1
|
Monsinjon T, Knigge T. Endocrine disrupters affect the immune system of fish: The example of the European seabass. FISH & SHELLFISH IMMUNOLOGY 2025; 162:110303. [PMID: 40180203 DOI: 10.1016/j.fsi.2025.110303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 03/19/2025] [Accepted: 03/29/2025] [Indexed: 04/05/2025]
Abstract
An organism's fitness critically relies on its immune system to provide protection against parasites and pathogens. The immune system has reached its highest complexity in vertebrates, combining the highly specific adaptive with the non-specific innate immunity. In vertebrates, a complex system of steroid hormones regulates major physiological functions comprising energy metabolism, growth, reproduction and immune system performance. This allows the organism to allocate available energy according to life-history traits and environmental conditions, thus maintaining homeostasis and survival of the individual and of the population. Immune system activation must take into account the developmental stage and the nutritional state of the organism. It should respond adequately to different pathogens, but should not overperform or consume all resources for other physiological functions. This important trade-off between immunity and reproduction is balanced by oestrogen. Many of the thousands of chemicals released by humans into the environment, so-called xenobiotics, have the ability to disrupt normal endocrine function. Such endocrine-disrupting chemicals have been demonstrated to impair reproductive functions and to be responsible for numerous diseases in humans and wild life. Given that oestrogens are established modulators of immune cell populations, exogenous oestrogens and oestrogen mimics can modulate immune functions in aquatic animals, such as fish, potentially affecting wildlife and aquaculture. This review highlights the interaction of xenoestrogens with fish immunity. It particularly focusses on the thymus, a major primary immune organ, in the European seabass, Dicentrarchus labrax an important species, both for fisheries and aquaculture.
Collapse
Affiliation(s)
- Tiphaine Monsinjon
- University of Le Havre Normandy, University of Reims Champagne-Ardenne, INERIS, Normandie Univ, FR CNRS 3730 SCALE, UMR I-02 SEBIO, F-76600, Le Havre, France.
| | - Thomas Knigge
- University of Le Havre Normandy, University of Reims Champagne-Ardenne, INERIS, Normandie Univ, FR CNRS 3730 SCALE, UMR I-02 SEBIO, F-76600, Le Havre, France
| |
Collapse
|
2
|
Li S, Wang K, Wu J, Zhu Y. The immunosenescence clock: A new method for evaluating biological age and predicting mortality risk. Ageing Res Rev 2025; 104:102653. [PMID: 39746402 DOI: 10.1016/j.arr.2024.102653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 12/12/2024] [Accepted: 12/29/2024] [Indexed: 01/04/2025]
Abstract
Precisely assessing an individual's immune age is critical for developing targeted aging interventions. Although traditional methods for evaluating biological age, such as the use of cellular senescence markers and physiological indicators, have been widely applied, these methods inherently struggle to capture the full complexity of biological aging. We propose the concept of an 'immunosenescence clock' that evaluates immune system changes on the basis of changes in immune cell abundance and omics data (including transcriptome and proteome data), providing a complementary indicator for understanding age-related physiological transformations. Rather than claiming to definitively measure biological age, this approach can be divided into a biological age prediction clock and a mortality prediction clock. The main function of the biological age prediction clock is to reflect the physiological state through the transcriptome data of peripheral blood mononuclear cells (PBMCs), whereas the mortality prediction clock emphasizes the ability to identify people at high risk of mortality and disease. We hereby present nearly all of the immunosenescence clocks developed to date, as well as their functional differences. Critically, we explicitly acknowledge that no single diagnostic test can exhaustively capture the intricate changes associated with biological aging. Furthermore, as these biological functions are based on the acceleration or delay of immunosenescence, we also summarize the factors that accelerate immunosenescence and the methods for delaying it. A deep understanding of the regulatory mechanisms of immunosenescence can help establish more accurate immune-age models, providing support for personalized longevity interventions and improving quality of life in old age.
Collapse
Affiliation(s)
- Shuyu Li
- Laboratory of Gastroenterology, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Ke Wang
- Department of Breast Surgery, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Jingni Wu
- Department of International Healthcare Center and General Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yongliang Zhu
- Laboratory of Gastroenterology, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
| |
Collapse
|
3
|
Clark MS, Christie M, Jones M, Ashley S, Tang MLK. Seasonal variation in sunlight exposure is differently associated with changes in T regulatory and T-helper 17 cell blood counts in adolescent and adults females: a pilot study. Photochem Photobiol Sci 2025; 24:23-35. [PMID: 39648192 DOI: 10.1007/s43630-024-00668-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 11/25/2024] [Indexed: 12/10/2024]
Abstract
Higher prevalence of multiple sclerosis at higher latitudes is associated with reduced sunlight during childhood. Alterations in inflammatory Th17 and regulatory T cells (Treg) are associated with autoimmunity. In Hobart, Australia (latitude 42.8° south), thirteen girls (aged 12-13) and sixteen women had blood collected in the evening in daylight in February/March, (post 15 h daylight) and at the same time in the dark in August/September (post 9 h daylight). Height and weight were measured. Participants completed online surveys prior around menstruation, sleep, exercise and time outside. Proportions of Th17 (CD4 + , CXCR3-, CCR4 + , CCR6 + , CD161 +), total Treg (CD4 + , CD25 + , CD127low), naïve Treg (CD45RA + , CD4 + , CD25 + , CD127low) and memory Treg (CD45RA low, CD4 + , CD25 + , CD127low) were enumerated by flow cytometry (Cytek Aurora). Hours spent outside was greater in summer than winter (12.5 v 10.5, p = 0.0003). The %Treg/CD4 + was higher in summer than winter (7.3% vs 7%, p = 0.004) including memory Treg (3.1% vs 2.9%, p = 0.02) for all females (n = 29), and naïve Treg were 13% higher in summer in women (3.2% vs 2.8%, p = 0.0009), whereas %Th17 remained unchanged. In women, a negative correlation between the number of hours spent outside in summer and %Th17 was observed (r = - 0.53, p = 0.035). The %Th17 cells were higher in women than girls (4.9% vs 3.1%, p = 0.001), whereas girls had a higher total %Treg (7.6% vs 6.7%, p = 0.005), consisting of a higher naïve Treg (5.5% vs 3.7%, p = 0.0001) while the women had a higher %memory Treg (3.4% vs 2.6%, p = 0.0001). These light-dependent seasonal differences may influence immune development in adolescents.
Collapse
Affiliation(s)
- Margaret S Clark
- ALIS: Adolescent Latitude Immune Study, Melbourne, VIC, Australia.
| | - Michael Christie
- Allergy Immunology, Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, VIC, Australia
| | - Melinda Jones
- ALIS: Adolescent Latitude Immune Study, Melbourne, VIC, Australia
| | - Sarah Ashley
- Allergy Immunology, Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, VIC, Australia
| | - Mimi L K Tang
- Allergy Immunology, Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, VIC, Australia
- Department of Paediatrics, University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
4
|
Zhang J, Guan X, Zhong X. Immunosenescence in digestive system cancers: Mechanisms, research advances, and therapeutic strategies. Semin Cancer Biol 2024; 106-107:234-250. [PMID: 39510149 DOI: 10.1016/j.semcancer.2024.10.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 10/21/2024] [Accepted: 10/30/2024] [Indexed: 11/15/2024]
Abstract
Increasing lifespans and external environmental factors have contributed to the increase of age-related diseases, particularly cancer. A decrease in immune surveillance and clearance of cancer cells is the result of immunosenescence, which involves the remodeling of immune organs, the changes and functional decline of immune cell subsets, in association with systemic low-grade chronic inflammation. Stem cells aging in bone marrow and thymic involution are the most important causes of immunosenescence. Senescent cancer cells promote the differentiation, recruitment, and functional upregulation of immune-suppressive cell subsets e.g. regulatory T cells (Tregs), myeloid-derived suppressor cell (MDSC), tumor-associated macrophages (TAMS) through senescence-associated secretory phenotype (SASP) further exacerbating the immunosuppressive microenvironment. For digestive system cancers, age-related damage to the intestinal mucosal barrier, the aging of gut-associated lymphoid tissue (GALT), exposure to xenobiotic stimuli throughout life, and dysbiosis make the local immune microenvironment more vulnerable. This article systematically reviews the research progress of immunosenescence and immune microenvironment in digestive system cancers, as well as the exploration of related therapy strategies, hoping to point out new directions for research in the digestive system cancers.
Collapse
Affiliation(s)
- Junyan Zhang
- Department of Surgical Oncology and General Surgery, First Affiliated Hospital, China Medical University, Shenyang, China
| | - Xiaojiao Guan
- Department of Pathology, Shengjing Hospital, China Medical University, Shenyang, China.
| | - Xinwen Zhong
- Department of Thoracic Surgery, First Affiliated Hospital, China Medical University, Shenyang, China.
| |
Collapse
|
5
|
Li D, Yao H, Cao X, Han X, Song T, Zeng X. Testosterone regulates thymic remodeling by activating glucocorticoid receptor signaling pathway to accelerate thymocyte apoptosis in male rats. J Reprod Immunol 2024; 164:104288. [PMID: 38924811 DOI: 10.1016/j.jri.2024.104288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 06/11/2024] [Accepted: 06/20/2024] [Indexed: 06/28/2024]
Abstract
Thymic atrophy affects T cell generation and migration to the periphery, thereby affecting T cell pool diversity. However, the mechanisms underlying thymic atrophy have not been fully elucidated. Here, gonadotropin-releasing hormone (GnRH) immunization and surgical castration did not affect thymocyte proliferation, but significantly reduced the apoptosis and increased the survival rate of CD4-CD8-, CD4+CD8+, CD4+CD8-, and CD4-CD8+ thymocytes. Following testosterone supplementation in rats subjected to GnRH immunization and surgical castration, thymocyte proliferation remained unchange, but the apoptosis of CD4-CD8-, CD4+CD8+, CD4+CD8-, and CD4-CD8+ thymocytes significantly increased. Transcriptome analyses of the thymus after GnRH immunization and surgical castration showed a significant reduction in the thymus's response to corticosterone. Cholesterol metabolism and the synthesis and secretion of corticosterone were significantly reduced. Analysis of the enzyme levels involved in the corticosterone synthesis pathway revealed that corticosterone synthesis in thymocytes was significantly reduced after GnRH immunization and surgical castration, whereas exogenous testosterone supplementation relieved this process. Testosterone promoted thymocyte apoptosis in a concentration-dependent manner, and induced corticosterone secretion in vitro. Blocking the intracellular androgen receptor (AR) signaling pathway did not significantly affect thymocyte apoptosis, but blocking the glucocorticoid receptor (GR) signaling pathway significantly reduced it. Our findings indicate that testosterone regulates thymus remodeling by affecting corticosterone synthesis in thymocytes, which activates GR signal transduction and promotes thymocyte apoptosis.
Collapse
Affiliation(s)
- Dong Li
- College of Life Science, Sichuan Agricultural University, Ya'an, Sichuan 625014, PR China
| | - Huan Yao
- College of Life Science, Sichuan Agricultural University, Ya'an, Sichuan 625014, PR China
| | - Xiaohan Cao
- College of Life Science, Sichuan Agricultural University, Ya'an, Sichuan 625014, PR China
| | - Xingfa Han
- College of Life Science, Sichuan Agricultural University, Ya'an, Sichuan 625014, PR China
| | - Tianzeng Song
- Institute of animal science, Tibet Academy of Agricultural and Animal Husbandry Science, Lhasa, Xizang 850009, PR China.
| | - Xianyin Zeng
- College of Life Science, Sichuan Agricultural University, Ya'an, Sichuan 625014, PR China.
| |
Collapse
|
6
|
Kientega T, Marcoux S, Bourbonnais J, Montpetit J, Caru M, Cardin GB, Arbour N, Marcil V, Curnier D, Laverdière C, Sinnett D, Rodier F. Premature thymic functional senescence is a hallmark of childhood acute lymphoblastic leukemia survivorship. Blood Cancer J 2024; 14:96. [PMID: 38871704 PMCID: PMC11176394 DOI: 10.1038/s41408-024-01071-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 05/07/2024] [Accepted: 05/16/2024] [Indexed: 06/15/2024] Open
Abstract
Childhood acute lymphoblastic leukemia (cALL) survivors suffer early-onset chronic diseases classically associated with aging. Normal aging is accompanied by organ dysfunctions, including immunological ones. We hypothesize that thymic immunosenescence occurs in cALL survivors and that its severity may correlate with early-onset chronic diseases. The PETALE study is a cALL survivor cohort with an extensive cardiovascular and metabolic evaluation. The thymic immunosenescence biomarker, signal joint T-cell receptor excision circles (TREC), was evaluated and was highly correlated with age in healthy participants (n = 281) and cALL survivors (n = 248). We observed a systematic thymic immunoage accentuation in each cALL survivor compared to controls ranging from 5.9 to 88.3 years. The immunoage gain was independent of age at diagnosis and treatment modalities and was more severe for females. Thymic aging was associated with several pathophysiological parameters, was greater in survivors suffering from metabolic syndrome, but there was no significant association with global physical condition. The decrease in TREC was independent from blood cell counts, which were normal, suggesting a segmental aging of the thymic compartment. Indeed, increased plasmatic T cell regulatory cytokines IL-6, IL-7 and GM-CSF accompanied high immunoage gain. Our data reveal that cALL or its treatment trigger a rapid immunoage gain followed by further gradual thymic immunosenescence, similar to normal aging. This leads to an enduring shift in accentuated immunoage compared to chronological age. Thus, accentuated thymic immunosenescence is a hallmark of cALL survivorship and TREC levels could be useful immunosenescence biomarkers to help monitoring the health of cancer survivors.
Collapse
Affiliation(s)
- Tibila Kientega
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CHUM), Montréal, QC, Canada
- Institut du cancer de Montréal, Montréal, QC, Canada
| | - Sophie Marcoux
- Université Laval, Département de médecine sociale et préventive, Québec, QC, Canada
- Centre de recherche du Centre hospitalier universitaire de Québec-Université Laval, Québec, QC, Canada
| | - Jessica Bourbonnais
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CHUM), Montréal, QC, Canada
- Institut du cancer de Montréal, Montréal, QC, Canada
| | - Jade Montpetit
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CHUM), Montréal, QC, Canada
- Institut du cancer de Montréal, Montréal, QC, Canada
| | - Maxime Caru
- Centre de recherche Azrieli du CHU Sainte-Justine, Montréal, QC, Canada
- Department of Pediatrics, Division of Hematology and Oncology, Penn State College of Medicine, Hershey, PA, USA
| | - Guillaume B Cardin
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CHUM), Montréal, QC, Canada
- Institut du cancer de Montréal, Montréal, QC, Canada
| | - Nathalie Arbour
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CHUM), Montréal, QC, Canada
- Université de Montréal, Département de Neurosciences, Montréal, QC, Canada
| | - Valérie Marcil
- Centre de recherche Azrieli du CHU Sainte-Justine, Montréal, QC, Canada
- Université de Montréal, Département de Nutrition, Montréal, QC, Canada
| | - Daniel Curnier
- Centre de recherche Azrieli du CHU Sainte-Justine, Montréal, QC, Canada
- Université de Montréal, Faculté de médecine, École de kinésiologie et des sciences de l'activité physique, Laboratoire de physiopathologie de l'exercice (LPEX), Montréal, QC, Canada
| | - Caroline Laverdière
- Centre de recherche Azrieli du CHU Sainte-Justine, Montréal, QC, Canada
- Université de Montréal, Département de Pédiatrie, Montréal, QC, Canada
| | - Daniel Sinnett
- Centre de recherche Azrieli du CHU Sainte-Justine, Montréal, QC, Canada
- Université de Montréal, Département de Pédiatrie, Montréal, QC, Canada
| | - Francis Rodier
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CHUM), Montréal, QC, Canada.
- Institut du cancer de Montréal, Montréal, QC, Canada.
- Université de Montréal, Département de Radiologie, radio-oncologie et médecine nucléaire, Montréal, QC, Canada.
| |
Collapse
|
7
|
Zelleroth S, Stam F, Nylander E, Kjellgren E, Gising J, Larhed M, Grönbladh A, Hallberg M. The decanoate esters of nandrolone, testosterone, and trenbolone induce steroid specific memory impairment and somatic effects in the male rat. Horm Behav 2024; 161:105501. [PMID: 38368844 DOI: 10.1016/j.yhbeh.2024.105501] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 01/26/2024] [Accepted: 02/05/2024] [Indexed: 02/20/2024]
Abstract
Long-term use of anabolic androgenic steroids (AAS) in supratherapeutic doses is associated with severe adverse effects, including physical, mental, and behavioral alterations. When used for recreational purposes several AAS are often combined, and in scientific studies of the physiological impact of AAS either a single compound or a cocktail of several steroids is often used. Because of this, steroid-specific effects have been difficult to define and are not fully elucidated. The present study used male Wistar rats to evaluate potential somatic and behavioral effects of three different AAS; the decanoate esters of nandrolone, testosterone, and trenbolone. The rats were exposed to 15 mg/kg of nandrolone decanoate, testosterone decanoate, or trenbolone decanoate every third day for 24 days. Body weight gain and organ weights (thymus, liver, kidney, testis, and heart) were measured together with the corticosterone plasma levels. Behavioral effects were studied in the novel object recognition-test (NOR-test) and the multivariate concentric square field-test (MCSF-test). The results conclude that nandrolone decanoate, but neither testosterone decanoate nor trenbolone decanoate, caused impaired recognition memory in the NOR-test, indicating an altered cognitive function. The behavioral profile and stress hormone level of the rats were not affected by the AAS treatments. Furthermore, the study revealed diverse AAS-induced somatic effects i.e., reduced body weight development and changes in organ weights. Of the three AAS included in the study, nandrolone decanoate was identified to cause the most prominent impact on the male rat, as it affected body weight development, the weights of multiple organs, and caused an impaired memory function.
Collapse
Affiliation(s)
- Sofia Zelleroth
- The Beijer Laboratory, Department of Pharmaceutical Biosciences, Neuropharmacology and Addiction Research, SE-751 24, Uppsala University, Sweden.
| | - Frida Stam
- The Beijer Laboratory, Department of Pharmaceutical Biosciences, Neuropharmacology and Addiction Research, SE-751 24, Uppsala University, Sweden.
| | - Erik Nylander
- The Beijer Laboratory, Department of Pharmaceutical Biosciences, Neuropharmacology and Addiction Research, SE-751 24, Uppsala University, Sweden.
| | - Ellinor Kjellgren
- The Beijer Laboratory, Department of Pharmaceutical Biosciences, Neuropharmacology and Addiction Research, SE-751 24, Uppsala University, Sweden
| | - Johan Gising
- The Beijer Laboratory, Science for Life Laboratory, Department of Medicinal Chemistry, SE-751 23, Uppsala University, Sweden.
| | - Mats Larhed
- The Beijer Laboratory, Science for Life Laboratory, Department of Medicinal Chemistry, SE-751 23, Uppsala University, Sweden.
| | - Alfhild Grönbladh
- The Beijer Laboratory, Department of Pharmaceutical Biosciences, Neuropharmacology and Addiction Research, SE-751 24, Uppsala University, Sweden.
| | - Mathias Hallberg
- The Beijer Laboratory, Department of Pharmaceutical Biosciences, Neuropharmacology and Addiction Research, SE-751 24, Uppsala University, Sweden.
| |
Collapse
|
8
|
Li YR, Zúñiga-Pflücker JC. Thymus aging and immune reconstitution, progresses and challenges. Semin Immunol 2023; 70:101837. [PMID: 37659170 DOI: 10.1016/j.smim.2023.101837] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 08/25/2023] [Accepted: 08/25/2023] [Indexed: 09/04/2023]
Abstract
Thymus is a primary lymphoid organ essential for the development of T lymphocytes. Age-related thymic involution is a prominent feature of immune senescence. The thymus undergoes rapid growth during fetal and neonatal development, peaks in size before puberty and then begins to undergo a decrease in cellularity with age. Dramatic changes occur with age-associated thymic involution. The most prominent features of thymic involution include: (i) epithelial structure disruption, (ii) adipogenesis, and (iii) thymocyte development arrest. There is a sex disparity in thymus aging. It is a multifactorial process controlled and regulated by a series of molecules, including the transcription factor FOXN1, fibroblast and keratinocyte growth factors (FGF and KGF, respectively), sex steroids, Notch signaling, WNT signaling, and microRNAs. Nevertheless, there is still no satisfactory evolutionary or physiological explanation for age-associated thymic involution, and understanding the precise mechanism(s) for thymus aging remains challenging. Sustained thymic regeneration has yet to be achieved by sex steroid ablation. Recent preclinical studies indicate that long-term thymic reconstitution can be achieved via adoptive transfer of in vitro-generated progenitor T (proT) cells, and improvements in the methods for the generation of human proT cells make this an attractive approach. Future clinical applications may rely on new applications integrating proT cells, cytokine support and sex-steroid inhibition treatments.
Collapse
Affiliation(s)
- Yue Ru Li
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Juan Carlos Zúñiga-Pflücker
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada; Biological Sciences, Sunnybrook Research Institute, Toronto, Ontario, Canada.
| |
Collapse
|
9
|
Li D, Yao H, Han X, Cao X, Du X, Meng F, Bu G, Kong F, Song T, Zeng X. Active immunization against gonadotropin-releasing hormone affects thymic T cell production, migration, and colonization in male rat lymphoid tissue. J Reprod Immunol 2023; 159:104132. [PMID: 37591181 DOI: 10.1016/j.jri.2023.104132] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 07/19/2023] [Accepted: 08/08/2023] [Indexed: 08/19/2023]
Abstract
Active immunization against gonadotropin-releasing hormone (GnRH) inhibits animal reproduction and has become a friendly alternative to surgical castration, which has been reported to affect the proportion of thymic T cell subpopulations. The effects of active immunization against GnRH on T cell migration from the thymus to the periphery and T cell distribution in lymphoid tissues remain unclear. Here, we showed that active immunization against GnRH increased thymic size and weight, enlarged the number of thymocytes, and enhanced CD4+ recent thymic emigrants (RTEs) and CD8+ RTEs migration to the blood and spleen. Active immunization against GnRH had no significant effect on naïve CD4+, naïve CD8+, CD4+ memory/activated, or CD8+ memory/activated T cells. In addition, active immunization against GnRH increased the proportion of CD3+ T cells in the spleen and lymph nodes. The percentages of CD3+CD4+ and CD3+CD8+ T cells in the blood, spleen, and lymph nodes were not significantly affected by GnRH immunization. Overall, these results enhance our understanding of thymic T cell production, migration, and colonization in rat lymphoid tissues affected by GnRH immunization.
Collapse
Affiliation(s)
- Dong Li
- College of Life Science, Sichuan Agricultural University, Ya'an 625014, Sichuan, PR China
| | - Huan Yao
- College of Life Science, Sichuan Agricultural University, Ya'an 625014, Sichuan, PR China
| | - Xinfa Han
- College of Life Science, Sichuan Agricultural University, Ya'an 625014, Sichuan, PR China
| | - Xiaohan Cao
- College of Life Science, Sichuan Agricultural University, Ya'an 625014, Sichuan, PR China
| | - Xiaogang Du
- College of Life Science, Sichuan Agricultural University, Ya'an 625014, Sichuan, PR China
| | - Fengyan Meng
- College of Life Science, Sichuan Agricultural University, Ya'an 625014, Sichuan, PR China
| | - Guixian Bu
- College of Life Science, Sichuan Agricultural University, Ya'an 625014, Sichuan, PR China
| | - Fanli Kong
- College of Life Science, Sichuan Agricultural University, Ya'an 625014, Sichuan, PR China
| | - Tianzeng Song
- Institute of animal science, Tibet academy of Agricultural and Animal Husbandry Science, Lhasa 850009, Xizang, PR China.
| | - Xianyin Zeng
- College of Life Science, Sichuan Agricultural University, Ya'an 625014, Sichuan, PR China.
| |
Collapse
|
10
|
Lagou MK, Karagiannis GS. Obesity-induced thymic involution and cancer risk. Semin Cancer Biol 2023; 93:3-19. [PMID: 37088128 DOI: 10.1016/j.semcancer.2023.04.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 04/19/2023] [Accepted: 04/20/2023] [Indexed: 04/25/2023]
Abstract
Declining thymic functions associated either with old age (i.e., age-related thymic involution), or with acute involution as a result of stress, infectious disease, or cytoreductive therapies (e.g., chemotherapy/radiotherapy), have been associated with cancer development. A key mechanism underlying such increased cancer risk is the thymus-dependent debilitation of adaptive immunity, which is responsible for orchestrating immunoediting mechanisms and tumor immune surveillance. In the past few years, a blooming set of evidence has intriguingly linked obesity with cancer development and progression. The majority of such studies has focused on obesity-driven chronic inflammation, steroid/sex hormone and adipokine production, and hyperinsulinemia, as principal factors affecting the tumor microenvironment and driving the development of primary malignancy. However, experimental observations about the negative impact of obesity on T cell development and maturation have existed for more than half a century. Here, we critically discuss the molecular and cellular mechanisms of obesity-driven thymic involution as a previously underrepresented intermediary pathology leading to cancer development and progression. This knowledge could be especially relevant in the context of childhood obesity, because impaired thymic function in young individuals leads to immune system abnormalities, and predisposes to various pediatric cancers. A thorough understanding behind the molecular and cellular circuitries governing obesity-induced thymic involution could therefore help towards the rationalized development of targeted thymic regeneration strategies for obese individuals at high risk of cancer development.
Collapse
Affiliation(s)
- Maria K Lagou
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, USA; Tumor Microenvironment of Metastasis Program, Albert Einstein Cancer Center, Bronx, NY, USA
| | - George S Karagiannis
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, USA; Tumor Microenvironment of Metastasis Program, Albert Einstein Cancer Center, Bronx, NY, USA; Cancer Dormancy and Tumor Microenvironment Institute, Albert Einstein College of Medicine, Bronx, NY, USA; Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY, USA; Integrated Imaging Program for Cancer Research, Albert Einstein College of Medicine, Bronx, NY, USA.
| |
Collapse
|
11
|
Hanson ED, Sakkal S, Bates-Fraser LC, Que S, Cho E, Spielmann G, Kadife E, Violet JA, Battaglini CL, Stoner L, Bartlett DB, McConell GK, Hayes A. Acute exercise induces distinct quantitative and phenotypical T cell profiles in men with prostate cancer. Front Sports Act Living 2023; 5:1173377. [PMID: 37325799 PMCID: PMC10266416 DOI: 10.3389/fspor.2023.1173377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 05/12/2023] [Indexed: 06/17/2023] Open
Abstract
Background Reduced testosterone levels can influence immune system function, particularly T cells. Exercise during cancer reduces treatment-related side effects and provide a stimulus to mobilize and redistribute immune cells. However, it is unclear how conventional and unconventional T cells (UTC) respond to acute exercise in prostate cancer survivors compared to healthy controls. Methods Age-matched prostate cancer survivors on androgen deprivation therapy (ADT) and those without ADT (PCa) along with non-cancer controls (CON) completed ∼45 min of intermittent cycling with 3 min at 60% of peak power interspersed by 1.5 min of rest. Fresh, unstimulated immune cell populations and intracellular perforin were assessed before (baseline), immediately following (0 h), 2 h, and 24 h post-exercise. Results At 0 h, conventional T cell counts increased by 45%-64% with no differences between groups. T cell frequency decreased by -3.5% for CD3+ and -4.5% for CD4+ cells relative to base at 0 h with CD8+ cells experiencing a delayed decrease of -4.5% at 2 h with no group differences. Compared to CON, the frequency of CD8+CD57+ cells was -18.1% lower in ADT. Despite a potential decrease in maturity, ADT increased CD8+perforin+ GMFI. CD3+Vα7.2+CD161+ counts, but not frequencies, increased by 69% post-exercise while CD3+CD56+ cell counts increased by 127% and were preferentially mobilized (+1.7%) immediately following the acute cycling bout. There were no UTC group differences. Cell counts and frequencies returned to baseline by 24 h. Conclusion Following acute exercise, prostate cancer survivors demonstrate normal T cell and UTC responses that were comparable to CON. Independent of exercise, ADT is associated with lower CD8+ cell maturity (CD57) and perforin frequency that suggests a less mature phenotype. However, higher perforin GMFI may attenuate these changes, with the functional implications of this yet to be determined.
Collapse
Affiliation(s)
- Erik D. Hanson
- Department of Exercise & Sport Science, University of North Carolina, Chapel Hill, NC, United States
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, United States
- Human Movement Science Curriculum, University of North Carolina, Chapel Hill, NC, United States
- Institute for Health and Sport, Victoria University, Melbourne, VIC, Australia
| | - Samy Sakkal
- Institute for Health and Sport, Victoria University, Melbourne, VIC, Australia
| | - Lauren C. Bates-Fraser
- Department of Exercise & Sport Science, University of North Carolina, Chapel Hill, NC, United States
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, United States
- Human Movement Science Curriculum, University of North Carolina, Chapel Hill, NC, United States
| | - Shadney Que
- Institute for Health and Sport, Victoria University, Melbourne, VIC, Australia
| | - Eunhan Cho
- School of Kinesiology, Louisiana State University, Baton Rouge, LA, United States
| | - Guillaume Spielmann
- School of Kinesiology, Louisiana State University, Baton Rouge, LA, United States
| | - Elif Kadife
- Institute for Health and Sport, Victoria University, Melbourne, VIC, Australia
| | - John A. Violet
- Division of Radiation Oncology and Cancer Imaging, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Claudio L. Battaglini
- Department of Exercise & Sport Science, University of North Carolina, Chapel Hill, NC, United States
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, United States
- Human Movement Science Curriculum, University of North Carolina, Chapel Hill, NC, United States
| | - Lee Stoner
- Department of Exercise & Sport Science, University of North Carolina, Chapel Hill, NC, United States
- Human Movement Science Curriculum, University of North Carolina, Chapel Hill, NC, United States
| | - David B. Bartlett
- School of Biosciences and Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford, United Kingdom
| | - Glenn K. McConell
- Institute for Health and Sport, Victoria University, Melbourne, VIC, Australia
| | - Alan Hayes
- Institute for Health and Sport, Victoria University, Melbourne, VIC, Australia
- Australian Institute for Musculoskeletal Science (AIMSS), Victoria University, Melbourne, VIC, Australia
- Department of Medicine—Western Health, Melbourne Medical School, The University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
12
|
McCombe PA, Greer JM. Effects of biological sex and pregnancy in experimental autoimmune encephalomyelitis: It's complicated. Front Immunol 2022; 13:1059833. [PMID: 36518769 PMCID: PMC9742606 DOI: 10.3389/fimmu.2022.1059833] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Accepted: 11/03/2022] [Indexed: 11/29/2022] Open
Abstract
Experimental autoimmune encephalomyelitis (EAE) can be induced in many animal strains by inoculation with central nervous system antigens and adjuvant or by the passive transfer of lymphocytes reactive with these antigens and is widely used as an animal model for multiple sclerosis (MS). There are reports that female sex and pregnancy affect EAE. Here we review the effects of biological sex and the effects of pregnancy on the clinical features (including disease susceptibility) and pathophysiology of EAE. We also review reports of the possible mechanisms underlying these differences. These include sex-related differences in the immune system and in the central nervous system, the effects of hormones and the sex chromosomes and molecules unique to pregnancy. We also review sex differences in the response to factors that can modify the course of EAE. Our conclusion is that the effects of biological sex in EAE vary amongst animal models and should not be widely extrapolated. In EAE, it is therefore essential that studies looking at the effects of biological sex or pregnancy give full information about the model that is used (i.e. animal strain, sex, the inducing antigen, timing of EAE induction in relation to pregnancy, etc.). In addition, it would be preferable if more than one EAE model were used, to show if any observed effects are generalizable. This is clearly a field that requires further work. However, understanding of the mechanisms of sex differences could lead to greater understanding of EAE, and suggest possible therapies for MS.
Collapse
Affiliation(s)
| | - Judith M. Greer
- UQ Centre for Clinical Research, The University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
13
|
Lagou MK, Anastasiadou DP, Karagiannis GS. A Proposed Link Between Acute Thymic Involution and Late Adverse Effects of Chemotherapy. Front Immunol 2022; 13:933547. [PMID: 35844592 PMCID: PMC9283860 DOI: 10.3389/fimmu.2022.933547] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Accepted: 05/31/2022] [Indexed: 11/13/2022] Open
Abstract
Epidemiologic data suggest that cancer survivors tend to develop a protuberant number of adverse late effects, including second primary malignancies (SPM), as a result of cytotoxic chemotherapy. Besides the genotoxic potential of these drugs that directly inflict mutational burden on genomic DNA, the precise mechanisms contributing to SPM development are poorly understood. Cancer is nowadays perceived as a complex process that goes beyond the concept of genetic disease and includes tumor cell interactions with complex stromal and immune cell microenvironments. The cancer immunoediting theory offers an explanation for the development of nascent neoplastic cells. Briefly, the theory suggests that newly emerging tumor cells are mostly eliminated by an effective tissue immunosurveillance, but certain tumor variants may occasionally escape innate and adaptive mechanisms of immunological destruction, entering an equilibrium phase, where immunologic tumor cell death "equals" new tumor cell birth. Subsequent microenvironmental pressures and accumulation of helpful mutations in certain variants may lead to escape from the equilibrium phase, and eventually cause an overt neoplasm. Cancer immunoediting functions as a dedicated sentinel under the auspice of a highly competent immune system. This perspective offers the fresh insight that chemotherapy-induced thymic involution, which is characterized by the extensive obliteration of the sensitive thymic epithelial cell (TEC) compartment, can cause long-term defects in thymopoiesis and in establishment of diverse T cell receptor repertoires and peripheral T cell pools of cancer survivors. Such delayed recovery of T cell adaptive immunity may result in prolonged hijacking of the cancer immunoediting mechanisms, and lead to development of persistent and mortal infections, inflammatory disorders, organ-specific autoimmunity lesions, and SPMs. Acknowledging that chemotherapy-induced thymic involution is a potential risk factor for the emergence of SPM demarcates new avenues for the rationalized development of pharmacologic interventions to promote thymic regeneration in patients receiving cytoreductive chemotherapies.
Collapse
Affiliation(s)
- Maria K. Lagou
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, United States
- Tumor Microenvironment and Metastasis Program, Albert Einstein Cancer Center, Bronx, NY, United States
| | - Dimitra P. Anastasiadou
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, United States
- Tumor Microenvironment and Metastasis Program, Albert Einstein Cancer Center, Bronx, NY, United States
| | - George S. Karagiannis
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, United States
- Tumor Microenvironment and Metastasis Program, Albert Einstein Cancer Center, Bronx, NY, United States
- Cancer Dormancy and Tumor Microenvironment Institute, Albert Einstein Cancer Center, Bronx, NY, United States
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY, United States
- Integrated Imaging Program, Albert Einstein College of Medicine, Bronx, NY, United States
| |
Collapse
|
14
|
Kernen L, Phan A, Bo J, Herzog EL, Huynh J, Segner H, Baumann L. Estrogens as immunotoxicants: 17α-ethinylestradiol exposure retards thymus development in zebrafish (Danio rerio). AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2022; 242:106025. [PMID: 34837781 DOI: 10.1016/j.aquatox.2021.106025] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 11/10/2021] [Indexed: 06/13/2023]
Abstract
Estrogenic endocrine disrupting compounds (EEDCs) can cause alterations in sexual development and reproductive function of fish. Growing evidence suggests that EEDCs can also interfere with development and function of innate immunity of fish. The present study examined a potential disruptive effect of EEDCs at field-relevant concentrations on the development of adaptive immunity, more specifically the thymus. Zebrafish (Danio rerio) were exposed from fertilization until 64 days post-fertilization (dpf) to environmentally relevant (3 and 10 ng/L) concentrations of the synthetic estrogen 17α-ethinylestradiol (EE2). The exposure duration covered the period of initial thymus differentiation to maximum growth. Thymus development was assessed by histological and morphometric (thymus area) analysis, thymocyte number, and transcript levels of thymocyte marker genes. Additionally, transcript levels of the estrogen receptors (esr1 and esr2a) were determined. The EE2 exposure altered sexual development (gonad differentiation, transcript levels of hepatic vitellogenin and estrogen receptors) of zebrafish, as expected. At the same time, the EE2 treatment reduced the thymus growth (thymus area, thymocyte number) and transcript levels of thymus marker genes. The expression of the thymic estrogen receptors responded to the EE2 exposure but in a different pattern than the hepatic estrogen receptors. After the 64-day-exposure period, the juvenile fish were transferred into clean water for another 95 days to assess the reversibility of EE2-induced effects. The thymic alterations were found to be reversible in female zebrafish but persisted in males. The present study provides the first evidence that the development of the fish adaptive immune system is sensitive to EEDCs, and that this takes place at concentrations similar to those that disrupt sexual development.
Collapse
Affiliation(s)
- Larissa Kernen
- Centre for Fish and Wildlife Health, University of Bern, Länggassstrasse 122, 3012 Bern, Switzerland
| | - Audrey Phan
- Centre for Fish and Wildlife Health, University of Bern, Länggassstrasse 122, 3012 Bern, Switzerland
| | - Jun Bo
- Laboratory of Marine Biology and Ecology, Third Institute of Oceanography, Xiamen 361102, China
| | - Elio L Herzog
- Centre for Fish and Wildlife Health, University of Bern, Länggassstrasse 122, 3012 Bern, Switzerland
| | - John Huynh
- Centre for Fish and Wildlife Health, University of Bern, Länggassstrasse 122, 3012 Bern, Switzerland
| | - Helmut Segner
- Centre for Fish and Wildlife Health, University of Bern, Länggassstrasse 122, 3012 Bern, Switzerland
| | - Lisa Baumann
- Centre for Fish and Wildlife Health, University of Bern, Länggassstrasse 122, 3012 Bern, Switzerland; Aquatic Ecology & Toxicology, Centre for Organismal Studies, University of Heidelberg, Im Neuenheimer Feld 504, 69120 Heidelberg, Germany.
| |
Collapse
|
15
|
Hu C, Zhang K, Jiang F, Wang H, Shao Q. Epigenetic modifications in thymic epithelial cells: an evolutionary perspective for thymus atrophy. Clin Epigenetics 2021; 13:210. [PMID: 34819170 PMCID: PMC8612001 DOI: 10.1186/s13148-021-01197-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 11/08/2021] [Indexed: 02/06/2023] Open
Abstract
Background The thymic microenvironment is mainly comprised of thymic epithelial cells, the cytokines, exosomes, surface molecules, and hormones from the cells, and plays a vital role in the development, differentiation, maturation and homeostasis of T lymphocytes. However, the thymus begins to degenerate as early as the second year of life and continues through aging in human beings, leading to a decreased output of naïve T cells, the limited TCR diversity and an expansion of monoclonal memory T cells in the periphery organs. These alternations will reduce the adaptive immune response to tumors and emerging infectious diseases, such as COVID-19, also it is easier to suffer from autoimmune diseases in older people. In the context of global aging, it is important to investigate and clarify the causes and mechanisms of thymus involution. Main body Epigenetics include histone modification, DNA methylation, non-coding RNA effects, and chromatin remodeling. In this review, we discuss how senescent thymic epithelial cells determine and control age-related thymic atrophy, how this process is altered by epigenetic modification. How the thymus adipose influences the dysfunctions of the thymic epithelial cells, and the prospects of targeting thymic epithelial cells for the treatment of thymus atrophy. Conclusion Epigenetic modifications are emerging as key regulators in governing the development and senescence of thymic epithelial cells. It is beneficial to re-establish effective thymopoiesis, identify the potential therapeutic strategy and rejuvenate the immune function in the elderly.
Collapse
Affiliation(s)
- Cexun Hu
- Department of Immunology, School of Medicine, Jiangsu University, Zhenjiang, 212013, Jiangsu, People's Republic of China.,Department of Immunology, Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, No. 301 Xuefu Road, Zhenjiang, 212013, Jiangsu, People's Republic of China
| | - Keyu Zhang
- Department of Immunology, School of Medicine, Jiangsu University, Zhenjiang, 212013, Jiangsu, People's Republic of China.,Department of Immunology, Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, No. 301 Xuefu Road, Zhenjiang, 212013, Jiangsu, People's Republic of China
| | - Feng Jiang
- Department of Immunology, School of Medicine, Jiangsu University, Zhenjiang, 212013, Jiangsu, People's Republic of China.,Department of Immunology, Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, No. 301 Xuefu Road, Zhenjiang, 212013, Jiangsu, People's Republic of China
| | - Hui Wang
- Department of Immunology, School of Medicine, Jiangsu University, Zhenjiang, 212013, Jiangsu, People's Republic of China. .,Department of Immunology, Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, No. 301 Xuefu Road, Zhenjiang, 212013, Jiangsu, People's Republic of China.
| | - Qixiang Shao
- Department of Immunology, School of Medicine, Jiangsu University, Zhenjiang, 212013, Jiangsu, People's Republic of China. .,Department of Immunology, Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, No. 301 Xuefu Road, Zhenjiang, 212013, Jiangsu, People's Republic of China. .,Jiangsu College of Nursing, School of Medical Science and Laboratory Medicine, Huai'an, 223002, Jiangsu, People's Republic of China.
| |
Collapse
|
16
|
Moreira C, Hétru J, Paiola M, Duflot A, Chan P, Vaudry D, Pinto PIS, Monsinjon T, Knigge T. Proteomic changes in the extracellular environment of sea bass thymocytes exposed to 17α-ethinylestradiol in vitro. COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY D-GENOMICS & PROTEOMICS 2021; 40:100911. [PMID: 34583305 DOI: 10.1016/j.cbd.2021.100911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Revised: 08/14/2021] [Accepted: 08/28/2021] [Indexed: 11/28/2022]
Abstract
The thymus is an important immune organ providing the necessary microenvironment for the development of a diverse, self-tolerant T cell repertoire, which is selected to allow for the recognition of foreign antigens while avoiding self-reactivity. Thymus function and activity are known to be regulated by sex steroid hormones, such as oestrogen, leading to sexual dimorphisms in immunocompetence between males and females. The oestrogenic modulation of the thymic function provides a potential target for environmental oestrogens, such as 17α-ethynylestradiol (EE2), to interfere with the cross-talk between the endocrine and the immune system. Oestrogen receptors have been identified on thymocytes and the thymic microenvironment, but it is unclear how oestrogens regulate thymic epithelial and T cell communication including paracrine signalling. Much less is known regarding intrathymic signalling in fish. Secretomics allows for the analysis of complex mixtures of immunomodulatory signalling factors secreted by T cells. Thus, in the present study, isolated thymocytes of the European sea bass, Dicentrarchus labrax, were exposed in vitro to 30 nM EE2 for 4 h and the T cell-secretome (i.e., extracellular proteome) was analysed by quantitative label-free mass-spectrometry. Progenesis revealed a total of 111 proteins differentially displayed between EE2-treated and control thymocytes at an α-level of 5% and a 1.3-fold change cut off (n = 5-6). The EE2-treatment significantly decreased the level of 90 proteins. Gene ontology revealed the proteasome to be the most impacted pathway. In contrast, the abundance of 21 proteins was significantly increased, with cathepsins showing the highest level of induction. However, no particular molecular pathway was significantly altered for these upregulated proteins. To the best of our knowledge, this work represents the first study of the secretome of the fish thymus exposed to the environmental oestrogen EE2, highlighting the impact on putative signalling pathways linked to immune surveillance, which may be of crucial importance for fish health and defence against pathogens.
Collapse
Affiliation(s)
- Catarina Moreira
- Normandie Univ, UNILEHAVRE, FR CNRS 3730 SCALE, UMR-I 02 Environmental Stress and Aquatic Biomonitoring (SEBIO), F-76600 Le Havre, France
| | - Julie Hétru
- Normandie Univ, UNILEHAVRE, FR CNRS 3730 SCALE, UMR-I 02 Environmental Stress and Aquatic Biomonitoring (SEBIO), F-76600 Le Havre, France
| | - Matthieu Paiola
- Normandie Univ, UNILEHAVRE, FR CNRS 3730 SCALE, UMR-I 02 Environmental Stress and Aquatic Biomonitoring (SEBIO), F-76600 Le Havre, France; Department of Microbiology and Immunology, University of Rochester Medical Center, 14642 Rochester, NY, United States
| | - Aurélie Duflot
- Normandie Univ, UNILEHAVRE, FR CNRS 3730 SCALE, UMR-I 02 Environmental Stress and Aquatic Biomonitoring (SEBIO), F-76600 Le Havre, France
| | - Philippe Chan
- Normandie Univ, UNIROUEN, PISSARO Proteomic Facility, IRIB, F-76820 Mont-Saint-Aignan, France; Normandie Univ, UNIROUEN, Institute for Research and Innovation in Biomedicine (IRIB), F-76183 Rouen, France
| | - David Vaudry
- Normandie Univ, UNIROUEN, PISSARO Proteomic Facility, IRIB, F-76820 Mont-Saint-Aignan, France; Normandie Univ, UNIROUEN, Neuronal and Neuroendocrine Differentiation and Communication (DC2N), Inserm U1239, 76821 Mont-Saint-Aignan, France; Normandie Univ, UNIROUEN, Institute for Research and Innovation in Biomedicine (IRIB), F-76183 Rouen, France
| | - Patrícia I S Pinto
- Centro de Ciências Do Mar (CCMAR), Universidade Do Algarve, 8005-139 Faro, Portugal
| | - Tiphaine Monsinjon
- Normandie Univ, UNILEHAVRE, FR CNRS 3730 SCALE, UMR-I 02 Environmental Stress and Aquatic Biomonitoring (SEBIO), F-76600 Le Havre, France
| | - Thomas Knigge
- Normandie Univ, UNILEHAVRE, FR CNRS 3730 SCALE, UMR-I 02 Environmental Stress and Aquatic Biomonitoring (SEBIO), F-76600 Le Havre, France.
| |
Collapse
|
17
|
Wu H, Li X, Zhou C, Yu Q, Ge S, Pan Z, Zhao Y, Xia S, Zhou X, Liu X, Wang H, Shao Q. Circulating mature dendritic cells homing to the thymus promote thymic epithelial cells involution via the Jagged1/Notch3 axis. Cell Death Discov 2021; 7:225. [PMID: 34462426 PMCID: PMC8404188 DOI: 10.1038/s41420-021-00619-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 08/13/2021] [Accepted: 08/20/2021] [Indexed: 11/24/2022] Open
Abstract
Multiple proinflammatory conditions, including chemotherapy, radiotherapy, transplant rejection, and microbial infections, have been identified to induce involution of the thymus. However, the underlying cellular and molecular mechanisms of these inflammatory conditions inducing apoptosis of thymic epithelial cells (TECs), the main components of the thymus, remain largely unknown. In the circulation, mature dendritic cells (mDCs), the predominant initiator of innate and adaptive immune response, can migrate into the thymus. Herein, we demonstrated that mDCs were able to directly inhibit TECs proliferation and induce their apoptosis by activating the Jagged1/Notch3 signaling pathway. Intrathymic injection of either mDCs or recombinant mouse Jagged1-human Fc fusion protein (rmJagged1-hFc) into mice resulted in acute atrophy of the thymus. Furthermore, DAPT, a γ-secretase inhibitor, reversed the effects induced by mDC or rmJagged1-hFc. These findings suggest that acute or aging-related thymus degeneration can be induced either by mass migration of circulating mDCs in a short period of time or by a few but constantly homing mDCs.
Collapse
Affiliation(s)
- Haojie Wu
- Reproductive Sciences Institute, Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Immunology, School of Medicine, Jiangsu University, Zhenjiang, 212013, Jiangsu, P. R. China
| | - Xiaohan Li
- Reproductive Sciences Institute, Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Immunology, School of Medicine, Jiangsu University, Zhenjiang, 212013, Jiangsu, P. R. China
| | - Chen Zhou
- Reproductive Sciences Institute, Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Immunology, School of Medicine, Jiangsu University, Zhenjiang, 212013, Jiangsu, P. R. China
| | - Qihong Yu
- Reproductive Sciences Institute, Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Immunology, School of Medicine, Jiangsu University, Zhenjiang, 212013, Jiangsu, P. R. China
| | - Shiyao Ge
- Reproductive Sciences Institute, Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Immunology, School of Medicine, Jiangsu University, Zhenjiang, 212013, Jiangsu, P. R. China
| | - Zihui Pan
- Reproductive Sciences Institute, Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Immunology, School of Medicine, Jiangsu University, Zhenjiang, 212013, Jiangsu, P. R. China
| | - Yangjing Zhao
- Reproductive Sciences Institute, Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Immunology, School of Medicine, Jiangsu University, Zhenjiang, 212013, Jiangsu, P. R. China
| | - Sheng Xia
- Reproductive Sciences Institute, Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Immunology, School of Medicine, Jiangsu University, Zhenjiang, 212013, Jiangsu, P. R. China
| | - Xiaoming Zhou
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Pathology, School of Medicine, Jiangsu University, Zhenjiang, 212013, Jiangsu, P. R. China
| | - Xia Liu
- Reproductive Sciences Institute, Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Immunology, School of Medicine, Jiangsu University, Zhenjiang, 212013, Jiangsu, P. R. China
| | - Hui Wang
- Reproductive Sciences Institute, Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Immunology, School of Medicine, Jiangsu University, Zhenjiang, 212013, Jiangsu, P. R. China.
| | - Qixiang Shao
- Reproductive Sciences Institute, Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Immunology, School of Medicine, Jiangsu University, Zhenjiang, 212013, Jiangsu, P. R. China.
- Institute of Medical Genetics and Reproductive Immunity, School of Medical Science and Laboratory Medicine, Jiangsu College of Nursing, Huai'an, 223002, Jiangsu, P. R. China.
| |
Collapse
|
18
|
Paiola M, Moreira C, Hétru J, Duflot A, Pinto PIS, Scapigliati G, Knigge T, Monsinjon T. Prepubertal gonad investment modulates thymus function: evidence in a teleost fish. J Exp Biol 2021; 224:238091. [PMID: 33789987 DOI: 10.1242/jeb.238576] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 01/20/2021] [Indexed: 12/12/2022]
Abstract
Thymus plasticity following gonadectomy or sex hormone replacement has long since exemplified sex hormone effects on the immune system in mammals and, to a lesser extent, in 'lower vertebrates', including amphibians and fish. Nevertheless, the underlying physiological significances as well as the ontogenetic establishment of this crosstalk remain largely unknown. Here, we used a teleost fish, the European sea bass, Dicentrarchus labrax, to investigate: (1) whether the regulation of thymus plasticity relies on resource trade-off with somatic growth and reproductive investment and (2) if the gonad-thymus interaction takes place during gonadal differentiation and development. Because gonadal development and, supposedly, thymus function in sea bass depend on environmental changes associated with the winter season, we evaluated thymus changes (foxn1 expression, and thymocyte and T cell content) in juvenile D. labrax raised for 1 year under either constant or fluctuating photoperiod and temperature. Importantly, in both conditions, intensive gonadal development following sex differentiation coincided with a halt of thymus growth, while somatic growth continued. To the best of our knowledge, this is the first study showing that gonadal development during prepuberty regulates thymus plasticity. This finding may provide an explanation for the initiation of the thymus involution related to ageing in mammals. Comparing fixed and variable environmental conditions, our work also demonstrates that the extent of the effects on the thymus, which are related to reproduction, depend on ecophysiological conditions, rather than being directly related to sexual maturity and sex hormone levels.
Collapse
Affiliation(s)
- Matthieu Paiola
- Normandy University, FR CNRS 3730 SCALE, UMR-I 02 INERIS-URCA-ULH Environmental Stress and Aquatic Biomonitoring (SEBIO), University of Le Havre Normandy, 76600 Le Havre, France
| | - Catarina Moreira
- Normandy University, FR CNRS 3730 SCALE, UMR-I 02 INERIS-URCA-ULH Environmental Stress and Aquatic Biomonitoring (SEBIO), University of Le Havre Normandy, 76600 Le Havre, France
| | - Julie Hétru
- Normandy University, FR CNRS 3730 SCALE, UMR-I 02 INERIS-URCA-ULH Environmental Stress and Aquatic Biomonitoring (SEBIO), University of Le Havre Normandy, 76600 Le Havre, France
| | - Aurélie Duflot
- Normandy University, FR CNRS 3730 SCALE, UMR-I 02 INERIS-URCA-ULH Environmental Stress and Aquatic Biomonitoring (SEBIO), University of Le Havre Normandy, 76600 Le Havre, France
| | - Patricia I S Pinto
- Laboratory of Comparative Endocrinology and Integrative Biology, CCMAR - Centre of Marine Sciences, University of Algarve, 8005-139 Faro, Portugal
| | - Giuseppe Scapigliati
- Department for Innovation in Biological, Agro-food and Forest Systems, Tuscia University, 01100 Viterbo, Italy
| | - Thomas Knigge
- Normandy University, FR CNRS 3730 SCALE, UMR-I 02 INERIS-URCA-ULH Environmental Stress and Aquatic Biomonitoring (SEBIO), University of Le Havre Normandy, 76600 Le Havre, France
| | - Tiphaine Monsinjon
- Normandy University, FR CNRS 3730 SCALE, UMR-I 02 INERIS-URCA-ULH Environmental Stress and Aquatic Biomonitoring (SEBIO), University of Le Havre Normandy, 76600 Le Havre, France
| |
Collapse
|
19
|
Kellogg C, Equils O. The role of the thymus in COVID-19 disease severity: implications for antibody treatment and immunization. Hum Vaccin Immunother 2021; 17:638-643. [PMID: 33064620 PMCID: PMC7993178 DOI: 10.1080/21645515.2020.1818519] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 08/27/2020] [Indexed: 12/20/2022] Open
Abstract
The thymus is a largely neglected organ but plays a significant role in the regulation of adaptive immune responses. The effect of aging on the thymus and immune senescence is well established, and the resulting inflammaging is found to be implicated in the development of many chronic diseases including atherosclerosis, hypertension and type 2 diabetes. Both aging and diseases of inflammaging are associated with severe COVID-19 disease, and a dysfunctional thymus may be a predisposing factor. In addition, insults on the thymus during childhood may lead to abnormal thymic function and may explain severe COVID-19 disease among younger individuals; therefore, measurement of thymic function may assist COVID-19 care. Those with poor thymic function may be treated prophylactically with convalescent serum or recombinant antibodies, and they may respond better to high-dose or adjuvanted COVID-19 vaccines. Treatments inducing thymic regeneration may improve patients' overall health and may be incorporated in COVID-19 management.
Collapse
Affiliation(s)
- Caitlyn Kellogg
- University of California, San Diego School of Medicine, San Diego, CA, USA
- Public Health Education , MiOra Foundation, Los Angeles, CA, USA
| | - Ozlem Equils
- Public Health Education , MiOra Foundation, Los Angeles, CA, USA
| |
Collapse
|
20
|
Yuan L, Cao J, Wang Z, Zhang L, Wang X, Wu Y, Dong J, Xie H, Lin X. Fetal thymus in the middle and late trimesters: Morphometry and development using post-mortem 3.0T MRI. Exp Ther Med 2020; 20:43. [PMID: 32952634 PMCID: PMC7480123 DOI: 10.3892/etm.2020.9172] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Accepted: 01/30/2020] [Indexed: 12/16/2022] Open
Abstract
The present study aimed to investigate the anatomical microstructure, features and signals of the fetal thymus by 3.0T FS-T2 weighted turbo spin echo sequences, which could provide imaging evidence for the evaluation of early-stage development of fetal thymus. In addition, the T2-weighted three-dimensional (3D) sequences and the 3D processing may contribute to the establishment of reference ranges for the fetal thymus. A total of 64 specimens obtained from the fetuses of 16-39 weeks of gestational age (GA) were scanned by 3.0T MRI. Morphological changes and quantitative measurements of the fetal thymus were assessed, including the anteroposterior diameter, width, height, surface area and volume. The shape of fetal thymus varied and the majority were X-shaped, with a narrow top and wide bottom. Morphology measurements demonstrated gradual growth with increasing GA. There were high linear correlations between width, height, surface area and volume and GA. No significant differences were observed between the sexes. Post-mortem 3.0T MRI clearly demonstrated changes in external contours and internal structure with GA. The images and data obtained reflect normal development of the fetal thymus and enrich the imaging data of fetal morphometry.
Collapse
Affiliation(s)
- Leilei Yuan
- Shandong University School of Medicine, Jinan, Shandong 250012, P.R. China.,Department of Radiology, Taian Central Hospital, Taian, Shandong 271000, P.R. China
| | - Jinfeng Cao
- Central Hospital of Zibo, Zibo, Shandong 255020, P.R. China
| | - Zhaohua Wang
- Department of Radiology, Taian Central Hospital, Taian, Shandong 271000, P.R. China
| | - Litao Zhang
- Department of Radiology, Taian Central Hospital, Taian, Shandong 271000, P.R. China
| | - Xia Wang
- Department of Radiology, Taian Central Hospital, Taian, Shandong 271000, P.R. China
| | - Yong Wu
- Department of MR, Shandong Medical Imaging Research Institute, Jinan, Shandong 250021, P.R. China
| | - Jinye Dong
- Department of MR, Shandong Medical Imaging Research Institute, Jinan, Shandong 250021, P.R. China
| | - Huihui Xie
- Department of MR, Shandong Medical Imaging Research Institute, Jinan, Shandong 250021, P.R. China
| | - Xiangtao Lin
- Shandong University School of Medicine, Jinan, Shandong 250012, P.R. China.,Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250012, P.R. China
| |
Collapse
|
21
|
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
|
22
|
Lavaert M, Valcke B, Vandekerckhove B, Leclercq G, Liang KL, Taghon T. Conventional and Computational Flow Cytometry Analyses Reveal Sustained Human Intrathymic T Cell Development From Birth Until Puberty. Front Immunol 2020; 11:1659. [PMID: 32849574 PMCID: PMC7417369 DOI: 10.3389/fimmu.2020.01659] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Accepted: 06/22/2020] [Indexed: 11/13/2022] Open
Abstract
The thymus is the organ where subsets of mature T cells are generated which subsequently egress to function as central mediators in the immune system. While continuously generating T cells even into adulthood, the thymus does undergo involution during life. This is characterized by an initial rapid decrease in thymic cellularity during early life and by a second age-dependent decline in adulthood. The thymic cellularity of neonates remains low during the first month after birth and the tissue reaches a maximum in cellularity at 6 months of age. In order to study the effect that this first phase of thymic involution has on thymic immune subset frequencies, we performed multi-color flow cytometry on thymic samples collected from birth to 14 years of age. In consideration of the inherent limitations posed by conventional flow cytometry analysis, we established a novel computational analysis pipeline that is adapted from single-cell transcriptome sequencing data analysis. This allowed us to overcome technical effects by batch correction, analyze multiple samples simultaneously, limit computational cost by subsampling, and to rely on KNN-graphs for graph-based clustering. As a result, we successfully identified rare, distinct and gradually developing immune subsets within the human thymus tissues. Although the thymus undergoes early involution from infanthood onwards, our data suggests that this does not affect human T-cell development as we did not observe significant alterations in the proportions of T-lineage developmental intermediates from birth to puberty. Thus, in addition to providing an interesting novel strategy to analyze conventional flow cytometry data for the thymus, our work shows that the early phase of human thymic involution mainly limits the overall T cell output since no obvious changes in thymocyte subsets could be observed.
Collapse
Affiliation(s)
- Marieke Lavaert
- Department of Diagnostic Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Brecht Valcke
- Department of Diagnostic Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Bart Vandekerckhove
- Department of Diagnostic Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Georges Leclercq
- Department of Diagnostic Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Kai Ling Liang
- Department of Diagnostic Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Tom Taghon
- Department of Diagnostic Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| |
Collapse
|
23
|
Blagosklonny MV. From causes of aging to death from COVID-19. Aging (Albany NY) 2020; 12:10004-10021. [PMID: 32534452 PMCID: PMC7346074 DOI: 10.18632/aging.103493] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 06/08/2020] [Indexed: 12/19/2022]
Abstract
COVID-19 is not deadly early in life, but mortality increases exponentially with age, which is the strongest predictor of mortality. Mortality is higher in men than in women, because men age faster, and it is especially high in patients with age-related diseases, such as diabetes and hypertension, because these diseases are manifestations of aging and a measure of biological age. At its deepest level, aging (a program-like continuation of developmental growth) is driven by inappropriately high cellular functioning. The hyperfunction theory of quasi-programmed aging explains why COVID-19 vulnerability (lethality) is an age-dependent syndrome, linking it to other age-related diseases. It also explains inflammaging and immunosenescence, hyperinflammation, hyperthrombosis, and cytokine storms, all of which are associated with COVID-19 vulnerability. Anti-aging interventions, such as rapamycin, may slow aging and age-related diseases, potentially decreasing COVID-19 vulnerability.
Collapse
|
24
|
Merrheim J, Villegas J, Van Wassenhove J, Khansa R, Berrih-Aknin S, le Panse R, Dragin N. Estrogen, estrogen-like molecules and autoimmune diseases. Autoimmun Rev 2020; 19:102468. [PMID: 31927086 DOI: 10.1016/j.autrev.2020.102468] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Accepted: 09/23/2019] [Indexed: 12/13/2022]
Abstract
In western countries, the slope of autoimmune disease (AD) incidence is increasing and affects 5-8% of the population. Mainly prevalent in women, these pathologies are due to thymic tolerance processes breakdown. The female sex hormone, estrogen, is involved in this AD female susceptibility. However, predisposition factors have to act in concert with unknown triggering environmental factors (virus, microbiota, pollution) to initiate AD. Individuals are exposed to various environmental compounds that display endocrine disruption abilities. The cellular effects of some of these molecules may be mediated through the aryl hydrocarbon receptor (AhR). Here, we review the effects of these molecules on the homeostasis of the thymic cells, the immune tolerance intrinsic factors (transcription factors, epigenetic marks) and on the immune tolerance extrinsic factors (microbiota, virus sensibility). This review highlights the contribution of estrogen and endocrine disruptors on the dysregulation of mechanisms sustaining AD development.
Collapse
Affiliation(s)
- Judith Merrheim
- Sorbonne Université, Paris, France; Inserm UMRS 974, Paris, France; AIM, Institute of Myology, Paris, France; Centre de Recherche en Myologie, Sorbonne Université, Inserm UMRS 974, Hôpital La Pitié- Salpêtrière, 105 Bd de l'hôpital, 75013 Paris, France
| | - José Villegas
- Sorbonne Université, Paris, France; Inserm UMRS 974, Paris, France; AIM, Institute of Myology, Paris, France; Centre de Recherche en Myologie, Sorbonne Université, Inserm UMRS 974, Hôpital La Pitié- Salpêtrière, 105 Bd de l'hôpital, 75013 Paris, France
| | - Jérôme Van Wassenhove
- Sorbonne Université, Paris, France; Inserm UMRS 974, Paris, France; AIM, Institute of Myology, Paris, France; Centre de Recherche en Myologie, Sorbonne Université, Inserm UMRS 974, Hôpital La Pitié- Salpêtrière, 105 Bd de l'hôpital, 75013 Paris, France
| | - Rémi Khansa
- Sorbonne Université, Paris, France; Inserm UMRS 974, Paris, France; AIM, Institute of Myology, Paris, France; Centre de Recherche en Myologie, Sorbonne Université, Inserm UMRS 974, Hôpital La Pitié- Salpêtrière, 105 Bd de l'hôpital, 75013 Paris, France
| | - Sonia Berrih-Aknin
- Sorbonne Université, Paris, France; Inserm UMRS 974, Paris, France; AIM, Institute of Myology, Paris, France; Centre de Recherche en Myologie, Sorbonne Université, Inserm UMRS 974, Hôpital La Pitié- Salpêtrière, 105 Bd de l'hôpital, 75013 Paris, France
| | - Rozen le Panse
- Sorbonne Université, Paris, France; Inserm UMRS 974, Paris, France; AIM, Institute of Myology, Paris, France; Centre de Recherche en Myologie, Sorbonne Université, Inserm UMRS 974, Hôpital La Pitié- Salpêtrière, 105 Bd de l'hôpital, 75013 Paris, France
| | - Nadine Dragin
- Sorbonne Université, Paris, France; Inserm UMRS 974, Paris, France; Inovarion, Paris, France; Centre de Recherche en Myologie, Sorbonne Université, Inserm UMRS 974, Hôpital La Pitié- Salpêtrière, 105 Bd de l'hôpital, 75013 Paris, France.
| |
Collapse
|
25
|
Brown MA, Su MA. An Inconvenient Variable: Sex Hormones and Their Impact on T Cell Responses. THE JOURNAL OF IMMUNOLOGY 2020; 202:1927-1933. [PMID: 30885988 DOI: 10.4049/jimmunol.1801403] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Accepted: 12/05/2018] [Indexed: 12/15/2022]
Abstract
Epidemiologic data demonstrate sex differences in autoimmune diseases, immune responses against infection, and antitumor immunity, and accumulating evidence suggests a major role for sex hormones in mediating these differences. In this study, we review recent advances in understanding how sex hormones regulate T cell responses to alter susceptibility to autoimmunity. Although sex hormones can directly alter gene transcriptional programs of T cells, we focus in this study on how sex hormones alter T cell development and function through their effects on thymic stromal cells and innate cell types. In addition to contributing to our understanding of sex differences, these findings also have implications for the therapeutic use of sex hormones and sex hormone modulators, which are now being prescribed to increasing numbers of patients for a wide variety of indications.
Collapse
Affiliation(s)
- Melissa A Brown
- Department of Microbiology and Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611
| | - Maureen A Su
- Department of Microbiology, Immunology and Molecular Genetics, David Geffen School of Medicine, College of Life Sciences, University of California at Los Angeles, Los Angeles, CA 90095; and .,Department of Pediatrics, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA 90095
| |
Collapse
|
26
|
Benagiano M, Bianchi P, D'Elios MM, Brosens I, Benagiano G. Autoimmune diseases: Role of steroid hormones. Best Pract Res Clin Obstet Gynaecol 2019; 60:24-34. [PMID: 31047850 DOI: 10.1016/j.bpobgyn.2019.03.001] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 03/01/2019] [Indexed: 02/06/2023]
Abstract
Autoimmune diseases (AIDs) are a heterogeneous group of disorders in terms of clinical manifestations, pathogenesis, and prevalence, and there is no agreement to date on a common classification. Adaptive immune responses are responsible for the existence of AIDs, although innate immunity is also involved in misguiding the immune response against self-antigens. Hormones, in general, and in particular steroid hormones, play a critical role in the physiology and pathology of the immune system, especially in adaptive immunity. Hormonal factors, alone or in relation to age, sex, and reproductive status, are involved in conditioning the onset of a number of AIDs. There is a well-defined sexual dimorphism for human AIDs. At the same time, the classic view has been that steroid hormones have well-defined effects, with one type, estrogens, being "pro-inflammatory" and the other two progestogens (progesterone and its synthetic analogs) and androgens being "anti-inflammatory." Although this view has been considered too simplistic and seems contradicted by numerous observations, it remains valid: progestogens and androgens are immunosuppressive and therefore protective against AIDs, whereas estrogens are immune-stimulatory and therefore pathogenic in AIDs.
Collapse
Affiliation(s)
- Marisa Benagiano
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Paola Bianchi
- Department of Medico-Surgical Sciences and Translational Medicine, Sant'Andrea Hospital, Sapienza, University of Rome, Rome, Italy.
| | - Mario Milco D'Elios
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Ivo Brosens
- Faculty of Medicine, Catholic University of Leuven, Leuven, Belgium
| | - Giuseppe Benagiano
- Department of Obstetrics, Gynaecology and Urology, Policlinico Umberto I, Sapienza, University of Rome, Rome, Italy
| |
Collapse
|
27
|
Abstract
The role of immune system is to protect the organism from the not built-in program-like alterations inside and against the agents penetrating from outside (bacteria, viruses, and protozoa). These functions were developed and formed during the evolution. Considering these functions, the immune system promotes the lengthening of lifespan and helps longevity. However, some immune functions have been conveyed by men to medical tools (e.g., pharmaceuticals, antibiotics, and prevention), especially in our modern age, which help the struggle against microbes, but evolutionarily weaken the immune system. Aging is a gradual slow attrition by autoimmunity, directed by the thymus and regulated by the central nervous system and pineal gland. Considering this, thymus could be a pacemaker of aging. The remodeling of the immune system, which can be observed in elderly people and centenarians, is probably not a cause of aging, but a consequence of it, which helps to suit immunity to the requirements. Oxidative stress also helps the attrition of the immune cells and antioxidants help to prolong lifespan. There are gender differences in the aging of the immune system as well as in the longevity. There is an advantage for women in both cases. This can be explained by hormonal differences (estrogens positively influences both processes); however, social factors are also not excluded. The endocrine disruptor chemicals act similar to estrogens, like stimulating or suppressing immunity and provoking autoimmunity; however, their role in longevity is controversial. There are some drugs (rapamycin, metformin, and selegiline) and antioxidants (as vitamins C and E) that prolong lifespan and also improve immunity. It is difficult to declare that longevity is exclusively dependent on the state of the immune system; however, there is a parallelism between the state of immune system and lifespan. It seems likely that there is not a real decline of immunity during aging, but there is a remodeling of the system according to the claims of senescence. This is manifested in the remaining (sometimes stronger) function of memory cells in contrast to the production and number of the new antigen-reactive naive T-cells.
Collapse
Affiliation(s)
- György Csaba
- 1 Department of Genetics, Cell- and Immunobiology, Semmelweis University, Budapest, Hungary
| |
Collapse
|
28
|
Shi M, Zhou H, Lei M, Chen L, Zellmer L, He Y, Yang W, Xu N, Liao DJ. Spontaneous Cancers, But Not Many Induced Ones in Animals, Resemble Semi-New Organisms that Possess a Unique Programmed Cell Death Mode Different from Apoptosis, Senescent Death, Necrosis and Stress-Induced Cell Death. J Cancer 2018; 9:4726-4735. [PMID: 30588258 PMCID: PMC6299389 DOI: 10.7150/jca.26502] [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: 04/05/2018] [Accepted: 09/11/2018] [Indexed: 01/26/2023] Open
Abstract
There are four basic cell death modes in animals, i.e. physiological senescent death (SD) and apoptosis as well as pathological necrosis and stress-induced cell death (SICD). There have been numerous publications describing “apoptosis” in cancer, mostly focused on killing cancer cells using radio- or chemo-therapy, with few on exploring how cancer cells die naturally without such treatments. Spontaneous benign or malignant neoplasms are immortal and autonomous, but they still retain some allegiance to their parental tissue or organ and thus are still somewhat controlled by the patient's body. Because of these properties of immortality, semi-autonomy, and semi-allegiance to the patient's body, spontaneous tumors have no redundant cells and resemble “semi-new organisms” parasitizing the patients, becoming a unique tissue type possessing a hitherto unannotated cell death mode besides SD, apoptosis, necrosis and SICD. Particularly, apoptosis aims to expunge redundant cells, whereas this new mode does not. In contrast to spontaneous tumors, many histologically malignant tumors induced in experimental animals, before they reach an advanced stage, regress after withdrawal of the inducer. This mortal and non-autonomous nature disqualifies these animal lesions as authentic neoplasms and as semi-new organisms but makes them a good tissue type for apoptosis studies. Ruminating over cell death in spontaneous cancers and many inauthentic tumors induced in animals from these new slants makes us realize that “whether cancer cells undergo apoptosis” is not an easy question with a simple answer. Our answer is that cancer cells have an uncharacterized programmed cell death mode, which is not apoptosis.
Collapse
Affiliation(s)
- Mingjun Shi
- Department of Pathophysiology, Guizhou Medical University, Guiyang 550025, Guizhou Province, China
| | - Haiyan Zhou
- Clinical Research Center, Guizhou Medical University Hospital, Guiyang 550004, Guizhou Province, China
| | - Mingjuan Lei
- Hormel Institute, University of Minnesota, Austin, MN 55912, USA
| | - Lichan Chen
- Hormel Institute, University of Minnesota, Austin, MN 55912, USA
| | - Lucas Zellmer
- Masonic Cancer Center, University of Minnesota, 435 E. River Road, Minneapolis, MN 55455, USA
| | - Yan He
- Key Lab of Endemic and Ethnic Diseases of the Ministry of Education of China at Guizhou Medical University, Guiyang 550004, Guizhou Province, China
| | - Wenxiu Yang
- Department of Pathology, Guizhou Medical University Hospital, Guiyang 550004, Guizhou province, China
| | - Ningzhi Xu
- Laboratory of Cell and Molecular Biology & State Key Laboratory of Molecular Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100021, China
| | - Dezhong Joshua Liao
- Key Lab of Endemic and Ethnic Diseases of the Ministry of Education of China at Guizhou Medical University, Guiyang 550004, Guizhou Province, China.,Department of Pathology, Guizhou Medical University Hospital, Guiyang 550004, Guizhou province, China
| |
Collapse
|
29
|
Dou X, Chen L, Lei M, Zellmer L, Jia Q, Ling P, He Y, Yang W, Liao DJ. Evaluating the Remote Control of Programmed Cell Death, with or without a Compensatory Cell Proliferation. Int J Biol Sci 2018; 14:1800-1812. [PMID: 30443184 PMCID: PMC6231223 DOI: 10.7150/ijbs.26962] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 08/24/2018] [Indexed: 12/23/2022] Open
Abstract
Organisms and their different component levels, whether organelle, cellular or other, come by birth and go by death, and the deaths are often balanced by new births. Evolution on the one hand has built demise program(s) in cells of organisms but on the other hand has established external controls on the program(s). For instance, evolution has established death program(s) in animal cells so that the cells can, when it is needed, commit apoptosis or senescent death (SD) in physiological situations and stress-induced cell death (SICD) in pathological situations. However, these programmed cell deaths are not predominantly regulated by the cells that do the dying but, instead, are controlled externally and remotely by the cells' superior(s), i.e. their host tissue or organ or even the animal's body. Currently, it is still unclear whether a cell has only one death program or has several programs respectively controlling SD, apoptosis and SICD. In animals, apoptosis exterminates, in a physiological manner, healthy but no-longer needed cells to avoid cell redundancy, whereas suicidal SD and SICD, like homicidal necrosis, terminate ill but useful cells, which may be followed by regeneration of the live cells and by scar formation to heal the damaged organ or tissue. Therefore, “who dies” clearly differentiates apoptosis from SD, SICD and necrosis. In animals, apoptosis can occur only in those cell types that retain a lifelong ability of proliferation and never occurs in those cell types that can no longer replicate in adulthood. In cancer cells, SICD is strengthened, apoptosis is dramatically weakened while SD has been lost. Most published studies professed to be about apoptosis are actually about SICD, which has four basic and well-articulated pathways involving caspases or involving pathological alterations in the mitochondria, endoplasmic reticula, or lysosomes.
Collapse
Affiliation(s)
- Xixi Dou
- Key Laboratory of Biopharmaceuticals, Shandong Academy of Pharmaceutical Sciences, Jinan 250101, Shandong Province, P.R. China.,Technology Center, Shandong Freda Pharmaceutical Group, Jinan 250101, Shandong Province, P.R. China
| | - Lichan Chen
- College of Chemical Engineering, Huaqiao University, Xiamen 361021, Fujian Province, P.R. China
| | - Mingjuan Lei
- Hormel Institute, University of Minnesota, Austin, MN 55912, USA
| | - Lucas Zellmer
- Masonic Cancer Center, University of Minnesota, 435 E. River Road, Minneapolis, MN 55455, USA
| | - Qingwen Jia
- Key Laboratory of Biopharmaceuticals, Shandong Academy of Pharmaceutical Sciences, Jinan 250101, Shandong Province, P.R. China
| | - Peixue Ling
- Key Laboratory of Biopharmaceuticals, Shandong Academy of Pharmaceutical Sciences, Jinan 250101, Shandong Province, P.R. China.,Technology Center, Shandong Freda Pharmaceutical Group, Jinan 250101, Shandong Province, P.R. China
| | - Yan He
- Key Lab of Endemic and Ethnic Diseases of the Ministry of Education of China in Guizhou Medical University, Guiyang 550004, Guizhou Province, P.R. China
| | - Wenxiu Yang
- Department of Pathology, Guizhou Medical University Hospital, Guiyang 550004, Guizhou province, P.R. China
| | - Dezhong Joshua Liao
- Key Lab of Endemic and Ethnic Diseases of the Ministry of Education of China in Guizhou Medical University, Guiyang 550004, Guizhou Province, P.R. China.,Department of Pathology, Guizhou Medical University Hospital, Guiyang 550004, Guizhou province, P.R. China
| |
Collapse
|
30
|
Wei C, Guo D, Li Y, Zhang K, Liang G, Li Y, Ma Y, Liu J, Li Y. Profiling analysis of 17β-estradiol-regulated lncRNAs in mouse thymic epithelial cells. Physiol Genomics 2018; 50:553-562. [DOI: 10.1152/physiolgenomics.00098.2017] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Thymus is the primary organ for T cell differentiation and maturation. Many studies have demonstrated that estrogen plays a crucial role in thymic epithelial cell (TEC) proliferation during thymic involution. LncRNAs are involved in various biological processes; however, estrogen-mediated lncRNA expression in TECs has not been yet reported. To address this question, the mouse medullary thymic epithelial cell line 1 (MTEC1) was treated with 17β-estradiol (E2). By using CCK8 assay and flow cytometry, we found that E2 was able to inhibit viability and proliferation of MTEC1 cells. The expression profiles of lncRNAs in MTEC1 cells with or without E2 treatment were then measured by RNA-Seq, and a total of 962 lncRNAs and 2,469 mRNAs were shown to be differentially expressed. The reliability of RNA-Seq was confirmed by quantitative RT-PCR. Correlation analysis was conducted to investigate the potential function of lncRNAs. According to gene ontology (GO) analysis, differentially expressed lncRNAs were mainly related to cell proliferation, cell cycle and cell apoptosis. KEGG pathway analysis indicated that these lncRNAs were associated with several pathways, namely immunological activity, metabolism and cytokine-cytokine receptor interaction. In conclusion, our study provided a novel direction for studying the relationship between lncRNAs and E2 in the thymus.
Collapse
Affiliation(s)
- Chaonan Wei
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Dongguang Guo
- School of Life Science and Technology, Xinxiang University, Xinxiang, China
| | - Ying Li
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Kaizhao Zhang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Guan Liang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Ying Li
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Yongjiang Ma
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Jilong Liu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Yugu Li
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| |
Collapse
|
31
|
Paiola M, Knigge T, Duflot A, Pinto PIS, Farcy E, Monsinjon T. Oestrogen, an evolutionary conserved regulator of T cell differentiation and immune tolerance in jawed vertebrates? DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2018; 84:48-61. [PMID: 29408048 DOI: 10.1016/j.dci.2018.01.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Revised: 01/18/2018] [Accepted: 01/18/2018] [Indexed: 06/07/2023]
Abstract
In teleosts, as in mammals, the immune system is tightly regulated by sexual steroid hormones, such as oestrogens. We investigated the effects of 17β-oestradiol on the expression of several genes related to T cell development and resulting T cell subpopulations in sea bass, Dicentrarchus labrax, for a primary lymphoid organ, the thymus, and two secondary lymphoid organs, the head-kidney and the spleen. In parallel, the oxidative burst capacity was assessed in leucocytes of the secondary lymphoid organs. Apoptosis- and proliferation-related genes, indicative of B and T cell clonal selection and lymphoid progenitor activity, were not affected by elevated oestrogen-levels. Sex-related oestrogen-responsiveness in T cell and antigen-presenting cell markers was observed, the expression of which was differentially induced by oestrogen-exposure in the three lymphoid organs. Remarkably, in the spleen, oestrogen increased regulatory T cell-related gene expression was associated with a decrease in oxidative burst capacity. To the best of our knowledge, this study indicates for the first time that physiological levels of oestrogen are likely to promote immune tolerance by modulating thymic function (i.e., T cell development and output) and peripheral T cells in teleosts, similar to previously reported oestrogenic effects in mammals.
Collapse
Affiliation(s)
- Matthieu Paiola
- Normandy University, FR CNRS 3730 SCALE, UMR-I 02 INERIS-URCA-ULH Environmental Stress and Aquatic Biomonitoring (SEBIO), University of Le Havre Normandy, 76600 Le Havre, France
| | - Thomas Knigge
- Normandy University, FR CNRS 3730 SCALE, UMR-I 02 INERIS-URCA-ULH Environmental Stress and Aquatic Biomonitoring (SEBIO), University of Le Havre Normandy, 76600 Le Havre, France
| | - Aurélie Duflot
- Normandy University, FR CNRS 3730 SCALE, UMR-I 02 INERIS-URCA-ULH Environmental Stress and Aquatic Biomonitoring (SEBIO), University of Le Havre Normandy, 76600 Le Havre, France
| | - Patricia I S Pinto
- Laboratory of Comparative Endocrinology and Integrative Biology, CCMAR - Centre of Marine Sciences, University of Algarve, 8005-139 Faro, Portugal
| | - Emilie Farcy
- Montpellier University, UMR MARBEC (UM, CNRS, Ifremer, IRD), 34095 Montpellier, France
| | - Tiphaine Monsinjon
- Normandy University, FR CNRS 3730 SCALE, UMR-I 02 INERIS-URCA-ULH Environmental Stress and Aquatic Biomonitoring (SEBIO), University of Le Havre Normandy, 76600 Le Havre, France.
| |
Collapse
|
32
|
The effects of supplemental vitamin E on hematological parameters in a rat model of ovarian hormone deficiency. Menopause 2018; 25:336-342. [DOI: 10.1097/gme.0000000000001003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
33
|
Salpietro V, Polizzi A, Recca G, Ruggieri M. The role of puberty and adolescence in the pathobiology of pediatric multiple sclerosis. ACTA ACUST UNITED AC 2018. [DOI: 10.1186/s40893-017-0032-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
|
34
|
Berrih-Aknin S, Panse RL, Dragin N. AIRE: a missing link to explain female susceptibility to autoimmune diseases. Ann N Y Acad Sci 2018; 1412:21-32. [PMID: 29291257 DOI: 10.1111/nyas.13529] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Revised: 09/20/2017] [Accepted: 09/25/2017] [Indexed: 12/13/2022]
Abstract
Women are more susceptible to autoimmune diseases than men. Autoimmunity results from tolerance breakdown toward self-components. Recently, three transcription modulators were identified in medullary thymic epithelial cells that orchestrate immune central tolerance processes: the autoimmune regulator (AIRE), FEZ family zinc finger 2 (FEZF2 or FEZ1), and PR domain zinc finger protein 1 (PRDM1). Interestingly, these three transcription modulators regulate nonredundant tissue-specific antigen subsets and thus cover broad antigen diversity. Recent data from different groups demonstrated that sex hormones (estrogen and testosterone) are involved in the regulation of thymic AIRE expression in humans and mice through direct transcriptional modulation and epigenetic changes. As a consequence, AIRE displays gender-biased thymic expression, with females showing a lower expression compared with males, a finding that could explain the female susceptibility to autoimmune diseases. So far, FEZF2 has not been related to an increased gender bias in autoimmune disease. PRDM1 expression has not been shown to display gender-differential thymic expression, but its expression level and its gene polymorphisms are associated with female-dependent autoimmune disease risk. Altogether, various studies have demonstrated that increased female susceptibility to autoimmune diseases is in part a consequence of hormone-driven reduced thymic AIRE expression.
Collapse
Affiliation(s)
- Sonia Berrih-Aknin
- UPMC Sorbonne Universities, Paris, France
- INSERM U974, Paris, France
- AIM, Institute of Myology, Paris, France
| | - Rozen Le Panse
- UPMC Sorbonne Universities, Paris, France
- INSERM U974, Paris, France
- AIM, Institute of Myology, Paris, France
| | - Nadine Dragin
- UPMC Sorbonne Universities, Paris, France
- INSERM U974, Paris, France
- AIM, Institute of Myology, Paris, France
- Inovarion, Paris, France
| |
Collapse
|
35
|
Paiola M, Knigge T, Picchietti S, Duflot A, Guerra L, Pinto PIS, Scapigliati G, Monsinjon T. Oestrogen receptor distribution related to functional thymus anatomy of the European sea bass, Dicentrarchus labrax. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2017; 77:106-120. [PMID: 28756001 DOI: 10.1016/j.dci.2017.07.023] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Revised: 07/24/2017] [Accepted: 07/24/2017] [Indexed: 06/07/2023]
Abstract
In jawed vertebrates, the crosstalk between immune and endocrine system as well as many fundamental mechanisms of T cell development are evolutionary conserved. Oestrogens affect mammalian thymic function and plasticity, but the mechanisms of action and the oestrogen receptors involved remain unclear. To corroborate the oestrogenic regulation of thymic function in teleosts and to identify the implicated oestrogen receptor subtypes, we examined the distribution of nuclear and membrane oestrogen receptors within the thymus of the European Sea bass, Dicentrarchus labrax, in relation to its morpho-functional organisation. Immunohistological analysis specified thymus histology and organisation in teleosts and described, for the first time, Hassall's corpuscle like structures in the medulla of sea bass. All oestrogen receptors were expressed at the transcript and protein level, both in T cells and in stromal cells belonging to specific functional areas. These observations suggest complex regulatory actions of oestrogen on thymic function, notably through the stromal microenvironment, comprising both, genomic and non-genomic pathways that are likely to affect T cell maturation and trafficking processes. Comparison with birds, rodents and humans supports the thymic localization of oestrogen receptors and suggests that oestrogens modulate T cell maturation in all gnathostomes.
Collapse
Affiliation(s)
- Matthieu Paiola
- Normandy University, FR CNRS 3730 SCALE, UMR-I 02 SEBIO, Université Le Havre Normandie, F-76600 Le Havre, France
| | - Thomas Knigge
- Normandy University, FR CNRS 3730 SCALE, UMR-I 02 SEBIO, Université Le Havre Normandie, F-76600 Le Havre, France
| | - Simona Picchietti
- Department for Innovation in Biological, Agro-food and Forest Systems, Tuscia University, 01100 Viterbo, Italy
| | - Aurélie Duflot
- Normandy University, FR CNRS 3730 SCALE, UMR-I 02 SEBIO, Université Le Havre Normandie, F-76600 Le Havre, France
| | - Laura Guerra
- Department for Innovation in Biological, Agro-food and Forest Systems, Tuscia University, 01100 Viterbo, Italy
| | - Patricia I S Pinto
- Laboratory of Comparative Endocrinology and Integrative Biology, CCMAR - Centre of Marine Sciences, University of Algarve, 8005-139 Faro, Portugal
| | - Giuseppe Scapigliati
- Department for Innovation in Biological, Agro-food and Forest Systems, Tuscia University, 01100 Viterbo, Italy
| | - Tiphaine Monsinjon
- Normandy University, FR CNRS 3730 SCALE, UMR-I 02 SEBIO, Université Le Havre Normandie, F-76600 Le Havre, France.
| |
Collapse
|
36
|
Wahlestedt M, Bryder D. The slippery slope of hematopoietic stem cell aging. Exp Hematol 2017; 56:1-6. [PMID: 28943295 DOI: 10.1016/j.exphem.2017.09.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Revised: 09/12/2017] [Accepted: 09/15/2017] [Indexed: 10/18/2022]
Abstract
The late stages of life, in most species including humans, are associated with a decline in the overall maintenance and health of the organism. This applies also to the hematopoietic system, where aging is not only associated with an increased predisposition for hematological malignancies, but also identified as a strong comorbidity factor for other diseases. Research during the last two decades has proposed that alterations at the level of hematopoietic stem cells (HSCs) might be a root cause for the hematological changes observed with age. However, the recent realization that not all HSCs are alike with regard to fundamental stem cell properties such as self-renewal and lineage potential has several implications for HSC aging, including the synchrony and the stability of the aging HSC state. To approach HSC aging from a clonal perspective, we recently took advantage of technical developments in cellular barcoding and combined this with the derivation of induced pluripotent stem cells (iPSCs). This allowed us to selectively approach HSCs functionally affected by age. The finding that such iPSCs were capable of fully regenerating multilineage hematopoiesis upon morula/blastocyst complementation provides compelling evidence that many aspects of HSC aging can be reversed, which indicates that a central mechanism underlying HSC aging is a failure to uphold the epigenomes associated with younger age. Here we discuss these findings in the context of the underlying causes that might influence HSC aging and the requirements and prospects for restoration of the aging HSC epigenome.
Collapse
Affiliation(s)
- Martin Wahlestedt
- Medical Faculty, Institution for Laboratory Medicine, Division of Molecular Hematology, Lund University, Lund, Sweden
| | - David Bryder
- Medical Faculty, Institution for Laboratory Medicine, Division of Molecular Hematology, Lund University, Lund, Sweden; StemTherapy, Lund University, Lund, Sweden.
| |
Collapse
|
37
|
Chaudhry MS, Velardi E, Malard F, van den Brink MRM. Immune Reconstitution after Allogeneic Hematopoietic Stem Cell Transplantation: Time To T Up the Thymus. THE JOURNAL OF IMMUNOLOGY 2017; 198:40-46. [PMID: 27994167 DOI: 10.4049/jimmunol.1601100] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Accepted: 09/01/2016] [Indexed: 01/09/2023]
Abstract
The success of allogeneic hematopoietic stem cell transplantation, a key treatment for many disorders, is intertwined with T cell immune reconstitution. The thymus plays a key role post allogeneic hematopoietic stem cell transplantation in the generation of a broad but self-tolerant T cell repertoire, but it is exquisitely sensitive to a range of insults during the transplant period, including conditioning regimens, corticosteroids, infections, and graft-versus-host disease. Although endogenous thymic repair is possible it is often suboptimal, and there is a need to develop exogenous strategies to help regenerate the thymus. Therapies currently in clinical trials in the transplant setting include keratinocyte growth factor, cytokines (IL-7 and IL-22), and hormonal modulation including sex steroid inhibition and growth hormone administration. Such regenerative strategies may ultimately enable the thymus to play as prominent a role after transplant as it once did in early childhood, allowing a more complete restoration of the T cell compartment.
Collapse
Affiliation(s)
- Mohammed S Chaudhry
- Immunology Program, Memorial Sloan-Kettering Cancer Center, New York, NY 10065
| | - Enrico Velardi
- Immunology Program, Memorial Sloan-Kettering Cancer Center, New York, NY 10065
| | - Florent Malard
- Immunology Program, Memorial Sloan-Kettering Cancer Center, New York, NY 10065
| | - Marcel R M van den Brink
- Immunology Program, Memorial Sloan-Kettering Cancer Center, New York, NY 10065; .,Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, NY 10065; and.,Department of Immunology and Microbial Pathogenesis, Weill Cornell Medical College, New York, NY 10021
| |
Collapse
|
38
|
Csaba G. The Role of Brain –Pineal –Thymus System in the Determination of Lifespan: The Autoimmune Aging Theory. ACTA ACUST UNITED AC 2017. [DOI: 10.3233/nib-160118] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Affiliation(s)
- G. Csaba
- Department of Genetics, Cell and Immunobiology, Semmelweis University, Budapest, Hungary
| |
Collapse
|
39
|
Effect of gonadotropin-releasing hormone vaccination on T lymphocyte changes in male rats. J Reprod Immunol 2017; 120:1-7. [PMID: 28196761 DOI: 10.1016/j.jri.2017.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Revised: 12/09/2016] [Accepted: 02/06/2017] [Indexed: 11/20/2022]
Abstract
The aim of this study was to detect the effect of immunization against gonadotropin-releasing hormone (GnRH) on cell-meditated immunity. Three-week-old male Sprague-Dawley rats (n=32) were randomly and equally assigned to two groups: 1) GnRH-tandem-ovalbumin immunized group; and 2) the control group (injected with an equivalent Al(OH)3 adjuvant). Blood samples were collected at two-week intervals to assess the level of GnRH-specific antibodies and testosterone. Moreover, blood and thymus samples were also collected to analyze the T lymphocyte subpopulations one and two months after the last booster immunization. T lymphocyte immunity against GnRH was activated during the first month post-immunization as exhibited by increased numbers of CD3+ (P<0.05) and CD4+ (P<0.05)T lymphocytes following testosterone suppression (P<0.01), which was then restored and maintained at appropriate levels in the second month. In contrast, the differentiation of T lymphocytes in the thymus was reduced during the first month after immunization as exhibited by the significant decreased number of CD3+ (P<0.05) cells, followed by the restoration and heightened numbers at later time points for both the number of CD3+ (P<0.05) and CD4+ (P<0.01)T lymphocytes. These results suggest that immunization against GnRH interferes with the number of lymphocytes during the early time points following immunization. The number of T lymphocytes initially decreased in the peripheral blood following immunization, but was replenished by newly exported cells from the thymus which eventually restored the T lymphocytes to normal levels.
Collapse
|
40
|
Saavedra D, Garcia B, Lage A. T Cell Subpopulations in Healthy Elderly and Lung Cancer Patients: Insights from Cuban Studies. Front Immunol 2017; 8:146. [PMID: 28261208 PMCID: PMC5306357 DOI: 10.3389/fimmu.2017.00146] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Accepted: 01/30/2017] [Indexed: 01/10/2023] Open
Abstract
The senescence of the immune system and the risk of cancer increase with aging. Age itself entails changes in the immune system, which are related to a decrease in thymic output of naïve lymphocytes, an accumulation of chronic antigenic load, notably chronic viral infections such as cytomegalovirus (CMV), and replicative senescence of lymphocytes. These changes could eventually contribute to cancer risk and affect the response to cancer treatment. However, several confounding factors make it difficult to draw a picture of causal relationships. Studies in diverse human populations could contribute to clarify these complex relationships. Here, we summarize the current knowledge about the senescence of the T cells, the relationship with CMV infection, cancer, and cancer treatment. We also review the results of a series of studies performed in Cuba whose population is characterized by the unusual combination of long life expectancy and high antigenic load, including high seroprevalence of CMV, typical of tropical countries. Although immunosenescence affects almost all components and functions of the immune response, its most salient feature is a decrease in numbers and proportions of naïve CD8+ T lymphocytes and an accretion of terminally differentiated CD8+ T lymphocytes. These features were confirmed by the Cuban studies, but interestingly a clear gender effect also appeared. Moreover, as aging is a global phenomenon, a fast increase in elderly with malignancies is expected; therefore, the evaluation of patient’s immune status would support the decision of treating them with immunotherapy and predict the efficacy of such treatments, thereby improving benefits for the patients.
Collapse
Affiliation(s)
- Danay Saavedra
- Clinical Immunology Department, Center of Molecular Immunology , Havana , Cuba
| | - Beatriz Garcia
- Clinical Immunology Department, Center of Molecular Immunology , Havana , Cuba
| | - Agustin Lage
- Clinical Immunology Department, Center of Molecular Immunology , Havana , Cuba
| |
Collapse
|
41
|
Holder A, Mella S, Palmer DB, Aspinall R, Catchpole B. An Age-Associated Decline in Thymic Output Differs in Dog Breeds According to Their Longevity. PLoS One 2016; 11:e0165968. [PMID: 27824893 PMCID: PMC5100965 DOI: 10.1371/journal.pone.0165968] [Citation(s) in RCA: 10] [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: 07/25/2016] [Accepted: 10/20/2016] [Indexed: 01/08/2023] Open
Abstract
The age associated decline in immune function is preceded in mammals by a reduction in thymic output. Furthermore, there is increasing evidence of a link between immune competence and lifespan. One approach to determining thymic output is to quantify signal joint T cell receptor excision circles (sj-TRECs), a method which has been developed and used in several mammalian species. Life expectancy and the rate of aging vary in dogs depending upon their breed. In this study, we quantified sj-TRECs in blood samples from dogs of selected breeds to determine whether there was a relationship between longevity and thymic output. In Labrador retrievers, a breed with a median expected lifespan of 11 years, there was an age-associated decline in sj-TREC values, with the greatest decline occurring before 5 years of age, but with sj-TREC still detectable in some geriatric animals, over 13 years of age. In large short-lived breeds (Burnese mountain dogs, Great Danes and Dogue de Bordeaux), the decline in sj-TREC values began earlier in life, compared with small long-lived breeds (Jack Russell terriers and Yorkshire terriers), and the presence of animals with undetectable sj-TRECs occurred at a younger age in the short-lived breeds. The study findings suggest that age-associated changes in canine sj-TRECs are related to breed differences in longevity, and this research highlights the use of dogs as a potential model of immunosenescence.
Collapse
Affiliation(s)
- Angela Holder
- Department of Pathology and Pathogen Biology, Royal Veterinary College, North Mymms, Hertfordshire, United Kingdom
| | - Stephanie Mella
- Department of Pathology and Pathogen Biology, Royal Veterinary College, North Mymms, Hertfordshire, United Kingdom
| | - Donald B. Palmer
- Department of Comparative Biomedical Sciences, Royal Veterinary College, London, United Kingdom
| | - Richard Aspinall
- Health and Wellbeing Academy, Postgraduate Medical Institute, Anglia Ruskin University, Chelmsford, Essex, United Kingdom
| | - Brian Catchpole
- Department of Pathology and Pathogen Biology, Royal Veterinary College, North Mymms, Hertfordshire, United Kingdom
| |
Collapse
|
42
|
Pilipović I, Vujnović I, Arsenović-Ranin N, Dimitrijević M, Kosec D, Stojić-Vukanić Z, Leposavić G. Peripubertal ovariectomy influences thymic adrenergic network plasticity in adult rats. J Neuroimmunol 2016; 297:103-16. [PMID: 27397083 DOI: 10.1016/j.jneuroim.2016.05.017] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Revised: 05/07/2016] [Accepted: 05/22/2016] [Indexed: 11/19/2022]
Abstract
The study investigated the influence of peripubertal ovariectomy on the thymic noradrenaline (NA) concentration, and the thymocyte NA content and β2- and α1-adrenoceptor (AR) expression in adult 2- and 11-month-old rats. In control rats, the thymic NA concentration increased with age. This increase reflected rise in the density of catecholamine (CA)-containing fluorescent nerve fibers and cells and their CA content. Additionally, the average β2- and α1-AR thymocyte surface density changed in the opposite direction with age; the density of β2-AR decreased, whereas that of α1-AR increased. Ovariectomy diminished the thymic NA concentration in 2-month-old rats. This reflected the decrease in the density of fluorescent nerve fibers, and CA content in fluorescent nerve fibers and non-lymphoid cells, since the thymocyte NA content was increased in ovariectomized (Ox) rats. Estrogen supplementation prevented the ovariectomy-induced changes. In Ox rats, the density of CA-synthesizing nerve fibers and non-lymphoid cells diminished with age. To the contrary, NA content in thymocytes increased with age, but it did not exceed that in 11-month-old controls. Additionally, ovariectomy diminished the average thymocyte surface density of β2-ARs, but it increased that of α1-ARs in 2-month-old-rats (due to estrogen, and estrogen and progesterone deficiency, respectively). These changes, despite of the rise in circulating estrogen level post-ovariectomy, remained stable with age. This most likely reflected a decreased sensitivity to estrogen action, as a consequence of the hormone misprinting in peripubertal age. The analysis of thymocyte proliferation in culture suggested that age- and ovariectomy-induced alterations in thymocyte NA synthesis and AR expression altered NA autocrine/paracrine action on thymocytes. In conclusion, the study indicates that the ovarian hormone deficiency in peripubertal age affects ovarian steroid-dependent remodeling of thymic adrenergic regulatory network in adult rats.
Collapse
Affiliation(s)
- Ivan Pilipović
- Immunology Research Centre "Branislav Janković", Institute of Virology, Vaccines and Sera "Torlak", 458 Vojvode Stepe, 11221 Belgrade, Serbia
| | - Ivana Vujnović
- Immunology Research Centre "Branislav Janković", Institute of Virology, Vaccines and Sera "Torlak", 458 Vojvode Stepe, 11221 Belgrade, Serbia
| | - Nevena Arsenović-Ranin
- Department of Microbiology and Immunology, Faculty of Pharmacy, University of Belgrade, 450 Vojvode Stepe, 11221 Belgrade, Serbia
| | - Mirjana Dimitrijević
- Department of Immunology, Institute for Biological Research "Siniša Stanković", University of Belgrade, Bulevar Despota Stefana 142, 11060 Belgrade, Serbia
| | - Duško Kosec
- Immunology Research Centre "Branislav Janković", Institute of Virology, Vaccines and Sera "Torlak", 458 Vojvode Stepe, 11221 Belgrade, Serbia
| | - Zorica Stojić-Vukanić
- Department of Microbiology and Immunology, Faculty of Pharmacy, University of Belgrade, 450 Vojvode Stepe, 11221 Belgrade, Serbia
| | - Gordana Leposavić
- Department of Physiology, Faculty of Pharmacy, University of Belgrade, 450 Vojvode Stepe, 11221 Belgrade, Serbia.
| |
Collapse
|
43
|
Abstract
As the primary site of T-cell development, the thymus plays a key role in the generation of a strong yet self-tolerant adaptive immune response, essential in the face of the potential threat from pathogens or neoplasia. As the importance of the role of the thymus has grown, so too has the understanding that it is extremely sensitive to both acute and chronic injury. The thymus undergoes rapid degeneration following a range of toxic insults, and also involutes as part of the aging process, albeit at a faster rate than many other tissues. The thymus is, however, capable of regenerating, restoring its function to a degree. Potential mechanisms for this endogenous thymic regeneration include keratinocyte growth factor (KGF) signaling, and a more recently described pathway in which innate lymphoid cells produce interleukin-22 (IL-22) in response to loss of double positive thymocytes and upregulation of IL-23 by dendritic cells. Endogenous repair is unable to fully restore the thymus, particularly in the aged population, and this paves the way toward the need for exogenous strategies to help regenerate or even replace thymic function. Therapies currently in clinical trials include KGF, use of the cytokines IL-7 and IL-22, and hormonal modulation including growth hormone administration and sex steroid inhibition. Further novel strategies are emerging in the preclinical setting, including the use of precursor T cells and thymus bioengineering. The use of such strategies offers hope that for many patients, the next regeneration of their thymus is a step closer.
Collapse
Affiliation(s)
- Mohammed S Chaudhry
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Enrico Velardi
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jarrod A Dudakov
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Program in Immunology, Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Marcel R M van den Brink
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Immunology and Microbial Pathogenesis, Weill Cornell Medical College, New York, NY, USA
| |
Collapse
|
44
|
Dragin N, Bismuth J, Cizeron-Clairac G, Biferi MG, Berthault C, Serraf A, Nottin R, Klatzmann D, Cumano A, Barkats M, Le Panse R, Berrih-Aknin S. Estrogen-mediated downregulation of AIRE influences sexual dimorphism in autoimmune diseases. J Clin Invest 2016; 126:1525-37. [PMID: 26999605 PMCID: PMC4811157 DOI: 10.1172/jci81894] [Citation(s) in RCA: 138] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Accepted: 01/21/2016] [Indexed: 01/01/2023] Open
Abstract
Autoimmune diseases affect 5% to 8% of the population, and females are more susceptible to these diseases than males. Here, we analyzed human thymic transcriptome and revealed sex-associated differences in the expression of tissue-specific antigens that are controlled by the autoimmune regulator (AIRE), a key factor in central tolerance. We hypothesized that the level of AIRE is linked to sexual dimorphism susceptibility to autoimmune diseases. In human and mouse thymus, females expressed less AIRE (mRNA and protein) than males after puberty. These results were confirmed in purified murine thymic epithelial cells (TECs). We also demonstrated that AIRE expression is related to sexual hormones, as male castration decreased AIRE thymic expression and estrogen receptor α-deficient mice did not show a sex disparity for AIRE expression. Moreover, estrogen treatment resulted in downregulation of AIRE expression in cultured human TECs, human thymic tissue grafted to immunodeficient mice, and murine fetal thymus organ cultures. AIRE levels in human thymus grafted in immunodeficient mice depended upon the sex of the recipient. Estrogen also upregulated the number of methylated CpG sites in the AIRE promoter. Together, our results indicate that in females, estrogen induces epigenetic changes in the AIRE gene, leading to reduced AIRE expression under a threshold that increases female susceptibility to autoimmune diseases.
Collapse
Affiliation(s)
- Nadine Dragin
- Sorbonne Universités, UPMC University of Paris 06, Paris, France
- INSERM U974, Paris, France
- CNRS FRE 3617, Paris, France
- AIM, Institute of Myology, Paris, France
| | - Jacky Bismuth
- Sorbonne Universités, UPMC University of Paris 06, Paris, France
- INSERM U974, Paris, France
- CNRS FRE 3617, Paris, France
- AIM, Institute of Myology, Paris, France
| | | | - Maria Grazia Biferi
- Sorbonne Universités, UPMC University of Paris 06, Paris, France
- INSERM U974, Paris, France
- CNRS FRE 3617, Paris, France
- AIM, Institute of Myology, Paris, France
| | - Claire Berthault
- INSERM U668, Unit for Lymphopoiesis, Immunology Department, Pasteur Institute, Paris, France
- Université Paris Diderot, Sorbonne Paris Cité, Cellule Pasteur, Paris, France
| | - Alain Serraf
- Hôpital Marie Lannelongue, Le Plessis–Robinson, France
| | - Rémi Nottin
- Hôpital Marie Lannelongue, Le Plessis–Robinson, France
| | - David Klatzmann
- Assistance Publique – Hôpitaux de Paris (AP-HP), Pitié-Salpêtrière Hospital, Biotherapy, Paris, France
| | - Ana Cumano
- INSERM U668, Unit for Lymphopoiesis, Immunology Department, Pasteur Institute, Paris, France
| | - Martine Barkats
- Sorbonne Universités, UPMC University of Paris 06, Paris, France
- INSERM U974, Paris, France
- CNRS FRE 3617, Paris, France
- AIM, Institute of Myology, Paris, France
| | - Rozen Le Panse
- Sorbonne Universités, UPMC University of Paris 06, Paris, France
- INSERM U974, Paris, France
- CNRS FRE 3617, Paris, France
- AIM, Institute of Myology, Paris, France
| | - Sonia Berrih-Aknin
- Sorbonne Universités, UPMC University of Paris 06, Paris, France
- INSERM U974, Paris, France
- CNRS FRE 3617, Paris, France
- AIM, Institute of Myology, Paris, France
| |
Collapse
|
45
|
Lucarelli B, Merli P, Bertaina V, Locatelli F. Strategies to accelerate immune recovery after allogeneic hematopoietic stem cell transplantation. Expert Rev Clin Immunol 2015; 12:343-58. [PMID: 26588325 DOI: 10.1586/1744666x.2016.1123091] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The interplay existing between immune reconstitution and patient outcome has been extensively demonstrated in allogeneic hematopoietic stem cell transplantation. One of the leading causes of infection-related mortality is the slow recovery of T-cell immunity due to the conditioning regimen and/or age-related thymus damage, poor naïve T-cell output, and restricted T-cell receptor (TCR) repertoires. With the aim of improving posttransplantation immune reconstitution, several immunotherapy approaches have been explored. Donor leukocyte infusions are widely used to accelerate immune recovery, but they carry the risk of provoking graft-versus-host disease. This review will focus on sophisticated strategies of thymus function-recovery, adoptive infusion of donor-derived, allodepleted T cells, T-cell lines/clones specific for life-threatening pathogens, regulatory T cells, and of T cells transduced with suicide genes.
Collapse
Affiliation(s)
- Barbarella Lucarelli
- a Department of Pediatric Hematology-Oncology , IRCCS, Bambino Gesù Children's Hospital , Rome , Italy
| | - Pietro Merli
- a Department of Pediatric Hematology-Oncology , IRCCS, Bambino Gesù Children's Hospital , Rome , Italy
| | - Valentina Bertaina
- a Department of Pediatric Hematology-Oncology , IRCCS, Bambino Gesù Children's Hospital , Rome , Italy
| | - Franco Locatelli
- a Department of Pediatric Hematology-Oncology , IRCCS, Bambino Gesù Children's Hospital , Rome , Italy.,b Department of Pediatrics , University of Pavia , Pavia , Italy
| |
Collapse
|
46
|
Brown SG, Shirachi S, Zandbergen D. HEALTH SELECTION THEORY: AN EXPLANATION FOR THE PARADOX BETWEEN PERCEIVED MALE WELL-BEING AND MORTALITY. QUARTERLY REVIEW OF BIOLOGY 2015; 90:3-21. [PMID: 26434163 DOI: 10.1086/679761] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Paradoxically, men report better health and quality of life than women, but men experience higher mortality rates than women at most ages. One conclusion from these findings is that men have been selected to disregard signs of ill health, or even to deceive themselves about their health, to their detriment because presenting themselves as healthy has fitness benefits. We hypothesize that men have been sexually selected to present themselves to women as healthy but that the cost of not attending to their minor health problems results in earlier mortality than women. We present a review of the human and primate literature that supports health selection theory, the hypothesis that females have preferentially selected males who present themselves as healthy.
Collapse
|
47
|
Okoye AA, Rohankhedkar M, Konfe AL, Abana CO, Reyes MD, Clock JA, Duell DM, Sylwester AW, Sammader P, Legasse AW, Park BS, Axthelm MK, Nikolich-Žugich J, Picker LJ. Effect of IL-7 Therapy on Naive and Memory T Cell Homeostasis in Aged Rhesus Macaques. THE JOURNAL OF IMMUNOLOGY 2015; 195:4292-305. [PMID: 26416281 DOI: 10.4049/jimmunol.1500609] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Accepted: 08/28/2015] [Indexed: 12/16/2022]
Abstract
Aging is associated with gradual deterioration of adaptive immune function, a hallmark of which is the profound loss of naive T cells (TN) associated with decline in thymic output and export of new cells into the peripheral T cell pool. Because the lymphotropic cytokine IL-7 plays crucial roles in both development of TN in the thymus and TN homeostasis in the periphery, we sought to determine the extent to which therapeutic administration of IL-7 could reverse TN deficiency in aging rhesus macaques (RM), either by enhancement of the demonstrably reduced thymopoiesis or by peripheral TN expansion. Our results indicate that treatment of both adult (8-15 y) and old (>20 y) RM with recombinant simian IL-7 (rsIL-7) results in only transient increases in peripheral CD4(+) and CD8(+) TN numbers with no long-term benefit, even with repeated therapy. This transient effect was due to peripheral TN expansion and not enhanced thymic function, and appeared to be limited by induction of IL-7 nonresponsiveness. However, rsIL-7 therapy had a more promising effect on the central memory T cell (TCM) population (both CD4(+) and CD8(+)) in adult and old RM, doubling the numbers of these cells in circulation and maintaining this larger population long term. IL-7 therapy did not reduce TCR diversity of the memory T cell compartment, suggesting that rsIL-7-induced expansion was symmetrical. Thus, although rsIL-7 failed to counter age-associated TN loss, the ability of this therapy to expand clonotypically diverse CD4(+) and CD8(+) TCM populations might potentially improve adaptive immune responsiveness in the elderly.
Collapse
Affiliation(s)
- Afam A Okoye
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR 97006; Oregon National Primate Research Center, Beaverton, OR 97006
| | - Mukta Rohankhedkar
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR 97006; Oregon National Primate Research Center, Beaverton, OR 97006
| | - Audrie L Konfe
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR 97006; Oregon National Primate Research Center, Beaverton, OR 97006
| | - Chike O Abana
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR 97006; Oregon National Primate Research Center, Beaverton, OR 97006
| | - Matthew D Reyes
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR 97006; Oregon National Primate Research Center, Beaverton, OR 97006
| | - Joseph A Clock
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR 97006; Oregon National Primate Research Center, Beaverton, OR 97006
| | - Derick M Duell
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR 97006; Oregon National Primate Research Center, Beaverton, OR 97006
| | - Andrew W Sylwester
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR 97006; Oregon National Primate Research Center, Beaverton, OR 97006
| | | | - Alfred W Legasse
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR 97006; Oregon National Primate Research Center, Beaverton, OR 97006
| | - Byung S Park
- Division of Biostatistics, Department of Public Health and Preventive Medicine, Oregon Health & Science University, Portland, OR 97239
| | - Michael K Axthelm
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR 97006; Oregon National Primate Research Center, Beaverton, OR 97006
| | - Janko Nikolich-Žugich
- Department of Immunobiology, University of Arizona College of Medicine, Tucson, AZ 85724; and The Arizona Center on Aging, University of Arizona College of Medicine, Tucson, AZ 85724.
| | - Louis J Picker
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR 97006; Oregon National Primate Research Center, Beaverton, OR 97006;
| |
Collapse
|
48
|
Chapman JC, Chapman FM, Michael SD. The production of alpha/beta and gamma/delta double negative (DN) T-cells and their role in the maintenance of pregnancy. Reprod Biol Endocrinol 2015; 13:73. [PMID: 26164866 PMCID: PMC4499209 DOI: 10.1186/s12958-015-0073-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Accepted: 07/08/2015] [Indexed: 01/19/2023] Open
Abstract
The ability of the thymus gland to convert bone marrow-derived progenitor cells into single positive (SP) T-cells is well known. In this review we present evidence that the thymus, in addition to producing SP T-cells, also has a pathway for the production of double negative (DN) T-cells. The existence of this pathway was noted during our examination of relevant literature to determine the cause of sex steroid-induced thymocyte loss. In conducting this search our objective was to answer the question of whether thymocyte loss is the end product of a typical interaction between the reproductive and immune systems, or evidence that the two systems are incompatible. We can now report that "thymocyte loss" is a normal process that occurs during the production of DN T-cells. The DN T-cell pathway is unique in that it is mediated by thymic mast cells, and becomes functional following puberty. Sex steroids initiate the development of the pathway by binding to an estrogen receptor alpha located in the outer membrane of the mast cells, causing their activation. This results in their uptake of extracellular calcium, and the production and subsequent release of histamine and serotonin. Lymphatic vessels, located in the subcapsular region of the thymus, respond to the two vasodilators by undergoing a substantial and preferential uptake of gamma/delta and alpha/beta DN T- cells. These T- cells exit the thymus via efferent lymphatic vessels and enter the lymphatic system.The DN pathway is responsible for the production of three subsets of gamma/delta DN T-cells and one subset of alpha/beta DN T-cells. In postpubertal animals approximately 35 % of total thymocytes exit the thymus as DN T-cells, regardless of sex. In pregnant females, their levels undergo a dramatic increase. Gamma/delta DN T-cells produce cytokines that are essential for the maintenance of pregnancy.
Collapse
Affiliation(s)
- John C Chapman
- Department of Biological Sciences, Binghamton University, Binghamton, New York, 13902-6000, USA.
| | - Fae M Chapman
- Department of Biological Sciences, Binghamton University, Binghamton, New York, 13902-6000, USA.
| | - Sandra D Michael
- Department of Biological Sciences, Binghamton University, Binghamton, New York, 13902-6000, USA.
| |
Collapse
|
49
|
|
50
|
Leclercq C, Prunier A, Thomas F, Merlot E. Neonatal surgical castration of male pigs reduces thymic growth but has moderate consequences on thymocytes. J Anim Sci 2014; 92:2415-21. [PMID: 24668957 DOI: 10.2527/jas.2013-7202] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Thymus integrates numerous signals from the neuroendocrine-immune system, including sex steroids, glucocorticoids, and catecholamines. Neonatal surgical castration, commonly practiced in pig husbandry, modifies thymic hormonal environment, for example, sex steroids and probably glucocorticoids and catecholamines, which are important modulators of thymic function. This study aimed at investigating, in pubescent male pigs, the consequences of neonatal suppression of testicular hormones on thymic T cell differentiation and hormonal control of thymocyte proliferation. A total of 34 male pigs were allocated to 2 experimental groups: control (CT) intact males and males surgically castrated (SC) at 5 or 6 d of age. At slaughter, thymus was weighed and thymic samples were collected to determine fat content and distribution of thymocyte subsets by identification of CD1, CD4, CD8, and γδ T cell receptor (TCR) cell surface markers and to measure thymocyte proliferation in presence of cortisol, norepinephrine, and sex steroids. Results showed that absolute and relative thymus weights were greater (P < 0.01 and P < 0.01, respectively) whereas thymic fat content was less (P < 0.01) in CT than in SC pigs. Surgical castration did not change the frequency of CD1+ immature thymocytes. The proportion of γδ T cells tended to be greater in CT than in SC pigs (P < 0.1) but the proportions of CD4+, CD8+, and CD4+CD8+ thymocytes were similar in both groups (P > 0.1) indicating that the Tαβ lineage was not influenced by early castration. Proliferation of thymocytes in response to concanavalin A (ConA) was greater in SC than in CT pigs (P < 0.05). Cortisol and norepinephrine decreased the ConA-induced proliferation in CT and SC pigs (P < 0.05). In addition, proliferation of thymocytes was less inhibited by norepinephrine in SC than in CT males (P < 0.05). The greatest concentration of testosterone (25 ng/mL) increased (SC males, P < 0.05) or tended to increase (CT males, P < 0.1) the proliferative responsiveness to ConA but the lowest dose (2.5 ng/mL) and the greatest dose of testosterone combined with estradiol had no significant effect (P > 0.1). Overall, our data show little effect of neonatal castration on thymocyte differentiation as well as of sex hormones on thymocyte proliferation. However, thymic cells seem to be more sensitive to the inhibitory influence of norepinephrine in CT than in CS pigs. The significance of such difference for animal health remains to be explored.
Collapse
Affiliation(s)
- C Leclercq
- INRA, UMR1348 PEGASE, F-35590 Saint-Gilles, France Agrocampus Ouest, UMR1348 PEGASE, F-35000 Rennes, France
| | - A Prunier
- INRA, UMR1348 PEGASE, F-35590 Saint-Gilles, France Agrocampus Ouest, UMR1348 PEGASE, F-35000 Rennes, France
| | - F Thomas
- INRA, UMR1348 PEGASE, F-35590 Saint-Gilles, France Agrocampus Ouest, UMR1348 PEGASE, F-35000 Rennes, France
| | - E Merlot
- INRA, UMR1348 PEGASE, F-35590 Saint-Gilles, France Agrocampus Ouest, UMR1348 PEGASE, F-35000 Rennes, France
| |
Collapse
|