1
|
Gatti DM, Reynolds LA. Thymic eosinophils: What are you doing here? J Leukoc Biol 2025; 117:qiaf001. [PMID: 39776203 DOI: 10.1093/jleuko/qiaf001] [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: 10/07/2024] [Revised: 11/29/2024] [Accepted: 01/06/2025] [Indexed: 01/11/2025] Open
Abstract
The thymus is a primary lymphoid organ where major types of T lymphocytes undergo essential developmental processes. Eosinophils are among the cell types present in microenvironments within the thymus, and perhaps surprisingly, the role of thymic eosinophils, especially during homeostatic conditions, remains unclear. Major physiological events impact thymic organization and function throughout life, including age-related involution, pregnancy, and exposure to chemotherapy or radiation. In this review, we summarize literature that has explored factors that regulate the accumulation, phenotype, and location of thymic eosinophils during homeostatic development and during conditions in which homeostasis is perturbed. Further, we discuss the current theories as to the function of thymic eosinophils and consider how the heterogeneity of thymic eosinophil populations may reflect a temporal, spatial, and situational multifunctionality of thymic eosinophils.
Collapse
Affiliation(s)
- Dominique M Gatti
- Department of Biochemistry and Microbiology, PO Box 1700 STN CSC, Faculty of Science, University of Victoria, Victoria, British Columbia, Canada, V8W 2Y2
| | - Lisa A Reynolds
- Department of Biochemistry and Microbiology, PO Box 1700 STN CSC, Faculty of Science, University of Victoria, Victoria, British Columbia, Canada, V8W 2Y2
| |
Collapse
|
2
|
Yang Z, Zhang Y, Cao Z, Li Z, Zhang L, Yang L. Expression of Estrogen Receptors in Main Immune Organs in Sheep During Early Pregnancy. Int J Mol Sci 2025; 26:3528. [PMID: 40331991 PMCID: PMC12027075 DOI: 10.3390/ijms26083528] [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: 02/27/2025] [Revised: 04/03/2025] [Accepted: 04/05/2025] [Indexed: 05/08/2025] Open
Abstract
Estrogen exerts its action via estrogen receptors (ERs), including ERα and ERβ, and has effects on immunomodulation during pregnancy. It is known that there are changes in the function of the maternal immune organs during pregnancy. However, it is not clear if early pregnancy has effects on the expression of ERα and ERβ in the ovine maternal thymus, lymph nodes, spleen, and liver. In this study, these maternal immune organs were harvested at day 16 of the estrous cycle and at days 13, 16, and 25 of pregnancy (n = 6 for each group) after the ewes were euthanized. The mRNA and protein expression of ERα and ERβ were analyzed using real-time PCR and Western blot and immunohistochemical analyses. The results reveal that the mRNA and protein expression of both ERα and ERβ were upregulated in the maternal spleen and the expression of ERα and ERβ in the thymus, lymph nodes, and liver was modulated during early pregnancy. In conclusion, early pregnancy modulates the expression of ERα and ERβ in the maternal thymus, lymph nodes, spleen, and liver in a tissue-specific manner, which is related to the regulation of maternal immune function during early pregnancy in ewes.
Collapse
Affiliation(s)
| | | | | | | | | | - Ling Yang
- School of Life Sciences and Food Engineering, Hebei University of Engineering, Handan 056038, China; (Z.Y.); (Y.Z.); (Z.C.); (Z.L.); (L.Z.)
| |
Collapse
|
3
|
Zhou M, Shu Y, Gao J. Thymus Degeneration in Women and the Influence of Female Sexual Hormones on Thymic Epithelial Cells. Int J Mol Sci 2025; 26:3014. [PMID: 40243626 PMCID: PMC11988661 DOI: 10.3390/ijms26073014] [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: 02/20/2025] [Revised: 03/19/2025] [Accepted: 03/24/2025] [Indexed: 04/18/2025] Open
Abstract
The thymus is a central immune organ for T cell development and plays an extremely important role in immune and aging. The unique physiological processes that occur in women, such as the menstrual cycle, pregnancy, and menopause, contribute to sexual dimorphism in thymic immunity. Thymic epithelial cells (TECs) are key stromal cells that affect thymus development and degeneration. Interestingly, TECs in women have stronger proliferation potentiality and ability for output of T cells than those in men. In comparison to men, women exhibit higher susceptibility to autoimmune disease, which can be attributed to lower AIRE expression in the female thymus, which is influenced by fluctuating hormone levels. In this review, we summarize the principles of female thymus regulation by hormones, particularly the influence of female sex hormones in the development and function of TECs, as well as the underlying mechanisms, with the aim of providing new ideas and strategies to inhibit or slow down female thymus degeneration.
Collapse
Affiliation(s)
| | | | - Jianli Gao
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China; (M.Z.); (Y.S.)
| |
Collapse
|
4
|
Ruiz Pérez M, Vandenabeele P, Tougaard P. The thymus road to a T cell: migration, selection, and atrophy. Front Immunol 2024; 15:1443910. [PMID: 39257583 PMCID: PMC11384998 DOI: 10.3389/fimmu.2024.1443910] [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: 06/04/2024] [Accepted: 08/08/2024] [Indexed: 09/12/2024] Open
Abstract
The thymus plays a pivotal role in generating a highly-diverse repertoire of T lymphocytes while preventing autoimmunity. Thymus seeding progenitors (TSPs) are a heterogeneous group of multipotent progenitors that migrate to the thymus via CCR7 and CCR9 receptors. While NOTCH guides thymus progenitors toward T cell fate, the absence or disruption of NOTCH signaling renders the thymus microenvironment permissive to other cell fates. Following T cell commitment, developing T cells undergo multiple selection checkpoints by engaging with the extracellular matrix, and interacting with thymic epithelial cells (TECs) and other immune subsets across the different compartments of the thymus. The different selection checkpoints assess the T cell receptor (TCR) performance, with failure resulting in either repurposing (agonist selection), or cell death. Additionally, environmental cues such as inflammation and endocrine signaling induce acute thymus atrophy, contributing to the demise of most developing T cells during thymic selection. We discuss the occurrence of acute thymus atrophy in response to systemic inflammation. The thymus demonstrates high plasticity, shaping inflammation by abrogating T cell development and undergoing profound structural changes, and facilitating regeneration and restoration of T cell development once inflammation is resolved. Despite the challenges, thymic selection ensures a highly diverse T cell repertoire capable of discerning between self and non-self antigens, ultimately egressing to secondary lymphoid organs where they complete their maturation and exert their functions.
Collapse
Affiliation(s)
- Mario Ruiz Pérez
- Molecular Signaling and Cell Death Unit, VIB-UGent, Center for Inflammation Research, Flanders Institute for Biotechnology, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Peter Vandenabeele
- Molecular Signaling and Cell Death Unit, VIB-UGent, Center for Inflammation Research, Flanders Institute for Biotechnology, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Peter Tougaard
- Molecular Signaling and Cell Death Unit, VIB-UGent, Center for Inflammation Research, Flanders Institute for Biotechnology, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
- Laboratory of Immunoregulation and Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent, Belgium
| |
Collapse
|
5
|
Lagou MK, Argyris DG, Vodopyanov S, Gunther-Cummins L, Hardas A, Poutahidis T, Panorias C, DesMarais S, Entenberg C, Carpenter RS, Guzik H, Nishku X, Churaman J, Maryanovich M, DesMarais V, Macaluso FP, Karagiannis GS. Morphometric Analysis of the Thymic Epithelial Cell (TEC) Network Using Integrated and Orthogonal Digital Pathology Approaches. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.11.584509. [PMID: 38559037 PMCID: PMC10979902 DOI: 10.1101/2024.03.11.584509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
The thymus, a central primary lymphoid organ of the immune system, plays a key role in T cell development. Surprisingly, the thymus is quite neglected with regards to standardized pathology approaches and practices for assessing structure and function. Most studies use multispectral flow cytometry to define the dynamic composition of the thymus at the cell population level, but they are limited by lack of contextual insight. This knowledge gap hinders our understanding of various thymic conditions and pathologies, particularly how they affect thymic architecture, and subsequently, immune competence. Here, we introduce a digital pathology pipeline to address these challenges. Our approach can be coupled to analytical algorithms and utilizes rationalized morphometric assessments of thymic tissue, ranging from tissue-wide down to microanatomical and ultrastructural levels. This pipeline enables the quantitative assessment of putative changes and adaptations of thymic structure to stimuli, offering valuable insights into the pathophysiology of thymic disorders. This versatile pipeline can be applied to a wide range of conditions that may directly or indirectly affect thymic structure, ranging from various cytotoxic stimuli inducing acute thymic involution to autoimmune diseases, such as myasthenia gravis. Here, we demonstrate applicability of the method in a mouse model of age-dependent thymic involution, both by confirming established knowledge, and by providing novel insights on intrathymic remodeling in the aged thymus. Our orthogonal pipeline, with its high versatility and depth of analysis, promises to be a valuable and practical toolset for both basic and translational immunology laboratories investigating thymic function and disease.
Collapse
Affiliation(s)
- Maria K Lagou
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, USA
- Tumor Microenvironment and Metastasis Program, Montefiore-Einstein Comprehensive Cancer Center, Bronx, NY, USA
| | - Dimitrios G Argyris
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, USA
- Tumor Microenvironment and Metastasis Program, Montefiore-Einstein Comprehensive Cancer Center, Bronx, NY, USA
- Integrated Imaging Program for Cancer Research, Montefiore-Einstein Comprehensive Cancer Center, Bronx, NY, USA
| | - Stepan Vodopyanov
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, USA
- Tumor Microenvironment and Metastasis Program, Montefiore-Einstein Comprehensive Cancer Center, Bronx, NY, USA
| | - Leslie Gunther-Cummins
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY, USA
- Analytical Imaging Facility, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY, USA
- Montefiore-Einstein Comprehensive Cancer, Center, Bronx, NY, USA
| | - Alexandros Hardas
- Department of Pathobiology and Population Sciences, The Royal Veterinary College, North Mymms, Hatfield, United Kingdom
| | - Theofilos Poutahidis
- Laboratory of Pathology, School of Veterinary Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Christos Panorias
- Division of Statistics and Operational Research, Department of Mathematics, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Sophia DesMarais
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Conner Entenberg
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Randall S Carpenter
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY, USA
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Hillary Guzik
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY, USA
- Analytical Imaging Facility, Albert Einstein College of Medicine, Bronx, NY, USA
- Montefiore-Einstein Comprehensive Cancer, Center, Bronx, NY, USA
| | - Xheni Nishku
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY, USA
- Analytical Imaging Facility, Albert Einstein College of Medicine, Bronx, NY, USA
- Montefiore-Einstein Comprehensive Cancer, Center, Bronx, NY, USA
| | - Joseph Churaman
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY, USA
- Analytical Imaging Facility, Albert Einstein College of Medicine, Bronx, NY, USA
- Montefiore-Einstein Comprehensive Cancer, Center, Bronx, NY, USA
| | - Maria Maryanovich
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY, USA
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
- Cancer Dormancy and Tumor Microenvironment Institute, Montefiore-Einstein Comprehensive Cancer, Center, Bronx, NY, USA
| | - Vera DesMarais
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY, USA
- Analytical Imaging Facility, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY, USA
- Montefiore-Einstein Comprehensive Cancer, Center, Bronx, NY, USA
| | - Frank P Macaluso
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY, USA
- Analytical Imaging Facility, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY, USA
- Montefiore-Einstein Comprehensive Cancer, Center, Bronx, NY, USA
| | - George S Karagiannis
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, USA
- Tumor Microenvironment and Metastasis Program, Montefiore-Einstein Comprehensive Cancer Center, Bronx, NY, USA
- Integrated Imaging Program for Cancer Research, Montefiore-Einstein Comprehensive Cancer Center, Bronx, NY, USA
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY, USA
- Cancer Dormancy and Tumor Microenvironment Institute, Montefiore-Einstein Comprehensive Cancer, Center, Bronx, NY, USA
| |
Collapse
|
6
|
Nevo S, Frenkel N, Kadouri N, Gome T, Rosenthal N, Givony T, Avin A, Peligero Cruz C, Kedmi M, Lindzen M, Ben Dor S, Damari G, Porat Z, Haffner-Krausz R, Keren-Shaul H, Yarden Y, Munitz A, Leshkowitz D, Goldfarb Y, Abramson J. Tuft cells and fibroblasts promote thymus regeneration through ILC2-mediated type 2 immune response. Sci Immunol 2024; 9:eabq6930. [PMID: 38215193 DOI: 10.1126/sciimmunol.abq6930] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 11/15/2023] [Indexed: 01/14/2024]
Abstract
The thymus is a primary lymphoid organ that is essential for the establishment of adaptive immunity through generation of immunocompetent T cells. In response to various stress signals, the thymus undergoes acute but reversible involution. However, the mechanisms governing its recovery are incompletely understood. Here, we used a dexamethasone-induced acute thymic involution mouse model to investigate how thymic hematopoietic cells (excluding T cells) contribute to thymic regeneration. scRNA-seq analysis revealed marked transcriptional and cellular changes in various thymic populations and highlighted thymus-resident innate lymphoid cells type 2 (ILC2) as a key cell type involved in the response to damage. We identified that ILC2 are activated by the alarmins IL-25 and IL-33 produced in response to tissue damage by thymic tuft cells and fibroblasts, respectively. Moreover, using mouse models deficient in either tuft cells and/or IL-33, we found that these alarmins are required for effective thymus regeneration after dexamethasone-induced damage. We also demonstrate that upon their damage-dependent activation, thymic ILC2 produce several effector molecules linked to tissue regeneration, such as amphiregulin and IL-13, which in turn promote thymic epithelial cell differentiation. Collectively, our study elucidates a previously undescribed role for thymic tuft cells and fibroblasts in thymus regeneration through activation of the type 2 immune response.
Collapse
Affiliation(s)
- Shir Nevo
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Noga Frenkel
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Noam Kadouri
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Tom Gome
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel
- Department of Computer Science and Applied Mathematics, Weizmann Institute of Science, Rehovot, Israel
| | - Noa Rosenthal
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Tal Givony
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Ayelet Avin
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Cristina Peligero Cruz
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel
- IrsiCaixa AIDS Research Institute, Hospital Germans Trias i Pujol, Institute for Health Science Research Germans Trias i Pujol (IGTP), Badalona, Spain
| | - Merav Kedmi
- Genomics Unit, Life Science Core Facility, Weizmann Institute of Science, Rehovot, Israel
| | - Moshit Lindzen
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Shifra Ben Dor
- Bioinformatics Unit, Life Science Core Facility, Weizmann Institute of Science, Rehovot, Israel
| | - Golda Damari
- Department of Veterinary Resources, Weizmann Institute of Science, Rehovot, Israel
| | - Ziv Porat
- Flow Cytometry Unit, Life Science Core Facility, Weizmann Institute of Science, Rehovot, Israel
| | | | - Hadas Keren-Shaul
- Genomics Unit, Life Science Core Facility, Weizmann Institute of Science, Rehovot, Israel
| | - Yosef Yarden
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Ariel Munitz
- Department of Microbiology and Clinical Immunology, Sackler School of Medicine, Tel-Aviv University, Tel Aviv, Israel
| | - Dena Leshkowitz
- Bioinformatics Unit, Life Science Core Facility, Weizmann Institute of Science, Rehovot, Israel
| | - Yael Goldfarb
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Jakub Abramson
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
7
|
Fujimori S, Ohigashi I. The role of thymic epithelium in thymus development and age-related thymic involution. THE JOURNAL OF MEDICAL INVESTIGATION 2024; 71:29-39. [PMID: 38735722 DOI: 10.2152/jmi.71.29] [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] [Indexed: 05/14/2024]
Abstract
The establishment of an adaptive immune system is critical for protecting our bodies from neoplastic cancers and invading pathogens such as viruses and bacteria. As a primary lymphoid organ, the thymus generates lymphoid T cells that play a major role in the adaptive immune system. T cell generation in the thymus is controlled by interactions between thymocytes and other thymic cells, primarily thymic epithelial cells. Thus, the normal development and function of thymic epithelial cells are important for the generation of immunocompetent and self-tolerant T cells. On the other hand, the degeneration of the thymic epithelium due to thymic aging causes thymic involution, which is associated with the decline of adaptive immune function. Herein we summarize basic and current knowledge of the development and function of thymic epithelial cells and the mechanism of thymic involution. J. Med. Invest. 71 : 29-39, February, 2024.
Collapse
Affiliation(s)
- Sayumi Fujimori
- Division of Experimental Immunology, Institute of Advanced Medical Sciences, Tokushima University, Tokushima, Japan
| | - Izumi Ohigashi
- Division of Experimental Immunology, Institute of Advanced Medical Sciences, Tokushima University, Tokushima, Japan
| |
Collapse
|
8
|
Pereira KHNP, Fuchs KDM, Mendonça JC, Xavier GM, Knupp FC, Lourenço MLG. Topics on maternal, fetal and neonatal immunology of dogs and cats. Vet Immunol Immunopathol 2023; 266:110678. [PMID: 38016336 DOI: 10.1016/j.vetimm.2023.110678] [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: 09/19/2023] [Revised: 11/05/2023] [Accepted: 11/07/2023] [Indexed: 11/30/2023]
Abstract
Birth and the first few weeks of age are critical periods of developing the immune system of puppies and kittens and adapting to an environment containing a variety of infectious agents. The survival rate during these periods depends mainly on the newborn's immune capacity to prevent and combat infections. Although most components of innate and adaptive immunity are present at birth, responses are slow and immature compared to adults. Due to immunological immaturity and the endotheliochorial placental structure, circulating concentrations of immunoglobulins in dogs and cats at birth are quite low. Thus, newborns need a prompt and immediate immune response, which is essentially provided by defense cells and maternal antibodies via colostrum. Failure to ingest colostrum is correlated with high mortality rates in the neonatal period. Concurrently, factors related to pregnant, such as pregnancy physiological immunosuppression and nutritional and health states, can directly influence newborn immunity and health. Therefore, understanding the maternal and neonatal immunological aspects, importance of colostrum, risk factors for failure to transfer passive immunity and colostrum substitute strategies are essential to ensure the survival of the litter.
Collapse
Affiliation(s)
- Keylla Helena Nobre Pacífico Pereira
- Veterinary Neonatology Research Group, Department of Veterinary Clinic, School of Veterinary Medicine and Animal Science, São Paulo State University (UNESP), Botucatu, São Paulo, Brazil
| | - Kárita da Mata Fuchs
- Veterinary Neonatology Research Group, Department of Veterinary Clinic, School of Veterinary Medicine and Animal Science, São Paulo State University (UNESP), Botucatu, São Paulo, Brazil
| | - Júlia Cosenza Mendonça
- Veterinary Neonatology Research Group, Department of Veterinary Clinic, School of Veterinary Medicine and Animal Science, São Paulo State University (UNESP), Botucatu, São Paulo, Brazil
| | - Gleice Mendes Xavier
- Veterinary Neonatology Research Group, Department of Veterinary Clinic, School of Veterinary Medicine and Animal Science, São Paulo State University (UNESP), Botucatu, São Paulo, Brazil
| | - Fabíola Cardoso Knupp
- Veterinary Neonatology Research Group, Department of Veterinary Clinic, School of Veterinary Medicine and Animal Science, São Paulo State University (UNESP), Botucatu, São Paulo, Brazil
| | - Maria Lucia Gomes Lourenço
- Veterinary Neonatology Research Group, Department of Veterinary Clinic, School of Veterinary Medicine and Animal Science, São Paulo State University (UNESP), Botucatu, São Paulo, Brazil.
| |
Collapse
|
9
|
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
|
10
|
Gomez-Lopez N, Romero R, Escobar MF, Carvajal JA, Echavarria MP, Albornoz LL, Nasner D, Miller D, Gallo DM, Galaz J, Arenas-Hernandez M, Bhatti G, Done B, Zambrano MA, Ramos I, Fernandez PA, Posada L, Chaiworapongsa T, Jung E, Garcia-Flores V, Suksai M, Gotsch F, Bosco M, Than NG, Tarca AL. Pregnancy-specific responses to COVID-19 revealed by high-throughput proteomics of human plasma. COMMUNICATIONS MEDICINE 2023; 3:48. [PMID: 37016066 PMCID: PMC10071476 DOI: 10.1038/s43856-023-00268-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 03/03/2023] [Indexed: 04/06/2023] Open
Abstract
BACKGROUND Pregnant women are at greater risk of adverse outcomes, including mortality, as well as obstetrical complications resulting from COVID-19. However, pregnancy-specific changes that underlie such worsened outcomes remain unclear. METHODS Plasma samples were collected from pregnant women and non-pregnant individuals (male and female) with (n = 72 pregnant, 52 non-pregnant) and without (n = 29 pregnant, 41 non-pregnant) COVID-19. COVID-19 patients were grouped as asymptomatic, mild, moderate, severe, or critically ill according to NIH classifications. Proteomic profiling of 7,288 analytes corresponding to 6,596 unique protein targets was performed using the SOMAmer platform. RESULTS Herein, we profile the plasma proteome of pregnant and non-pregnant COVID-19 patients and controls and show alterations that display a dose-response relationship with disease severity; yet, such proteomic perturbations are dampened during pregnancy. In both pregnant and non-pregnant state, the proteome response induced by COVID-19 shows enrichment of mediators implicated in cytokine storm, endothelial dysfunction, and angiogenesis. Shared and pregnancy-specific proteomic changes are identified: pregnant women display a tailored response that may protect the conceptus from heightened inflammation, while non-pregnant individuals display a stronger response to repel infection. Furthermore, the plasma proteome can accurately identify COVID-19 patients, even when asymptomatic or with mild symptoms. CONCLUSION This study represents the most comprehensive characterization of the plasma proteome of pregnant and non-pregnant COVID-19 patients. Our findings emphasize the distinct immune modulation between the non-pregnant and pregnant states, providing insight into the pathogenesis of COVID-19 as well as a potential explanation for the more severe outcomes observed in pregnant women.
Collapse
Affiliation(s)
- Nardhy Gomez-Lopez
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Detroit, MI, USA.
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA.
- Department of Biochemistry, Microbiology and Immunology, Wayne State University School of Medicine, Detroit, MI, USA.
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI, USA.
| | - Roberto Romero
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Detroit, MI, USA.
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI, USA.
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI, USA.
- Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, MI, USA.
- Detroit Medical Center, Detroit, MI, USA.
| | - María Fernanda Escobar
- Departamento de Ginecología y Obstetricia, Fundación Valle del Lili, Cali, Colombia
- Departamento de Ginecología y Obstetricia, Facultad de Ciencias de la Salud, Universidad Icesi, Cali, Colombia
| | - Javier Andres Carvajal
- Departamento de Ginecología y Obstetricia, Fundación Valle del Lili, Cali, Colombia
- Departamento de Ginecología y Obstetricia, Facultad de Ciencias de la Salud, Universidad Icesi, Cali, Colombia
| | - Maria Paula Echavarria
- Departamento de Ginecología y Obstetricia, Fundación Valle del Lili, Cali, Colombia
- Departamento de Ginecología y Obstetricia, Facultad de Ciencias de la Salud, Universidad Icesi, Cali, Colombia
| | - Ludwig L Albornoz
- Departamento de Laboratorio Clínico y Patología, Fundación Valle del Lili, Cali, Colombia
- Facultad de Ciencias de la Salud, Universidad Icesi, Cali, Colombia
| | - Daniela Nasner
- Centro de Investigaciones Clínicas, Fundación Valle del Lili, Cali, Colombia
| | - Derek Miller
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Dahiana M Gallo
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Jose Galaz
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
- Division of Obstetrics and Gynecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Marcia Arenas-Hernandez
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Gaurav Bhatti
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Bogdan Done
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Maria Andrea Zambrano
- Departamento de Ginecología y Obstetricia, Facultad de Ciencias de la Salud, Universidad Icesi, Cali, Colombia
| | - Isabella Ramos
- Departamento de Ginecología y Obstetricia, Facultad de Ciencias de la Salud, Universidad Icesi, Cali, Colombia
| | - Paula Andrea Fernandez
- Departamento de Ginecología y Obstetricia, Facultad de Ciencias de la Salud, Universidad Icesi, Cali, Colombia
| | - Leandro Posada
- Departamento de Ginecología y Obstetricia, Facultad de Ciencias de la Salud, Universidad Icesi, Cali, Colombia
| | - Tinnakorn Chaiworapongsa
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Eunjung Jung
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Valeria Garcia-Flores
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Manaphat Suksai
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Francesca Gotsch
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Mariachiara Bosco
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Nandor Gabor Than
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Detroit, MI, USA
- Systems Biology of Reproduction Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary
- Maternity Private Clinic of Obstetrics and Gynecology, Budapest, Hungary
- Department of Obstetrics and Gynecology, Semmelweis University, Budapest, Hungary
- Genesis Theranostix Group, Budapest, Hungary
| | - Adi L Tarca
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Detroit, MI, USA.
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA.
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI, USA.
- Department of Computer Science, Wayne State University College of Engineering, Detroit, MI, USA.
| |
Collapse
|
11
|
Depoërs L, Dumont-Lagacé M, Trinh VQH, Houques C, Côté C, Larouche JD, Brochu S, Perreault C. Klf4 protects thymus integrity during late pregnancy. Front Immunol 2023; 14:1016378. [PMID: 37180153 PMCID: PMC10174329 DOI: 10.3389/fimmu.2023.1016378] [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: 08/10/2022] [Accepted: 04/12/2023] [Indexed: 05/15/2023] Open
Abstract
Pregnancy causes abrupt thymic atrophy. This atrophy is characterized by a severe decrease in the number of all thymocyte subsets and qualitative (but not quantitative) changes in thymic epithelial cells (TECs). Pregnancy-related thymic involution is triggered by progesterone-induced functional changes affecting mainly cortical TECs (cTECs). Remarkably, this severe involution is rapidly corrected following parturition. We postulated that understanding the mechanisms of pregnancy-related thymic changes could provide novel insights into signaling pathways regulating TEC function. When we analyzed genes whose expression in TECs was modified during late pregnancy, we found a strong enrichment in genes bearing KLF4 transcription factor binding motifs. We, therefore, engineered a Psmb11-iCre : Klf4lox/lox mouse model to study the impact of TEC-specific Klf4 deletion in steady-state conditions and during late pregnancy. Under steady-state conditions, Klf4 deletion had a minimal effect on TEC subsets and did not affect thymic architecture. However, pregnancy-induced thymic involution was much more pronounced in pregnant females lacking Klf4 expression in TECs. These mice displayed a substantial ablation of TECs with a more pronounced loss of thymocytes. Transcriptomic and phenotypic analyses of Klf4 -/- TECs revealed that Klf4 maintains cTEC numbers by supporting cell survival and preventing epithelial-to-mesenchymal plasticity during late pregnancy. We conclude that Klf4 is essential for preserving TEC's integrity and mitigating thymic involution during late pregnancy.
Collapse
Affiliation(s)
- Lucyle Depoërs
- Department of Medicine, Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC, Canada
| | - Maude Dumont-Lagacé
- ExCellThera, Inc., Montréal, QC, Canada
- Piercing Star Technologies, Rabat, Morocco
| | - Vincent Quoc-Huy Trinh
- Department of Medicine, Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC, Canada
- Department of Pathology and Cellular Biology, Institute for Research in Immunology and Cancer, and Centre de recherche du Centre hospitalier de l’Université de Montréal, Université de Montréal, Montréal, QC, Canada
| | - Chloé Houques
- Institut de Génétique Moléculaire de Montpellier, Université de Montpellier, Montpellier, France
| | - Caroline Côté
- Department of Medicine, Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC, Canada
| | - Jean-David Larouche
- Department of Medicine, Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC, Canada
| | - Sylvie Brochu
- Department of Medicine, Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC, Canada
- *Correspondence: Sylvie Brochu, ; Claude Perreault,
| | - Claude Perreault
- Department of Medicine, Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC, Canada
- *Correspondence: Sylvie Brochu, ; Claude Perreault,
| |
Collapse
|
12
|
Taves MD, Ashwell JD. Effects of sex steroids on thymic epithelium and thymocyte development. Front Immunol 2022; 13:975858. [PMID: 36119041 PMCID: PMC9478935 DOI: 10.3389/fimmu.2022.975858] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 07/18/2022] [Indexed: 11/13/2022] Open
Abstract
Sex steroid hormones have major effects on the thymus. Age-related increases in androgens and estrogens and pregnancy-induced increases in progestins all cause dramatic thymic atrophy. Atrophy can also be induced by treatment with exogenous sex steroids and reversed by ablation of endogenous sex steroids. Although these observations are frequently touted as evidence of steroid lymphotoxicity, they are often driven by steroid signaling in thymic epithelial cells (TEC), which are highly steroid responsive. Here, we outline the effects of sex steroids on the thymus and T cell development. We focus on studies that have examined steroid signaling in vivo, aiming to emphasize the actions of endogenous steroids which, via TEC, have remarkable programming effects on the TCR repertoire. Due to the dramatic effects of steroids on TEC, especially thymic involution, the direct effects of sex steroid signaling in thymocytes are less well understood. We outline studies that could be important in addressing these possibilities, and highlight suggestive findings of sex steroid generation within the thymus itself.
Collapse
Affiliation(s)
- Matthew D. Taves
- Laboratory of Immune Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
- Department of Neurobiology and Behavior, Cornell University, Ithaca, NY, United States
| | - Jonathan D. Ashwell
- Laboratory of Immune Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
13
|
Petroff MG, Nguyen SL, Ahn SH. Fetal‐placental
antigens and the maternal immune system: Reproductive immunology comes of age. Immunol Rev 2022; 308:25-39. [PMID: 35643905 PMCID: PMC9328203 DOI: 10.1111/imr.13090] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Revised: 05/07/2022] [Accepted: 05/11/2022] [Indexed: 12/20/2022]
Abstract
Reproductive physiology and immunology as scientific disciplines each have rich, largely independent histories. The physicians and philosophers of ancient Greece made remarkable observations and inferences to explain regeneration as well as illness and immunity. The scientific enlightenment of the renaissance and the technological advances of the past century have led to the explosion of knowledge that we are experiencing today. Breakthroughs in transplantation, immunology, and reproduction eventually culminated with Medawar’s discovery of acquired immunological tolerance, which helped to explain the transplantation success and failure. Medawar’s musings also keenly pointed out that the fetus apparently breaks these newly discovered rules, and with this, the field of reproductive immunology was launched. As a result of having stemmed from transplantation immunology, scientist still analogizes the fetus to a successful allograft. Although we now know of the fundamental differences between the two, this analogy remains a useful tool to understand how the fetus thrives despite its immunological disparity with the mother. Here, we review the history of reproductive immunology, and how major and minor histocompatibility antigens, blood group antigens, and tissue‐specific “self” antigens from the fetus and transplanted organs parallel and differ.
Collapse
Affiliation(s)
- Margaret G. Petroff
- Department of Pathobiology and Diagnostic Investigation, College of Veterinary Medicine Michigan State University East Lansing Michigan USA
- Departments of Microbiology and Molecular Genetics, College of Veterinary Medicine and College of Human Medicine Michigan State University East Lansing Michigan USA
- Cell and Molecular Biology Program, College of Natural Science Michigan State University East Lansing Michigan USA
| | - Sean L. Nguyen
- Department of Pathobiology and Diagnostic Investigation, College of Veterinary Medicine Michigan State University East Lansing Michigan USA
- Cell and Molecular Biology Program, College of Natural Science Michigan State University East Lansing Michigan USA
| | - Soo Hyun Ahn
- Department of Pathobiology and Diagnostic Investigation, College of Veterinary Medicine Michigan State University East Lansing Michigan USA
| |
Collapse
|
14
|
Ahn SH, Nguyen SL, Kim TH, Jeong JW, Arora R, Lydon JP, Petroff MG. Nuclear Progesterone Receptor Expressed by the Cortical Thymic Epithelial Cells Dictates Thymus Involution in Murine Pregnancy. Front Endocrinol (Lausanne) 2022; 13:846226. [PMID: 35498436 PMCID: PMC9046655 DOI: 10.3389/fendo.2022.846226] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Accepted: 03/14/2022] [Indexed: 12/14/2022] Open
Abstract
Progesterone is a gonadal pro-gestational hormone that is absolutely necessary for the success of pregnancy. Most notable actions of progesterone are observed in the female reproductive organs, the uterus and the ovary. Acting through the nuclear progesterone receptor (PGR), progesterone prepares the endometrium for implantation of the embryo. Interestingly, the maternal thymus also is a known expressor of Pgr; its absence is associated with murine pregnancy complications. However, the localization of its expression and its functional importance were not known. Here, we used a transgenic dual fluorescent reporter mouse model and genetic deletion of Pgr in Foxn1+ thymic epithelial cells (TEC) to demonstrate TEC-specific Pgr expression in pregnancy, especially in the cortex where thymocyte maturation occurs. Using our TEC-specific Pgr deletion mouse model, we demonstrate that TEC-specific Pgr is necessary for pregnancy-induced thymic involution in pregnancy. Our investigation reveals that PGR expression is upregulated in the cortical thymic epithelial cells during pregnancy, and that PGR expression is important for thymic involution during murine pregnancy.
Collapse
Affiliation(s)
- Soo Hyun Ahn
- Department of Pathobiology Diagnostic Investigation, College of Veterinary Medicine, Michigan State University, East Lansing, MI, United States
| | - Sean L. Nguyen
- Institute for Integrative Toxicology, Michigan State University, East Lansing, MI, United States
- Cell and Molecular Biology Program, Michigan State University, East Lansing, MI, United States
| | - Tae Hoon Kim
- Department of Obstetrics, Gynecology & Reproductive Biology, Michigan State University, Grand Rapids, MI, United States
| | - Jae-Wook Jeong
- Department of Obstetrics, Gynecology & Reproductive Biology, Michigan State University, Grand Rapids, MI, United States
| | - Ripla Arora
- Department of Obstetrics, Gynecology, and Reproductive Biology, Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, United States
| | - John P. Lydon
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, United States
| | - Margaret G. Petroff
- Department of Pathobiology Diagnostic Investigation, College of Veterinary Medicine, Michigan State University, East Lansing, MI, United States
- Cell and Molecular Biology Program, Michigan State University, East Lansing, MI, United States
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI, United States
| |
Collapse
|
15
|
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
|
16
|
Barreira-Silva P, Melo-Miranda R, Nobrega C, Roque S, Serre-Miranda C, Borges M, Armada G, de Sá Calçada D, Behar SM, Appelberg R, Correia-Neves M. IFNγ and iNOS-Mediated Alterations in the Bone Marrow and Thymus and Its Impact on Mycobacterium avium-Induced Thymic Atrophy. Front Immunol 2021; 12:696415. [PMID: 34987496 PMCID: PMC8721011 DOI: 10.3389/fimmu.2021.696415] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Accepted: 11/30/2021] [Indexed: 11/13/2022] Open
Abstract
Disseminated infection with the high virulence strain of Mycobacterium avium 25291 leads to progressive thymic atrophy. We previously showed that M. avium-induced thymic atrophy results from increased glucocorticoid levels that synergize with nitric oxide (NO) produced by interferon gamma (IFNγ) activated macrophages. Where and how these mediators act is not understood. We hypothesized that IFNγ and NO promote thymic atrophy through their effects on bone marrow (BM) T cell precursors and T cell differentiation in the thymus. We show that M. avium infection cause a reduction in the percentage and number of common lymphoid progenitors (CLP). Additionally, BM precursors from infected mice show an overall impaired ability to reconstitute thymi of RAGKO mice, in part due to IFNγ. Thymi from infected mice present an IFNγ and NO-driven inflammation. When transplanted under the kidney capsule of uninfected mice, thymi from infected mice are unable to sustain T cell differentiation. Finally, we observed increased thymocyte death via apoptosis after infection, independent of both IFNγ and iNOS; and a decrease on active caspase-3 positive thymocytes, which is not observed in the absence of iNOS expression. Together our data suggests that M. avium-induced thymic atrophy results from a combination of defects mediated by IFNγ and NO, including alterations in the BM T cell precursors, the thymic structure and the thymocyte differentiation.
Collapse
Affiliation(s)
- Palmira Barreira-Silva
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- Life and Health Sciences Research Institute/Biomaterials, Biodegradables and Biomimetics Research Group (ICVS/3B’s), PT Government Associate Laboratory, Braga, Portugal
- *Correspondence: Palmira Barreira-Silva, ; Margarida Correia-Neves,
| | - Rita Melo-Miranda
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- Life and Health Sciences Research Institute/Biomaterials, Biodegradables and Biomimetics Research Group (ICVS/3B’s), PT Government Associate Laboratory, Braga, Portugal
| | - Claudia Nobrega
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- Life and Health Sciences Research Institute/Biomaterials, Biodegradables and Biomimetics Research Group (ICVS/3B’s), PT Government Associate Laboratory, Braga, Portugal
| | - Susana Roque
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- Life and Health Sciences Research Institute/Biomaterials, Biodegradables and Biomimetics Research Group (ICVS/3B’s), PT Government Associate Laboratory, Braga, Portugal
| | - Cláudia Serre-Miranda
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- Life and Health Sciences Research Institute/Biomaterials, Biodegradables and Biomimetics Research Group (ICVS/3B’s), PT Government Associate Laboratory, Braga, Portugal
| | - Margarida Borges
- Research Unit on Applied Molecular Biosciences (UCIBIO)/Rede de Química e Tecnologia (REQUINTE), Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, Porto, Portugal
| | - Gisela Armada
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- Life and Health Sciences Research Institute/Biomaterials, Biodegradables and Biomimetics Research Group (ICVS/3B’s), PT Government Associate Laboratory, Braga, Portugal
| | - Daniela de Sá Calçada
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- Life and Health Sciences Research Institute/Biomaterials, Biodegradables and Biomimetics Research Group (ICVS/3B’s), PT Government Associate Laboratory, Braga, Portugal
| | - Samuel M. Behar
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, MA, United States
| | - Rui Appelberg
- Instituto de Investigação e Inovação em Saúde (i3S), Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- IBMC-Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
- ICBAS-Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Margarida Correia-Neves
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- Life and Health Sciences Research Institute/Biomaterials, Biodegradables and Biomimetics Research Group (ICVS/3B’s), PT Government Associate Laboratory, Braga, Portugal
- Division of Infectious Diseases, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
- *Correspondence: Palmira Barreira-Silva, ; Margarida Correia-Neves,
| |
Collapse
|
17
|
Dos Santos Fagundes I, Brendler EP, Nunes Erthal I, Eder Ribeiro RJ, Caron-Lienert RS, Machado DC, Pinheiro da Costa BE, Poli-de-Figueiredo CE. Total Th1/Th2 cytokines profile from peripheral blood lymphocytes in normal pregnancy and preeclampsia syndrome. Hypertens Pregnancy 2021; 41:15-22. [PMID: 34812111 DOI: 10.1080/10641955.2021.2008424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
To evaluate total Th1/Th2 cytokines in CD3+ cells (immunocompetent T-lymphocytes) and peripheral blood lymphocytes, mostly CD4+ (T helper cells) and CD8+ (T-cytotoxic cells) subpopulations in preeclampsia. Total blood leukocytes and lymphocytes counts, percent cells: CD3+, INF-g+/CD3+, IL-4+/CD3+, and IL-10+/CD3+, CD4+/CD8+ were determined by flow-cytometry. Preeclampsia (n= 26) and normal pregnancy (n= 25) participants were age and gestational age matched. CD4+ lymphocytes count was higher in preeclampsia, compared with normal pregnancy (43.6 ± 5.8 vs 37.6 ± 5.6%; P< 0.001). CD3+ cells Th1/Th2 shift was not detected in preeclampsia, yet may be present in other cell types, such as CD4+ and CD3 - lymphocytes.
Collapse
Affiliation(s)
- Iara Dos Santos Fagundes
- Serviço de Imunologia Hospital de Clínicas de Porto Alegre, Federal University of Rio Grande Do Sul, Porto Alegre, Brazil
| | - Eduardo Pletsch Brendler
- School of Medicine, Pontifical Catholic University of Rio Grande Do Sul, Pucrs, Porto Alegre, Brazil
| | - Isadora Nunes Erthal
- School of Medicine, Pontifical Catholic University of Rio Grande Do Sul, Pucrs, Porto Alegre, Brazil
| | | | | | - Denise Cantarelli Machado
- School of Medicine, Pontifical Catholic University of Rio Grande Do Sul, Pucrs, Porto Alegre, Brazil
| | | | | |
Collapse
|
18
|
Willis SK, Aiello AE, Chatterjea D, Nelson JA, Hibberd P, Harlow BL. Characterizing Differences in Thymic Function in Women With and Without Vulvodynia: A Community-Based Study. J Low Genit Tract Dis 2021; 25:296-302. [PMID: 34542085 PMCID: PMC10155502 DOI: 10.1097/lgt.0000000000000620] [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] [Indexed: 11/27/2022]
Abstract
OBJECTIVE The aim of the study was to evaluate the association between vulvodynia and thymic function. MATERIALS AND METHODS In this case-control study of 200 clinically confirmed cases of vulvodynia and 205 general population controls residing in the Minneapolis/Saint Paul metropolitan area, we used DNA extracted from whole blood to measure levels of signal joint T-cell receptor excision circles (sjTRECs), a measure of thymic output. We used logistic regression to evaluate the association between vulvodynia and thymic function. RESULTS In 405 participants (aged 18-40 years), we observed an association between decreasing thymic function and increasing age. Women with vulvodynia had a steeper decline in sjTREC values across age categories compared with women without vulvodynia. In addition, at younger ages, women with vulvodynia had higher sjTREC values compared with women without vulvodynia. In older women, those with vulvodynia had lower sjTREC than those without vulvodynia. When accounting for recency of vulvar pain onset, women with a shorter time since pain onset had higher thymic function compared with women with a longer time since vulvar pain onset. CONCLUSIONS These findings suggest that at younger ages, women with vulvodynia have higher thymic output and a more precipitous decline of thymic function than those without vulvodynia. It also seems that a strong immune inflammatory response is present proximate to the onset of vulvar pain and may wane subsequently over time.
Collapse
Affiliation(s)
- Sydney K Willis
- Department of Epidemiology, Boston University School of Public Health, Boston, Massachusetts
| | - Allison E Aiello
- Department of Epidemiology, University of North Carolina Gillings School of Global Public Health, Chapel Hill, North Carolina
| | | | - Julie A. Nelson
- Department of Microbiology and Immunology, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| | - Patricia Hibberd
- Department of Global Health, Boston University School of Public Health, Boston, Massachusetts
| | - Bernard L. Harlow
- Department of Epidemiology, Boston University School of Public Health, Boston, Massachusetts
| |
Collapse
|
19
|
Han J, Zúñiga-Pflücker JC. A 2020 View of Thymus Stromal Cells in T Cell Development. THE JOURNAL OF IMMUNOLOGY 2021; 206:249-256. [PMID: 33397738 DOI: 10.4049/jimmunol.2000889] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 10/29/2020] [Indexed: 12/12/2022]
Abstract
The thymus is an intricate primary lymphoid organ, wherein bone marrow-derived lymphoid progenitor cells are induced to develop into functionally competent T cells that express a diverse TCR repertoire, which is selected to allow for the recognition of foreign Ags while avoiding self-reactivity or autoimmunity. Thymus stromal cells, which can include all non-T lineage cells, such as thymic epithelial cells, endothelial cells, mesenchymal/fibroblast cells, dendritic cells, and B cells, provide signals that are essential for thymocyte development as well as for the homeostasis of the thymic stroma itself. In this brief review, we focus on the key roles played by thymic stromal cells during early stages of T cell development, such as promoting the homing of thymic-seeding progenitors, inducing T lineage differentiation, and supporting thymocyte survival and proliferation. We also discuss recent advances on the transcriptional regulation that govern thymic epithelial cell function as well as the cellular and molecular changes that are associated with thymic involution and regeneration.
Collapse
Affiliation(s)
- Jianxun Han
- Sunnybrook Research Institute, Toronto, Ontario M4N 3M5, Canada; and
| | - Juan Carlos Zúñiga-Pflücker
- Sunnybrook Research Institute, Toronto, Ontario M4N 3M5, Canada; and.,Department of Immunology, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| |
Collapse
|
20
|
Nutrition-Based Management of Inflammaging in CKD and Renal Replacement Therapies. Nutrients 2021; 13:nu13010267. [PMID: 33477671 PMCID: PMC7831904 DOI: 10.3390/nu13010267] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 01/15/2021] [Accepted: 01/15/2021] [Indexed: 12/12/2022] Open
Abstract
Access to renal transplantation guarantees a substantial improvement in the clinical condition and quality of life (QoL) for end-stage renal disease (ESRD) patients. In recent years, a greater number of older patients starting renal replacement therapies (RRT) have shown the long-term impact of conservative therapies for advanced CKD and the consequences of the uremic milieu, with a frail clinical condition that impacts not only their survival but also limits their access to transplantation. This process, referred to as “inflammaging,” might be reversible with a tailored approach, such as RRT accompanied by specific nutritional support. In this review, we summarize the evidence demonstrating the presence of several proinflammatory substances in the Western diet (WD) and the positive effect of unprocessed food consumption and increased fruit and vegetable intake, suggesting a new approach to reduce inflammaging with the improvement of ESRD clinical status. We conclude that the Mediterranean diet (MD), because of its modulative effects on microbiota and its anti-inflammaging properties, may be a cornerstone in a more precise nutritional support for patients on the waiting list for kidney transplantation.
Collapse
|
21
|
Ahn SH, Nguyen SL, Petroff MG. Exploring the Origin and Antigenic Specificity of Maternal Regulatory T Cells in Pregnancy. Front Immunol 2020; 11:1302. [PMID: 32670288 PMCID: PMC7330120 DOI: 10.3389/fimmu.2020.01302] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Accepted: 05/22/2020] [Indexed: 12/22/2022] Open
Abstract
Successful pregnancy outcome is partially determined by the suppression of reactive effector T cells by maternal regulatory T cells (TRegs) at the maternal-fetal interface. While a large area of research has focused on the regulation of peripherally-induced TReg (pTReg) distribution and differentiation using transgenic mouse models and human samples, studies focusing on the role of TRegs derived from the thymus (tTRegs), and the potential role of central tolerance in maternal-fetal tolerance is less explored. The genome of the fetus is composed of both the tissue-specific and paternally-inherited antigens, and a break in maternal immune tolerance to either antigen may result in adverse pregnancy outcomes. Notably, "self"-antigens, including antigens that are highly restricted to the fetus and placenta, are promiscuously expressed by medullary thymic epithelial cells under the control of Autoimmune Regulator (Aire), which skews the tTReg T cell receptor (TCR) repertoire to be specific toward these antigens. TRegs that circulate in mothers during pregnancy may be comprised of TRegs that stem from the thymus as well as those induced in the periphery. Moreover, despite a wealth of research dedicated to elucidating the function of TRegs in maternal-fetal tolerance, little is understood about the origin of these cells, and whether/how tTRegs may contribute. Investigation into this question is complicated by the absence of reliable markers to distinguish between the two. In this review, we discuss how distinct types of fetal/placental antigens may determine the generation of different subtypes of TReg cells in the mother, and in turn how these may promote maternal tolerance to the fetus in pregnancy.
Collapse
Affiliation(s)
- Soo Hyun Ahn
- Department of Pathobiology Diagnostic Investigation, College of Veterinary Medicine, Michigan State University, East Lansing, MI, United States
| | - Sean L Nguyen
- Institute for Integrative Toxicology, Michigan State University, East Lansing, MI, United States.,Cell and Molecular Biology Program, Michigan State University, East Lansing, MI, United States
| | - Margaret G Petroff
- Department of Pathobiology Diagnostic Investigation, College of Veterinary Medicine, Michigan State University, East Lansing, MI, United States.,Cell and Molecular Biology Program, Michigan State University, East Lansing, MI, United States.,Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI, United States
| |
Collapse
|
22
|
Cowan JE, Takahama Y, Bhandoola A, Ohigashi I. Postnatal Involution and Counter-Involution of the Thymus. Front Immunol 2020; 11:897. [PMID: 32477366 PMCID: PMC7235445 DOI: 10.3389/fimmu.2020.00897] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 04/17/2020] [Indexed: 11/15/2022] Open
Abstract
Thymus involution occurs in all vertebrates. It is thought to impact on immune responses in the aged, and in other clinical circumstances such as bone marrow transplantation. Determinants of thymus growth and size are beginning to be identified. Ectopic expression of factors like cyclin D1 and Myc in thymic epithelial cells (TEC)s results in considerable increase in thymus size. These models provide useful experimental tools that allow thymus function to be understood. In future, understanding TEC-specific controllers of growth will provide new approaches to thymus regeneration.
Collapse
Affiliation(s)
- Jennifer E Cowan
- Laboratory of Genome Integrity, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Yousuke Takahama
- Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Avinash Bhandoola
- Laboratory of Genome Integrity, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Izumi Ohigashi
- Division of Experimental Immunology, Institute of Advanced Medical Sciences, University of Tokushima, Tokushima, Japan
| |
Collapse
|
23
|
Molecular Mechanisms of Premature Aging in Hemodialysis: The Complex Interplay Between Innate and Adaptive Immune Dysfunction. Int J Mol Sci 2020; 21:ijms21103422. [PMID: 32408613 PMCID: PMC7279398 DOI: 10.3390/ijms21103422] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 05/01/2020] [Accepted: 05/07/2020] [Indexed: 02/07/2023] Open
Abstract
Hemodialysis (HD) patient are known to be susceptible to a wide range of early and long-term complication such as chronic inflammation, infections, malnutrition, and cardiovascular disease that significantly affect the incidence of mortality. A large gap between the number of people with end-stage kidney disease (ESKD) and patients who received kidney transplantation has been identified. Therefore, there is a huge need to explore the underlying pathophysiology of HD complications in order to provide treatment guidelines. The immunological dysregulation, involving both the innate and adaptive response, plays a crucial role during the HD sessions and in chronic, maintenance treatments. Innate immune system mediators include the dysfunction of neutrophils, monocytes, and natural killer (NK) cells with signaling mediated by NOD-like receptor P3 (NLRP3) and Toll-like receptor 4 (TLR4); in addition, there is a significant activation of the complement system that is mediated by dialysis membrane-surfaces. These effectors induce a persistent, systemic, pro-inflammatory, and pro-coagulant milieu that has been described as inflammaging. The adaptive response, the imbalance in the CD4+/CD8+ T cell ratio, and the reduction of Th2 and regulatory T cells, together with an altered interaction with B lymphocyte by CD40/CD40L, have been mainly implicated in immune system dysfunction. Altogether, these observations suggest that intervention targeting the immune system in HD patients could improve morbidity and mortality. The purpose of this review is to expand our understanding on the role of immune dysfunction in both innate and adaptive response in patients undergoing hemodialysis treatment.
Collapse
|
24
|
Dumont-Lagacé M, Daouda T, Depoërs L, Zumer J, Benslimane Y, Brochu S, Harrington L, Lemieux S, Perreault C. Qualitative Changes in Cortical Thymic Epithelial Cells Drive Postpartum Thymic Regeneration. Front Immunol 2020; 10:3118. [PMID: 32010151 PMCID: PMC6974522 DOI: 10.3389/fimmu.2019.03118] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Accepted: 12/20/2019] [Indexed: 12/05/2022] Open
Abstract
During gestation, sex hormones cause a significant thymic involution which enhances fertility. This thymic involution is rapidly corrected following parturition. As thymic epithelial cells (TECs) are responsible for the regulation of thymopoiesis, we analyzed the sequential phenotypic and transcriptomic changes in TECs during the postpartum period in order to identify mechanisms triggering postpartum thymic regeneration. In particular, we performed flow cytometry analyses and deep RNA-sequencing on purified TEC subsets at several time points before and after parturition. We report that pregnancy-induced involution is not caused by loss of TECs since their number does not change during or after pregnancy. However, during pregnancy, we observed a significant depletion of all thymocyte subsets downstream of the double-negative 1 (DN1) differentiation stage. Variations in thymocyte numbers correlated with conspicuous changes in the transcriptome of cortical TECs (cTECs). The transcriptomic changes affected predominantly cTEC expression of Foxn1, its targets and several genes that are essential for thymopoiesis. By contrast, medullary TECs (mTECs) showed very little transcriptomic changes in the early postpartum regenerative phase, but seemed to respond to the expansion of single-positive (SP) thymocytes in the late phase of regeneration. Together, these results show that postpartum thymic regeneration is orchestrated by variations in expression of a well-defined subset of cTEC genes, that occur very early after parturition.
Collapse
Affiliation(s)
- Maude Dumont-Lagacé
- Immunobiology Research Unit, Department of Medicine, Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC, Canada
| | - Tariq Daouda
- Immunobiology Research Unit, Department of Medicine, Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC, Canada.,Functional and Structural Bioinformatics Research Unit, Department of Biochemistry and Molecular Medicine, Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC, Canada
| | - Lucyle Depoërs
- Immunobiology Research Unit, Department of Medicine, Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC, Canada
| | - Jérémie Zumer
- Functional and Structural Bioinformatics Research Unit, Department of Biochemistry and Molecular Medicine, Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC, Canada
| | - Yahya Benslimane
- Telomere Length Homeostasis and Genomic Instability Research Unit, Department of Medicine, Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC, Canada
| | - Sylvie Brochu
- Immunobiology Research Unit, Department of Medicine, Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC, Canada
| | - Lea Harrington
- Telomere Length Homeostasis and Genomic Instability Research Unit, Department of Medicine, Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC, Canada
| | - Sébastien Lemieux
- Functional and Structural Bioinformatics Research Unit, Department of Biochemistry and Molecular Medicine, Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC, Canada
| | - Claude Perreault
- Immunobiology Research Unit, Department of Medicine, Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC, Canada
| |
Collapse
|
25
|
[Premature immune senescence and chronic kidney disease: Update and perspectives]. Nephrol Ther 2019; 16:9-18. [PMID: 31848067 DOI: 10.1016/j.nephro.2019.04.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Accepted: 04/12/2019] [Indexed: 12/14/2022]
Abstract
Immune senescence is associated with age-related diseases (i.e. infectious disease, cardiovascular diseases and cancers). Chronic kidney disease patients die prematurely when compared with general population, because of a higher occurrence of infections, cardiovascular events and cancer. These diseases are commonly observed in the elderly population and frequently associated with immune senescence. Indeed, chronic kidney disease causes a premature aging of the T lymphocyte compartment, widely related to a decrease in thymic function, a phenomenon that plays a key role in the onset of age-related diseases in chronic kidney disease patients. The degree of immune senescence also influences patients' outcome after renal transplantation, particularly the risk of acute rejection and infections. Partial reversion of pre-transplant immune senescence is observed for some renal transplant patients. In conclusion, to reduce the increasing incidence of morbidity and mortality of chronic kidney disease patients, a better knowledge of uremia-induced immune senescence would help to pave the way to build clinical studies and promote innovative therapeutic approaches. We believe that therapeutic reversion and immune senescence prevention approaches will be part of the management of chronic kidney disease patients in the future.
Collapse
|
26
|
Pereira M, Valério-Bolas A, Saraiva-Marques C, Alexandre-Pires G, Pereira da Fonseca I, Santos-Gomes G. Development of Dog Immune System: From in Uterus to Elderly. Vet Sci 2019; 6:E83. [PMID: 31640234 PMCID: PMC6958461 DOI: 10.3390/vetsci6040083] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 10/10/2019] [Accepted: 10/12/2019] [Indexed: 12/12/2022] Open
Abstract
Immune system recognize and fight back foreign microorganisms and inner modifications that lead to deficient cell and tissue functions. During a dog's life, the immune system needs to adapt to different physiological conditions, assuring surveillance and protection in a careful and controlled way. Pregnancy alters normal homeostasis, requiring a balance between immunity and tolerance. The embryos and fetus should be protected from infections, while the female dog must tolerate the growing of semi-allografts in her uterus. After birth, newborn puppies are at great risk of developing infectious diseases, because their immune system is in development and immune memory is absent. Passive transfer of immunity through colostrum is fundamental for puppy survival in the first weeks of life, but hampers the development of an active immune response to vaccination. At the end of life, dogs experience a decline in the structure and functional competence of the immune system, compromising the immune responses to novel antigenic challenges, such as infections and vaccines. Therefore, the current article reviews the general processes related to the development of the dog´s immune system, providing an overview of immune activity throughout the dog's life and its implications in canine health, and highlighting priority research goals.
Collapse
Affiliation(s)
- Maria Pereira
- Global Health and Tropical Medicine (GHTM), Instituto de Higiene e Medicina Tropical (IHMT), Universidade Nova de Lisboa (UNL), R. da Junqueira 100, 1349-008 Lisboa, Portugal.
- Instituto Politécnico de Portalegre (IPP), Praça do Município 11, 7300-110 Portalegre, Portugal.
- Agrarian School of the Polytechnic Institute of Viseu, Quinta da Alagoa-Estrada de Nelas Ranhados, 3500-606 Viseu, Portugal.
| | - Ana Valério-Bolas
- Global Health and Tropical Medicine (GHTM), Instituto de Higiene e Medicina Tropical (IHMT), Universidade Nova de Lisboa (UNL), R. da Junqueira 100, 1349-008 Lisboa, Portugal.
| | - Cátia Saraiva-Marques
- Centro Interdisciplinar de Investigação em Sanidade Animal (CIISA), Faculdade de Medicina Veterinária (FMV), Universidade de Lisboa (UL), Av. Universidade Técnica, 1300-477 Lisboa, Portugal.
| | - Graça Alexandre-Pires
- Centro Interdisciplinar de Investigação em Sanidade Animal (CIISA), Faculdade de Medicina Veterinária (FMV), Universidade de Lisboa (UL), Av. Universidade Técnica, 1300-477 Lisboa, Portugal.
| | - Isabel Pereira da Fonseca
- Centro Interdisciplinar de Investigação em Sanidade Animal (CIISA), Faculdade de Medicina Veterinária (FMV), Universidade de Lisboa (UL), Av. Universidade Técnica, 1300-477 Lisboa, Portugal.
| | - Gabriela Santos-Gomes
- Global Health and Tropical Medicine (GHTM), Instituto de Higiene e Medicina Tropical (IHMT), Universidade Nova de Lisboa (UNL), R. da Junqueira 100, 1349-008 Lisboa, Portugal.
| |
Collapse
|
27
|
Moldenhauer LM, Schjenken JE, Hope CM, Green ES, Zhang B, Eldi P, Hayball JD, Barry SC, Robertson SA. Thymus-Derived Regulatory T Cells Exhibit Foxp3 Epigenetic Modification and Phenotype Attenuation after Mating in Mice. THE JOURNAL OF IMMUNOLOGY 2019; 203:647-657. [DOI: 10.4049/jimmunol.1900084] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 05/20/2019] [Indexed: 12/30/2022]
|
28
|
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
|