, maternal stress during fetal ontogeny) can permanently alter the responsiveness of the nervous system, an effect called programming or imprinting. Programming affects the hypothalamic-pituitary-adrenocortical (HPA) axis, brain neurotransmitter systems, sympathetic nervous system (SNS), and the cognitive abilities of the offspring, which can alter neural regulation of immune function. Prenatal or early life stress may contribute to the maladaptive immune responses to stress that occur later in life. This review focuses on the effect of maternal and early life stress on immune function in the offspring across life span. It highlights potential mechanisms by which prenatal stress impacts immune functions over life span. The literature discussed in this review suggests that psychosocial stress during pre- and early postnatal life may increase the vulnerability of infants to the effects of immunotoxicants or immune-mediated diseases, with long-term consequences. Neural-immune interactions may provide an indirect route through which immunotoxicants affect the developing immune system. A developmental approach to understanding how immunotoxicants interact with maternal and early life stress-induced changes in immunity is needed, because as the body changes physiologically across life span so do the effects of stress and immunotoxicants. In early and late life, the immune system is more vulnerable to the effects of stress. Stress can mimic the effects of aging and exacerbate age-related changes in immune function. This is important because immune dysregulation in the elderly is more frequently and seriously associated with clinical impairment and death. Aging, exposure to teratogens, and psychological stress interact to increase vulnerability and put the elderly at the greatest risk for disease.
Collapse
Affiliation(s)
- Denise L Bellinger
- Department of Human Anatomy and Pathology, Loma Linda University School of Medicine, Loma Linda, CA 92352, USA.
| | | | | |
Collapse
|
23
|
Gruver AL, Sempowski GD. Cytokines, leptin, and stress-induced thymic atrophy. J Leukoc Biol 2008; 84:915-23. [PMID: 18495786 DOI: 10.1189/jlb.0108025] [Citation(s) in RCA: 167] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Thymopoiesis is essential for development and maintenance of a robust and healthy immune system. Acute thymic atrophy is a complication of many infections, environmental stressors, clinical preparative regimens, and cancer treatments used today. This undesirable sequela can decrease host ability to reconstitute the peripheral T cell repertoire and respond to new antigens. Currently, there are no treatments available to protect against acute thymic atrophy or accelerate recovery, thus leaving the immune system compromised during acute stress events. Several useful murine models are available for mechanistic studies of acute thymic atrophy, including a sepsis model of endotoxin-induced thymic involution. We have identified the IL-6 cytokine gene family members (i.e., leukemia inhibitory factor, IL-6, and oncostatin M) as thymosuppressive agents by the observation that they can acutely involute the thymus when injected into a young, healthy mouse. We have gone on to explore the role of thymosuppressive cytokines and specifically defined a corticosteroid-dependent mechanism of action for the leukemia inhibitory factor in acute thymic atrophy. We also have identified leptin as a novel, thymostimulatory agent that can protect against endotoxin-induced acute thymic atrophy. This review will highlight mechanisms of stress-induced thymic involution and focus on thymosuppressive agents involved in atrophy induction and thymostimulatory agents that may be exploited for therapeutic use.
Collapse
Affiliation(s)
- Amanda L Gruver
- Department of Pathology, Duke University Medical Center, Durham, NC 27710, USA
| | | |
Collapse
|
24
|
van den Brandt J, Lühder F, McPherson KG, de Graaf KL, Tischner D, Wiehr S, Herrmann T, Weissert R, Gold R, Reichardt HM. Enhanced glucocorticoid receptor signaling in T cells impacts thymocyte apoptosis and adaptive immune responses. THE AMERICAN JOURNAL OF PATHOLOGY 2007; 170:1041-53. [PMID: 17322387 PMCID: PMC1864890 DOI: 10.2353/ajpath.2007.060804] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
To study the effect of enhanced glucocorticoid signaling on T cells, we generated transgenic rats overexpressing a mutant glucocorticoid receptor with increased ligand affinity in the thymus. We found that this caused massive thymocyte apoptosis at physiological hormone levels, which could be reversed by adrenalectomy. Due to homeostatic proliferation, a considerable number of mature T lymphocytes accumulated in the periphery, responding normally to costimulation but exhibiting a perturbed T-cell repertoire. Furthermore, the transgenic rats showed increased resistance to experimental autoimmune encephalomyelitis, which manifests in a delayed onset and milder disease course, impaired leukocyte infiltration into the central nervous system and a distinct cytokine profile. In contrast, the ability of the transgenic rats to mount an allergic airway response to ovalbumin was not compromised, although isotype switching of antigen-specific immunoglobulins was altered. Collectively, our findings suggest that endogenous glucocorticoids impact T-cell development and favor the selection of Th2- over Th1-dominated adaptive immune responses.
Collapse
Affiliation(s)
- Jens van den Brandt
- Institute for Virology and Immunobiology, University of Würzburg, Würzburg, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Lustig A, Weeraratna AT, Wood WW, Teichberg D, Bertak D, Carter A, Poosala S, Firman J, Becker KG, Zonderman AB, Longo DL, Taub DD. Transcriptome analysis of age-, gender- and diet-associated changes in murine thymus. Cell Immunol 2007; 245:42-61. [PMID: 17499630 PMCID: PMC2271048 DOI: 10.1016/j.cellimm.2007.03.008] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2007] [Revised: 03/25/2007] [Accepted: 03/27/2007] [Indexed: 01/18/2023]
Abstract
The loss of thymic function with age may be due to diminished numbers of T-cell progenitors and the loss of critical mediators within the thymic microenvironment. To assess the molecular changes associated with this loss, we examined transcriptomes of progressively aging mouse thymi, of different sexes and on caloric-restricted (CR) vs. ad libitum (AL) diets. Genes involved in various biological and molecular processes including transcriptional regulators, stress response, inflammation and immune function significantly changed during thymic aging. These differences depended on variables such as sex and diet. Interestingly, many changes associated with thymic aging are either muted or almost completely reversed in mice on caloric-restricted diets. These studies provide valuable insight into the molecular mechanisms associated with thymic aging and emphasize the need to account for biological variables such as sex and diet when elucidating the genomic correlates that influence the molecular pathways responsible for thymic involution.
Collapse
Affiliation(s)
- Ana Lustig
- Laboratory of Immunology, National Institute on Aging-Intramural Research Program, National Institutes of Health, 5600 Nathan Shock Drive, Baltimore, MD 21224, USA
| | - Ashani T. Weeraratna
- Laboratory of Immunology, National Institute on Aging-Intramural Research Program, National Institutes of Health, 5600 Nathan Shock Drive, Baltimore, MD 21224, USA
| | - William W. Wood
- The Research Resources Branch, National Institute on Aging-Intramural Research Program, National Institutes of Health, Baltimore, MD 21224, USA
| | - Diane Teichberg
- The Research Resources Branch, National Institute on Aging-Intramural Research Program, National Institutes of Health, Baltimore, MD 21224, USA
| | - Dorothy Bertak
- Laboratory of Immunology, National Institute on Aging-Intramural Research Program, National Institutes of Health, 5600 Nathan Shock Drive, Baltimore, MD 21224, USA
| | - Arnell Carter
- Laboratory of Immunology, National Institute on Aging-Intramural Research Program, National Institutes of Health, 5600 Nathan Shock Drive, Baltimore, MD 21224, USA
| | - Suresh Poosala
- The Research Resources Branch, National Institute on Aging-Intramural Research Program, National Institutes of Health, Baltimore, MD 21224, USA
| | - Jeffrey Firman
- The Research Resources Branch, National Institute on Aging-Intramural Research Program, National Institutes of Health, Baltimore, MD 21224, USA
| | - Kevin G. Becker
- The Research Resources Branch, National Institute on Aging-Intramural Research Program, National Institutes of Health, Baltimore, MD 21224, USA
| | - Alan B. Zonderman
- The Research Resources Branch, National Institute on Aging-Intramural Research Program, National Institutes of Health, Baltimore, MD 21224, USA
| | - Dan L. Longo
- Laboratory of Immunology, National Institute on Aging-Intramural Research Program, National Institutes of Health, 5600 Nathan Shock Drive, Baltimore, MD 21224, USA
| | - Dennis D. Taub
- Laboratory of Immunology, National Institute on Aging-Intramural Research Program, National Institutes of Health, 5600 Nathan Shock Drive, Baltimore, MD 21224, USA
| |
Collapse
|
26
|
Bianchini R, Nocentini G, Krausz LT, Fettucciari K, Coaccioli S, Ronchetti S, Riccardi C. Modulation of pro- and antiapoptotic molecules in double-positive (CD4+CD8+) thymocytes following dexamethasone treatment. J Pharmacol Exp Ther 2006; 319:887-97. [PMID: 16914556 DOI: 10.1124/jpet.106.108480] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Glucocorticoids play a role in regulation of T lymphocytes homeostasis and development. In particular, glucocorticoid treatment induces massive apoptosis of CD4(+)CD8(+) double-positive (DP) thymocytes. This effect is due to many mechanisms, mainly driven by modulation of gene transcription. To find out which genes are modulated, we analyzed DP thymocytes treated for 3 h with dexamethasone (a synthetic glucocorticoid) by global gene expression profiling. Results indicate modulation of 163 genes, also confirmed by either RNase protection assay or real-time polymerase chain reaction. In particular, dexamethasone caused down-regulation of genes promoting DP thymocyte survival (e.g., Notch1, suppressor of cytokine signaling 1, and inhibitor of DNA binding 3) or modulation of genes activating cell death through the ceramide pathway (UDP-glucose ceramide glucosyltransferase, sphingosine 1-phosphate phosphatase, dihydroceramide desaturase, isoform 1, and G protein-coupled receptor 65) or through the mitochondrial machinery. Among the latter, there are Bcl-2 family members (Bim, Bfl-1, Bcl-xL, and Bcl-xbeta), genes involved in the control of redox status (thioredoxin reductase, thioredoxin reductase inhibitor, and NADP(+)-dependent isocitrate dehydrogenase) and genes belonging to Tis11 family that are involved in mRNA stability. Our study suggests that dexamethasone treatment of DP thymocytes modulates several genes belonging to apoptosis-related systems that can contribute to their apoptosis.
Collapse
Affiliation(s)
- Rodolfo Bianchini
- Dipartimento di Medicina Clinica e Sperimentale, Sezione di Farmacologia, Tossicologia e Chemioterapia, Università di Perugia, Istituto di Biotecnologie Trapiantologiche and Polo Scientifico e Didattico di Terni, Perugia, Italy
| | | | | | | | | | | | | |
Collapse
|
27
|
Abstract
The thymus is a very sensitive target for environmental pollutants, which can affect this organ as well as thymocyte differentiation. A failure in thymocyte development can be due to the exacerbation of apoptosis, arrest of thymocyte maturation, generation of autoreactive T cells, and inhibition or stimulation of the output of recent thymic emigrants to the periphery. Recent data demonstrate that the immune system has the potential to maintain homeostasis under conditions of elevated risk, and the thymus plays a crucial role in this process. Environmental xenobiotics can exert their effects through receptor-mediated interactions or independently on receptor involvement. Under natural conditions organisms are exposed to a variety of xenobiotics. The final effect of such exposure is not related to the action of a single chemical, but to the action of a mixture of chemicals. The toxic effect of environmental xenobiotics on the generation and functions of immune cells may result in suppression or stimulation of the immune response. The most intensive studies have been done on halogenated aromatic hydrocarbons, heavy metals and various chemicals acting as endocrine disrupters. Recently, special interest has focused on the action of air particulate matter.
Collapse
Affiliation(s)
- Nadzieja Drela
- Department of Immunology, Warsaw University, Warsaw, Poland.
| |
Collapse
|
28
|
Loftus SK, Cannons JL, Incao A, Pak E, Chen A, Zerfas PM, Bryant MA, Biesecker LG, Schwartzberg PL, Pavan WJ. Acinar cell apoptosis in Serpini2-deficient mice models pancreatic insufficiency. PLoS Genet 2006; 1:e38. [PMID: 16184191 PMCID: PMC1231717 DOI: 10.1371/journal.pgen.0010038] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2005] [Accepted: 08/18/2005] [Indexed: 11/18/2022] Open
Abstract
Pancreatic insufficiency (PI) when left untreated results in a state of malnutrition due to an inability to absorb nutrients. Frequently, PI is diagnosed as part of a larger clinical presentation in cystic fibrosis or Shwachman-Diamond syndrome. In this study, a mouse model for isolated exocrine PI was identified in a mouse line generated by a transgene insertion. The trait is inherited in an autosomal recessive pattern, and homozygous animals are growth retarded, have abnormal immunity, and have reduced life span. Mice with the disease locus, named pequeño (pq), exhibit progressive apoptosis of pancreatic acinar cells with severe exocrine acinar cell loss by 8 wk of age, while the islets and ductal tissue persist. The mutation in pq/pq mice results from a random transgene insertion. Molecular characterization of the transgene insertion site by fluorescent in situ hybridization and genomic deletion mapping identified an approximately 210-kb deletion on Chromosome 3, deleting two genes. One of these genes, Serpini2, encodes a protein that is a member of the serpin family of protease inhibitors. Reintroduction of only the Serpini2 gene by bacterial artificial chromosome transgenic complementation corrected the acinar cell defect as well as body weight and immune phenotypes, showing that deletion of Serpini2 causes the pequeño phenotype. Dietary supplementation of pancreatic enzymes also corrected body size, body weight, and immunodeficiency, and increased the life span of Serpini2-deficient mice, despite continued acinar cell loss. To our knowledge, this study describes the first characterized genetic animal model for isolated PI. Genetic complementation of the transgene insertion mutant demonstrates that Serpini2 deficiency directly results in the acinar cell apoptosis, malabsorption, and malnutrition observed in pq/pq mice. The rescue of growth retardation, immunodeficiency, and mortality by either Serpini2 bacterial artificial chromosome transgenic expression or by pancreatic enzyme supplementation demonstrates that these phenotypes are secondary to malnutrition in pq/pq mice.
Collapse
Affiliation(s)
- Stacie K Loftus
- Genetic Disease Research Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Nuotio-Antar AM, Hasty AH, Kovacs WJ. Quantitation and cellular localization of 11beta-HSD1 expression in murine thymus. J Steroid Biochem Mol Biol 2006; 99:93-9. [PMID: 16621520 DOI: 10.1016/j.jsbmb.2006.01.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2005] [Accepted: 01/05/2006] [Indexed: 11/28/2022]
Abstract
11beta-hydroxysteroid dehydrogenase type I (11beta-HSD1), an NADPH-dependent reductase, functions in intact cells to convert inactive 11-keto metabolites of glucocorticoids into biologically active glucocorticoids. The enzyme is thus capable of amplifying glucocorticoid action in tissues in which it is expressed. In the experiments presented here, we show that 11beta-HSD1 is expressed in the murine thymus and that expression increases from late fetal development to maximal levels in the adult thymus. Quantitative real time-PCR, immunoblots, and assays of enzymatic activity reveal adult thymic expression of 11beta-HSD1 mRNA and protein at levels approximately 6-7% of those observed in liver. Immunofluorescence experiments show that the enzyme is expressed in the medullary thymocytes and thymocytes present at the corticomedullary junction. These experiments extend our recognition of 11beta-HSD1 expression in cells of the immune system and lend support to the notion that glucocorticoid signaling and amplification of those signals by regeneration of active glucocorticoids from inactive 11-keto metabolites might impact intrathymic T cell development and the establishment of the immune repertoire.
Collapse
Affiliation(s)
- Alli M Nuotio-Antar
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | | | | |
Collapse
|
30
|
Abstract
Glucocorticoids (GCs) are a class of steroid hormones which regulate a variety of essential biological functions. The profound anti-inflammatory and immunosuppressive activity of synthetic GCs, combined with their power to induce lymphocyte apoptosis place them among the most commonly prescribed drugs worldwide. Endogenous GCs also exert a wide range of immunomodulatory activities, including the control of T cell homeostasis. Most, if not all of these effects are mediated through the glucocorticoid receptor, a member of the nuclear receptor superfamily. However, the signaling pathways and their cell type specificity remain poorly defined. In this review, we summarize our present knowledge on GC action, the mechanisms employed to induce apoptosis and the currently discussed models of how they may participate in thymocyte development. Although our knowledge in this field has substantially increased during recent years, we are still far from a comprehensive picture of the role that GCs play in T lymphocytes.
Collapse
Affiliation(s)
- M. J. Herold
- Molecular Immunology, Institute for Virology and Immunobiology, University of Würzburg, Versbacher Strasse 7, 97078 Würzburg, Germany
| | - K. G. McPherson
- Molecular Immunology, Institute for Virology and Immunobiology, University of Würzburg, Versbacher Strasse 7, 97078 Würzburg, Germany
| | - H. M. Reichardt
- Molecular Immunology, Institute for Virology and Immunobiology, University of Würzburg, Versbacher Strasse 7, 97078 Würzburg, Germany
| |
Collapse
|
31
|
Jang J, Choi YI, Choi J, Lee KY, Chung H, Jeon SH, Seong RH. Notch1 confers thymocytes a resistance to GC-induced apoptosis through Deltex1 by blocking the recruitment of p300 to the SRG3 promoter. Cell Death Differ 2005; 13:1495-505. [PMID: 16341126 DOI: 10.1038/sj.cdd.4401827] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
One notable phenotypic change during the differentiation of immature thymocytes into either mature CD4 or CD8 single-positive lineages is the acquisition of a resistance to glucocorticoid (GC)-induced apoptosis. We have previously reported that SRG3 is critical in determining the sensitivity for the GC-induced apoptosis in developing thymocytes. We report here that Notch signaling downregulates the transcriptional activation of SRG3 through N-box and/or E-box elements on its promoter. RBP-J represses SRG3 transcription through the N-box motif. On the other hand, Deltex1 competitively inhibits the binding of p300 to E2A/HEB protein bound to the E-box elements and represses the SRG3 promoter activity. Moreover, enforced expression of Deltex1 restored double-positive (DP) thymocyte survival from the GC-induced apoptosis. Our results suggest that Notch signaling confers differentiating DP thymocytes resistance to GCs by regulating the SRG3 expression through Deltex1, and that Deltex1 and SRG3 may play a significant role during DP thymocyte maturation.
Collapse
Affiliation(s)
- J Jang
- Department of Biological Sciences, Institute of Molecular Biology of Genetics, and Center for Functional Cellulomics, Seoul National University, Seoul 151-742, Korea
| | | | | | | | | | | | | |
Collapse
|
32
|
Abstract
The major function of the thymus is to eliminate developing thymocytes that are potentially useless or autoreactive, and select only those that bear functional T cell antigen receptors (TCRs) through fastidious screening. It is believed that glucocorticoids (GCs) are at least in part responsible for cell death during death by neglect. In this review, we will mainly cover the topic of the GC-induced apoptosis of developing thymocytes. We will also discuss how thymocytes that are fated to die by GCs can be rescued from GC-induced apoptosis in response to a variety of signals with antagonizing properties for GC receptor (GR) signaling. Currently, a lot of evidence supports the notion that the decision is made as a result of the integration of the multiple signal transduction networks that are triggered by GR, TCR, and Notch. A few candidate molecules at the converging point of these multiple signaling pathyways will be discussed. We will particularly describe the role of the SRG3 protein as a potent modulator of GC-induced apoptosis in the crosstalk.
Collapse
Affiliation(s)
- Heekyoung Chung
- School of Biological Sciences and Institute of Molecular Biology & Genetics, Seoul National University, Seoul 151-742, Korea
| | | | | | | |
Collapse
|
33
|
Lépine S, Le Stunff H, Lakatos B, Sulpice JC, Giraud F. ATP-induced apoptosis of thymocytes is mediated by activation of P2 X 7 receptor and involves de novo ceramide synthesis and mitochondria. Biochim Biophys Acta Mol Cell Biol Lipids 2005; 1761:73-82. [PMID: 16325464 DOI: 10.1016/j.bbalip.2005.10.001] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2005] [Revised: 10/03/2005] [Accepted: 10/05/2005] [Indexed: 11/26/2022]
Abstract
Thymocytes were reported to undergo apoptosis in the presence of extracellular ATP through the activation of the purinergic receptors P2 X 1R, P2 X 7R or both. We investigated the identity of the P2 X R and the signaling pathways involved in ATP-mediated apoptosis. Apoptosis elicited by ATP was prevented by inhibition of P2 X 7R, or in thymocytes bearing a mutated P2 X 7R, and reproduced with a P2 X 7R agonist, but not with a P2 X 1R agonist. Stimulation of thymocytes with either ATP or a P2 X 7R agonist was found to stimulate a late de novo ceramide synthesis and mitochondrial alterations. Inhibition of either processes attenuated apoptosis. Interestingly, stimulation with either ATP or a P2 X 1R agonist induced an early ceramide accumulation and a weak caspases-3/7 activation that did not lead to apoptosis. In conclusion, de novo ceramide generation and mitochondrial alterations, both resulting from P2 X 7R activation, were implicated in ATP-induced thymocyte apoptosis.
Collapse
Affiliation(s)
- S Lépine
- Biomembranes et Messagers Cellulaires, Centre National de la Recherche Scientifique, Unité Mixte de Recherche 8619, Université Paris XI-Orsay, France
| | | | | | | | | |
Collapse
|
34
|
Jondal M, Pazirandeh A, Okret S. Different roles for glucocorticoids in thymocyte homeostasis? Trends Immunol 2005; 25:595-600. [PMID: 15489188 DOI: 10.1016/j.it.2004.09.003] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Glucocorticoids (GCs) have important immunoregulatory effects on thymocytes and T cells. Ectopic production of GCs has been demonstrated in thymic epithelial cells (TECs) but the role of GCs in thymocyte homeostasis is controversial. Studies in several different mouse models, genetically modified for the GC receptor (GR) expression or function, have demonstrated conflicting results in terms of the effect of the hormone on thymocytes. Here, we summarize these data and suggest that GCs can mediate both positive and negative effects in the organ depending on the local hormonal concentration. Basal GC levels might promote growth of early thymocytes in young mice, and increased levels, generated through a stress reaction, apoptosis in these cells. A gradual loss of GC synthesis in TECs during aging might contribute to thymic involution, a process so far unexplained.
Collapse
Affiliation(s)
- Mikael Jondal
- Microbiology and Tumor Biology Center (MTC), Karolinska Institute, Box 280, S-171 77 Stockholm, Sweden.
| | | | | |
Collapse
|
35
|
Abstract
The deterioration of the immune system with progressive aging is believed to contribute to morbidity and mortality in elderly humans due to the increased incidence of infection, autoimmunity, and cancer. Dysregulation of T-cell function is thought to play a critical part in these processes. One of the consequences of an aging immune system is the process termed thymic involution, where the thymus undergoes a progressive reduction in size due to profound changes in its anatomy associated with loss of thymic epithelial cells and a decrease in thymopoiesis. This decline in the output of newly developed T cells results in diminished numbers of circulating naive T cells and impaired cell-mediated immunity. A number of theories have been forwarded to explain this 'thymic menopause' including the possible loss of thymic progenitors or epithelial cells, a diminished capacity to rearrange T-cell receptor genes and alterations in the production of growth factors and hormones. Although to date no interventions fully restore thymic function in the aging host, systemic administration of various cytokines and hormones or bone marrow transplantation have resulted in increased thymic activity and T-cell output with age. In this review, we shall examine the current literature on thymic involution and discuss several interventional strategies currently being explored to restore thymic function in elderly subjects.
Collapse
Affiliation(s)
- Dennis D Taub
- Laboratory of Immunology, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | | |
Collapse
|
36
|
Tuckermann JP, Kleiman A, McPherson KG, Reichardt HM. Molecular mechanisms of glucocorticoids in the control of inflammation and lymphocyte apoptosis. Crit Rev Clin Lab Sci 2005; 42:71-104. [PMID: 15697171 DOI: 10.1080/10408360590888983] [Citation(s) in RCA: 151] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The immune system must be tightly controlled not only to guarantee efficient protection from invading pathogens and oncogenic cells but also to avoid exaggerated immune responses and autoimmunity. This is achieved through interactions amongst leukocytes themselves, by signals from stromal cells and also by various hormones, including glucocorticoids. The glucocorticoids are a class of steroid hormones that exert a wide range of anti-inflammatory and immunosuppressive activities after binding to the glucocorticoid receptor. The power of these hormones was acknowledged many decades ago, and today synthetic derivatives are widely used in the treatment of inflammatory disorders, autoimmunity and cancer. In this review, we summarize our present knowledge of the molecular mechanisms of glucocorticoid action, their influence on specific leukocytes and the induction of thymocyte apoptosis, with an emphasis on how molecular genetics has contributed to our growing, although still incomplete, understanding of these processes.
Collapse
|
37
|
Ko M, Jang J, Ahn J, Lee K, Chung H, Jeon SH, Seong RH. T Cell Receptor Signaling Inhibits Glucocorticoid-induced Apoptosis by Repressing the SRG3 Expression via Ras Activation. J Biol Chem 2004; 279:21903-15. [PMID: 15016814 DOI: 10.1074/jbc.m402144200] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Activation of T cell antigen receptor (TCR) signaling inhibits glucocorticoid (GC)-induced apoptosis of T cells. However, the detailed mechanism regarding how activated T cells are protected from GC-induced apoptosis is unclear. Previously, we have shown that the expression level of SRG3, a murine homolog of BAF155 in humans, correlated well with the GC sensitivity of T cells either in vitro or in vivo. Intriguingly, the expression of SRG3 decreased upon positive selection in the thymus. Here we have shown that TCR signaling inhibits the SRG3 expression via Ras activation and thereby renders primary thymocytes and some thymoma cells resistant to GC-mediated apoptosis. By using pharmacological inhibitors, we have shown that Ras-mediated down-regulation of the SRG3 gene expression is mediated by MEK/ERK and phosphatidylinositol 3-kinase pathways. Moreover, TCR signals repressed the SRG3 transcription through the putative binding sites for E proteins and Ets family transcription factors in the proximal region of the SRG3 promoter. Introduction of mutations in these elements rendered the SRG3 promoter immune to the Ras or TCR signals. Taken together, these observations suggest that TCR signals result in GC desensitization in immature T cells by repressing SRG3 gene expression via Ras activation.
Collapse
MESH Headings
- Animals
- Apoptosis
- Binding Sites
- Binding, Competitive
- Blotting, Northern
- Cell Nucleus/metabolism
- Dexamethasone/pharmacology
- Dose-Response Relationship, Drug
- Down-Regulation
- Enzyme Activation
- Enzyme Inhibitors/pharmacology
- Flavonoids/pharmacology
- Flow Cytometry
- Genes, Reporter
- Glucocorticoids/metabolism
- Glucocorticoids/pharmacology
- Imidazoles/pharmacology
- Immunoblotting
- Kinetics
- Luciferases/metabolism
- Mice
- Mice, Inbred C57BL
- Mice, Transgenic
- Mutagenesis, Site-Directed
- Phosphatidylinositol 3-Kinases/metabolism
- Plasmids/metabolism
- Precipitin Tests
- Promoter Regions, Genetic
- Protein Binding
- Proto-Oncogene Proteins/metabolism
- Proto-Oncogene Proteins c-ets
- Pyridines/pharmacology
- Receptors, Antigen, T-Cell/metabolism
- Repressor Proteins
- Signal Transduction
- Thymus Gland/cytology
- Time Factors
- Trans-Activators/biosynthesis
- Transcription Factors/metabolism
- Transcription, Genetic
- Transfection
- ras Proteins/metabolism
Collapse
Affiliation(s)
- Myunggon Ko
- School of Biological Sciences and Institute of Molecular Biology and Genetics, Seoul National University, Seoul 151-742
| | | | | | | | | | | | | |
Collapse
|
38
|
Zilberman Y, Zafrir E, Ovadia H, Yefenof E, Guy R, Sionov RV. The glucocorticoid receptor mediates the thymic epithelial cell-induced apoptosis of CD4+8+ thymic lymphoma cells. Cell Immunol 2004; 227:12-23. [PMID: 15051511 DOI: 10.1016/j.cellimm.2004.01.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2003] [Accepted: 01/26/2004] [Indexed: 01/24/2023]
Abstract
"Negative selection" and "death by neglect" are governed by apoptotic processes occurring in the thymus that shape the repertoire of maturing T cells. We have previously developed an in vitro model that recapitulates "death by neglect": Co-cultivation of double positive (DP) thymocytes or thymic lymphoma cells (PD1.6) with thymic epithelial cells (TEC) caused TcR-independent apoptosis of the former. We further demonstrated that this apoptosis could be attenuated by aminoglutethimide, an inhibitor of steroid synthesis, suggesting a role of TEC-derived glucocorticoids (GC) in this death process. We have now substantiated the role of the GC-glucocorticoid receptor (GR) axis by using a GC-resistant subline (PD1.6Dex(-)) obtained from the GC-sensitive PD1.6 cells by repeated exposures to increasing doses of dexamethasone (Dex). The PD1.6Dex(-) cells barely express GR and are much less sensitive to TEC-induced apoptosis. Re-expression of GR in PD1.6Dex(-) cells restored their sensitivity to both Dex and TEC, highlighting the central role of GR in these apoptotic processes. Likewise, repeated exposures of PD1.6 cells to TEC led to the selection of TEC-resistant cells (PD1.6TEC(-)) that are insensitive to corticosterone and less sensitive to Dex, though their GR level was only moderately reduced. This is in line with the low levels of corticosterone secreted by TEC. Altogether, our data show that TEC eliminates DP thymic lymphoma cells in a GR-dependent manner and modulates the GC sensitivity of the surviving cells.
Collapse
Affiliation(s)
- Yael Zilberman
- Department of Pharmacology, Faculty of Dental Medicine Founded by the Alpha-Omega Fraternity, Jerusalem, Israel
| | | | | | | | | | | |
Collapse
|
39
|
van den Brandt J, Wang D, Reichardt HM. Resistance of single-positive thymocytes to glucocorticoid-induced apoptosis is mediated by CD28 signaling. Mol Endocrinol 2003; 18:687-95. [PMID: 14701943 DOI: 10.1210/me.2003-0390] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Glucocorticoids administered in pharmacological doses potently induce apoptosis in immature double-positive thymocytes. In contrast, single-positive thymocytes are completely resistant. We now provide evidence that this difference can be attributed to CD28 signaling. When taken into culture, single-positive thymocytes also become sensitive to glucocorticoid-induced apoptosis, which can be prevented by enforced CD28 engagement using a novel type of antibody. This is achieved, at least in part, by transcriptional regulation of apoptosis-related genes such as Bcl-X(L) via a calcium- and phosphatidylinositol 3 kinase-dependent pathway. Accordingly, deficiency of CD28 in genetically engineered mice leads to an increased sensitivity of single-positive thymocytes toward glucocorticoid-induced cell death in vivo. Taken together, we have identified CD28 signaling in the thymus as a key player in determining the differential sensitivity of double-positive and single-positive cells to glucocorticoid action.
Collapse
Affiliation(s)
- Jens van den Brandt
- Institute of Virology and Immunobiology, University of Würzburg, Versbacher Strasse 7, 97078 Würzburg, Germany
| | | | | |
Collapse
|
40
|
Szegezdi E, Kiss I, Simon A, Blaskó B, Reichert U, Michel S, Sándor M, Fésüs L, Szondy Z. Ligation of retinoic acid receptor alpha regulates negative selection of thymocytes by inhibiting both DNA binding of nur77 and synthesis of bim. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2003; 170:3577-84. [PMID: 12646620 DOI: 10.4049/jimmunol.170.7.3577] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Negative selection refers to the selective deletion of autoreactive thymocytes. Its molecular mechanisms have not been well defined. Previous studies in our laboratory have demonstrated that retinoic acids, physiological ligands for the nuclear retinoid receptors, selectively inhibit TCR-mediated death under in vitro conditions, and the inhibition is mediated via the retinoic acid receptor (RAR) alpha. The present studies were undertaken to investigate whether ligation of RARalpha leads to inhibition of TCR-mediated death in vivo and to identify the molecular mechanisms involved. Three models of TCR-mediated death were studied: anti-CD3-mediated death of thymocytes in wild-type mice, and Ag- and bacterial superantigen-driven thymocyte death in TCR-transgenic mice expressing a receptor specific for a fragment of pigeon cytochrome c in the context of the E(k) (class II MHC) molecule. Our data demonstrate that the molecular program of both anti-CD3- and Ag-driven, but not that of superantigen-mediated apoptosis involves up-regulation of nur77, an orphan nuclear receptor, and bim, a BH3-only member of the proapoptotic bcl-2 protein family, proteins previously implicated to participate in the negative selection. Ligation of RARalpha by the synthetic agonist CD336 inhibited apoptosis, DNA binding of nur77, and synthesis of bim induced by anti-CD3 or the specific Ag, but had no effect on the superantigen-driven cell death. Our data imply that retinoids are able to inhibit negative selection in vivo as well, and they interfere with multiple steps of the T cell selection signal pathway.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/administration & dosage
- Apoptosis/drug effects
- Apoptosis/immunology
- Apoptosis Regulatory Proteins
- Bcl-2-Like Protein 11
- Benzoates/administration & dosage
- CD3 Complex/immunology
- CD4-Positive T-Lymphocytes/immunology
- CD4-Positive T-Lymphocytes/metabolism
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/metabolism
- Carrier Proteins/antagonists & inhibitors
- Carrier Proteins/biosynthesis
- Clonal Deletion/drug effects
- Clonal Deletion/immunology
- Columbidae
- Cytochrome c Group/administration & dosage
- Cytochrome c Group/immunology
- DNA-Binding Proteins/antagonists & inhibitors
- DNA-Binding Proteins/biosynthesis
- DNA-Binding Proteins/metabolism
- Enterotoxins/administration & dosage
- Injections, Intraperitoneal
- Ligands
- Male
- Membrane Proteins
- Mice
- Mice, Transgenic
- Nuclear Receptor Subfamily 4, Group A, Member 1
- Protein Binding/drug effects
- Protein Binding/immunology
- Proto-Oncogene Proteins
- Receptors, Cytoplasmic and Nuclear
- Receptors, Retinoic Acid/agonists
- Receptors, Retinoic Acid/metabolism
- Receptors, Retinoic Acid/physiology
- Receptors, Steroid
- Retinoic Acid Receptor alpha
- Retinoids/metabolism
- Staphylococcus aureus/immunology
- Superantigens/administration & dosage
- T-Lymphocyte Subsets/cytology
- T-Lymphocyte Subsets/drug effects
- T-Lymphocyte Subsets/immunology
- T-Lymphocyte Subsets/metabolism
- Tetrahydronaphthalenes/administration & dosage
- Thymus Gland/cytology
- Thymus Gland/drug effects
- Thymus Gland/immunology
- Thymus Gland/metabolism
- Transcription Factors/antagonists & inhibitors
- Transcription Factors/biosynthesis
- Transcription Factors/metabolism
Collapse
Affiliation(s)
- Eva Szegezdi
- Department of Biochemistry and Molecular Biology,University of Debrecen, Debrecen, Hungary
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Khaled AR, Li WQ, Huang J, Fry TJ, Khaled AS, Mackall CL, Muegge K, Young HA, Durum SK. Bax deficiency partially corrects interleukin-7 receptor alpha deficiency. Immunity 2002; 17:561-73. [PMID: 12433363 DOI: 10.1016/s1074-7613(02)00450-8] [Citation(s) in RCA: 76] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
The requirement for cytokines in hematopoiesis is partly attributable to the protection of cells from apoptosis. Since IL-7 is required for normal T cell development, we evaluated the role of Bax in vivo by generating mice deficient in both Bax and the IL-7 receptor alpha chain (IL-7R). Starting at birth, we observed complete recovery of all stages of alphabeta thymocyte development up to 4 weeks of age. However, by 12 weeks of age, thymic cellularity had reverted to that of mice deficient in IL-7R alone. The BH3 only proteins, Bad and Bim, were also part of the death pathway repressed by IL-7. Thus, in young mice, Bax emerges as an essential protein in the death pathway induced by IL-7 deficiency.
Collapse
Affiliation(s)
- Annette R Khaled
- Laboratory of Molecular Immunoregulation, Center for Cancer Research, National Cancer Institute - Frederick, MD 21702, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Jang IK, Hu R, Lacaná E, D'Adamio L, Gu H. Apoptosis-linked gene 2-deficient mice exhibit normal T-cell development and function. Mol Cell Biol 2002; 22:4094-100. [PMID: 12024023 PMCID: PMC133871 DOI: 10.1128/mcb.22.12.4094-4100.2002] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
The apoptosis-linked gene product, ALG-2, is a member of the family of intracellular Ca(2+)-binding proteins and a part of the apoptotic machinery controlled by T-cell receptor (TCR), Fas, and glucocorticoid signals. To explore the physiologic function of ALG-2 in T-cell development and function, we generated mice harboring a null mutation in the alg-2 gene. The alg-2 null mutant mice were viable and fertile and showed neither gross developmental abnormality nor immune dysfunction. Analyses of apoptotic responses of ALG-2-deficient T cells demonstrated that ALG-2 deficiency failed to block apoptosis induced by TCR, Fas, or dexamethasone signals. These findings indicate that ALG-2 is physiologically dispensable for apoptotic responses induced by the above signaling pathways and suggest that other functionally redundant proteins might exist in mammalian cells.
Collapse
Affiliation(s)
- Ihn Kyung Jang
- Laboratory of Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland 20852, USA
| | | | | | | | | |
Collapse
|