1
|
Glorieux C, Buc Calderon P. Targeting catalase in cancer. Redox Biol 2024; 77:103404. [PMID: 39447253 PMCID: PMC11539659 DOI: 10.1016/j.redox.2024.103404] [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: 08/26/2024] [Revised: 10/11/2024] [Accepted: 10/17/2024] [Indexed: 10/26/2024] Open
Abstract
Healthy cells have developed a sophisticated network of antioxidant molecules to prevent the toxic accumulation of reactive oxygen species (ROS) generated by diverse environmental stresses. On the opposite, cancer cells often exhibit high levels of ROS and an altered levels of antioxidant molecules compared to normal cells. Among them, the antioxidant enzyme catalase plays an essential role in cell defense against oxidative stress through the dismutation of hydrogen peroxide into water and molecular oxygen, and its expression is often decreased in cancer cells. The elevation of ROS in cancer cells provides them proliferative advantages, and leads to metabolic reprogramming, immune escape and metastasis. In this context, catalase is of critical importance to control these cellular processes in cancer through various mechanisms. In this review, we will discuss the major progresses and challenges in understanding the role of catalase in cancer for this last decade. This review also aims to provide important updates regarding the regulation of catalase expression, subcellular localization and discuss about the potential role of microbial catalases in tumor environment. Finally, we will describe the different catalase-based therapies and address the advantages, disadvantages, and limitations associated with modulating catalase therapeutically in cancer treatment.
Collapse
Affiliation(s)
- Christophe Glorieux
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, 510060, Guangzhou, China.
| | - Pedro Buc Calderon
- Química y Farmacia, Facultad de Ciencias de La Salud, Universidad Arturo Prat, 1100000, Iquique, Chile; Instituto de Química Medicinal, Universidad Arturo Prat, 1100000, Iquique, Chile; Research Group in Metabolism and Nutrition, Louvain Drug Research Institute, Université Catholique de Louvain, 1200, Brussels, Belgium.
| |
Collapse
|
2
|
Yamaguchi N, Takakura Y, Akiyama T. Autophagy and proteasomes in thymic epithelial cells: essential bulk protein degradation systems for immune homeostasis maintenance. Front Immunol 2024; 15:1488020. [PMID: 39524450 PMCID: PMC11543444 DOI: 10.3389/fimmu.2024.1488020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Accepted: 10/09/2024] [Indexed: 11/16/2024] Open
Abstract
The thymus is a central organ that controls T cell development. Thymic epithelial cells (TECs) create a unique microenvironment essential for the differentiation of major histocompatibility complex (MHC)-restricted and self-tolerant T cells. TECs present a complex of self-peptides and MHC molecules (self-pMHCs) to immature T cells and regulate their survival and differentiation based on their affinity for self-pMHCs. The processing of self-peptides in TECs depends on bulk protein degradation systems, specifically autophagy and proteasomes. Studies using autophagy- and proteasome-deficient mouse models have demonstrated that these degradation systems in TECs are indispensable for maintaining immune homeostasis. Although autophagy and proteasomes are ubiquitous in nearly all eukaryotic cells, TECs exhibit unique characteristics in their autophagy and proteasome functions. Autophagy in TECs is constitutively active and independent of stress responses, while TEC proteasomes contain specialized catalytic subunits. This review summarizes the distinctive characteristics of autophagy and proteasomes in TECs and their roles in immune system regulation.
Collapse
Affiliation(s)
- Noritaka Yamaguchi
- Department of Molecular Cardiovascular Pharmacology, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, Japan
- Laboratory for Immune Homeostasis, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Yuki Takakura
- Department of Molecular Cardiovascular Pharmacology, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, Japan
- Laboratory for Immune Homeostasis, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Taishin Akiyama
- Laboratory for Immune Homeostasis, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| |
Collapse
|
3
|
Jing H, Song J, Sun J, Su S, Hu J, Zhang H, Bi Y, Wu B. METTL3 governs thymocyte development and thymic involution by regulating ferroptosis. NATURE AGING 2024:10.1038/s43587-024-00724-x. [PMID: 39443728 DOI: 10.1038/s43587-024-00724-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Accepted: 09/16/2024] [Indexed: 10/25/2024]
Abstract
Given its central role in immune aging, it is important to identify the regulators of thymic involution. While conventional programmed cell death has a fundamental role in thymocyte development, how cell death pathways contribute to thymic involution are unclear. In this study, we found that CD4+CD8+ double-positive (DP) thymocytes acquired the characteristics of senescence in aged mice undergoing thymic involution, while expression of the m6A methyltransferase-like protein 3 (METTL3), which is enriched in DP cells from young mice, decreased with aging. By conditionally deleting METTL3 in T cells, we revealed a critical role for METTL3 in DP cell survival and in restraining the features of aging in DP thymocytes by preventing ferroptosis signaling through glutathione peroxidase 4. Mechanistically, glutathione peroxidase 4 was maintained by METTL3 at the translational level, independently of its methyltransferase activity. Furthermore, we found that pharmacological inhibition of ferroptosis promoted DP cell survival and attenuated the features of aging in DP thymocytes. These findings uncover a role for METTL3-regulated ferroptosis in thymic involution and identify strategies to restore thymic function.
Collapse
Affiliation(s)
- Huiru Jing
- Department of Urology, State Key Laboratory of Virology, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
- Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, China
| | - Jiayu Song
- Department of Urology, State Key Laboratory of Virology, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
- Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, China
| | - Jie Sun
- Department of Urology, State Key Laboratory of Virology, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
- Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, China
| | - Shaojun Su
- Department of Urology, State Key Laboratory of Virology, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
- Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, China
| | - Jin Hu
- Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, China
| | - Haojian Zhang
- Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, China
| | - Yanmin Bi
- Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, China.
| | - Bing Wu
- Department of Urology, State Key Laboratory of Virology, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China.
- Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, China.
| |
Collapse
|
4
|
Dourado TMH, Nascimento DC, Rosa MH, Assis VO, Pimenta GF, Alves-Filho JC, Tirapelli CR. Mineralocorticoid receptor antagonism partially prevents dysfunction of T cell maturation in rats chronically treated with ethanol. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024:10.1007/s00210-024-03382-3. [PMID: 39172146 DOI: 10.1007/s00210-024-03382-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 08/14/2024] [Indexed: 08/23/2024]
Abstract
Ethanol consumption induces thymic atrophy and affects T cell maturation in the thymus. However, the mechanisms underlying such effects still need to be fully understood. We attempted to investigate the role of mineralocorticoid receptors (MR) on ethanol-induced thymic atrophy, T cell maturation dysfunction, and the role of oxidative stress in such responses. Male Wistar Hannover rats were treated with ethanol (20%; in volume ratio) and/or potassium canrenoate, an antagonist of MR (MRA; 30 mg/kg/day, gavage) for five weeks. Blockade of MR prevented ethanol-induced increases in the number of double-positive (CD4+CD8+), CD8+ single-positive (CD4-CD8+), CD4+ single-positive (CD4+CD8-), and Foxp3+CD4+ (Treg) cells in the thymus. Ethanol increased NOX2-derived superoxide (O2•-), lipoperoxidation, and superoxide dismutase (SOD) activity in the thymus. Pretreatment with the MRA fully prevented these responses. Apocynin, an antioxidant, prevented ethanol-induced increases in the number of double-positive and CD8+ single-positive cells but failed to prevent the rise in the number of CD4+ single-positive and Treg cells induced by ethanol. Apocynin, but not the MRA, prevented thymic atrophy induced by ethanol. Our findings provided novel evidence for the participation of MR in thymic dysfunction induced by ethanol consumption. Oxidative stress mediates the increase in double-positive and CD8+ single-positive cells in response to MR activation, while positive regulation of CD4+ single-positive and Treg cells is independent of oxidative stress. Oxidative stress is a significant mechanism of thymic atrophy associated with ethanol consumption, but this response is independent of MR activation.
Collapse
Affiliation(s)
- Thales M H Dourado
- Laboratório de Farmacologia, Escola de Enfermagem de Ribeirão Preto, Universidade de São Paulo (USP), Avenida Bandeirantes 3900, Ribeirão Preto, São Paulo, CEP 14040-902, Brazil
- Departamento de Farmacologia, Faculdade de Medicina de Ribeirão Preto, USP, Ribeirão Preto, São Paulo, Brazil
| | - Daniele C Nascimento
- Departamento de Farmacologia, Faculdade de Medicina de Ribeirão Preto, USP, Ribeirão Preto, São Paulo, Brazil
- Centro de Pesquisa Em Doenças Inflamatórias, Faculdade de Medicina de Ribeirão Preto, USP, Ribeirão Preto, São Paulo, Brazil
| | - Marcos H Rosa
- Departamento de Farmacologia, Faculdade de Medicina de Ribeirão Preto, USP, Ribeirão Preto, São Paulo, Brazil
- Centro de Pesquisa Em Doenças Inflamatórias, Faculdade de Medicina de Ribeirão Preto, USP, Ribeirão Preto, São Paulo, Brazil
| | - Victor O Assis
- Laboratório de Farmacologia, Escola de Enfermagem de Ribeirão Preto, Universidade de São Paulo (USP), Avenida Bandeirantes 3900, Ribeirão Preto, São Paulo, CEP 14040-902, Brazil
- Departamento de Farmacologia, Faculdade de Medicina de Ribeirão Preto, USP, Ribeirão Preto, São Paulo, Brazil
| | - Gustavo F Pimenta
- Laboratório de Farmacologia, Escola de Enfermagem de Ribeirão Preto, Universidade de São Paulo (USP), Avenida Bandeirantes 3900, Ribeirão Preto, São Paulo, CEP 14040-902, Brazil
- Departamento de Farmacologia, Faculdade de Medicina de Ribeirão Preto, USP, Ribeirão Preto, São Paulo, Brazil
| | - José C Alves-Filho
- Departamento de Farmacologia, Faculdade de Medicina de Ribeirão Preto, USP, Ribeirão Preto, São Paulo, Brazil
- Centro de Pesquisa Em Doenças Inflamatórias, Faculdade de Medicina de Ribeirão Preto, USP, Ribeirão Preto, São Paulo, Brazil
| | - Carlos R Tirapelli
- Laboratório de Farmacologia, Escola de Enfermagem de Ribeirão Preto, Universidade de São Paulo (USP), Avenida Bandeirantes 3900, Ribeirão Preto, São Paulo, CEP 14040-902, Brazil.
- Departamento de Ciências BioMoleculares, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, USP, Ribeirão Preto, São Paulo, Brazil.
| |
Collapse
|
5
|
Kologrivova IV, Naryzhnaya NV, Suslova TE. Thymus in Cardiometabolic Impairments and Atherosclerosis: Not a Silent Player? Biomedicines 2024; 12:1408. [PMID: 39061983 PMCID: PMC11273826 DOI: 10.3390/biomedicines12071408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 06/11/2024] [Accepted: 06/21/2024] [Indexed: 07/28/2024] Open
Abstract
The thymus represents a primary organ of the immune system, harboring the generation and maturation of T lymphocytes. Starting from childhood, the thymus undergoes involution, being replaced with adipose tissue, and by an advanced age nearly all the thymus parenchyma is represented by adipocytes. This decline of thymic function is associated with compromised maturation and selection of T lymphocytes, which may directly impact the development of inflammation and induce various autoinflammatory disorders, including atherosclerosis. For a long time, thymus health in adults has been ignored. The process of adipogenesis in thymus and impact of thymic fat on cardiometabolism remains a mysterious process, with many issues being still unresolved. Meanwhile, thymus functional activity has a potential to be regulated, since islets of thymopoeisis remain in adults even at an advanced age. The present review describes the intricate process of thymic adipose involution, focusing on the issues of the thymus' role in the development of atherosclerosis and metabolic health, tightly interconnected with the state of vessels. We also review the recent information on the key molecular pathways and biologically active substances that may be targeted to manipulate both thymic function and atherosclerosis.
Collapse
Affiliation(s)
- Irina V. Kologrivova
- Cardiology Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, 111A Kievskaya, Tomsk 634012, Russia; (N.V.N.); (T.E.S.)
| | | | | |
Collapse
|
6
|
Kaminski HJ, Kusner LL, Cutter GR, Le Panse R, Wright CD, Perry Y, Wolfe GI. Does Surgical Removal of the Thymus Have Deleterious Consequences? Neurology 2024; 102:e209482. [PMID: 38781559 PMCID: PMC11226319 DOI: 10.1212/wnl.0000000000209482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 03/28/2024] [Indexed: 05/25/2024] Open
Abstract
The role of immunosenescence, particularly the natural process of thymic involution during aging, is increasingly acknowledged as a factor contributing to the development of autoimmune diseases and cancer. Recently, a concern has been raised about deleterious consequences of the surgical removal of thymic tissue, including for patients who undergo thymectomy for myasthenia gravis (MG) or resection of a thymoma. This review adopts a multidisciplinary approach to scrutinize the evidence concerning the long-term risks of cancer and autoimmunity postthymectomy. We conclude that for patients with acetylcholine receptor antibody-positive MG and those diagnosed with thymoma, the removal of the thymus offers prominent benefits that well outweigh the potential risks. However, incidental removal of thymic tissue during other thoracic surgeries should be minimized whenever feasible.
Collapse
Affiliation(s)
- Henry J Kaminski
- From the Department of Neurology & Rehabilitation Medicine (H.J.K.), George Washington University, DC; Department of Pharmacology & Physiology (L.L.K.), and Department of Biostatistics (G.R.C.), University of Alabama at Birmingham; INSERM (R.L.P.), Institute of Myology, Center of Research in Myology, Sorbonne University, Paris, France; Department of Surgery (C.D.W.), Harvard Medical School, Boston, MA; and Department of Surgery (Y.P.), and Department of Neurology (G.I.W.), Jacobs School of Medicine and Biomedical Sciences, University at Buffalo/SUNY, NY
| | - Linda L Kusner
- From the Department of Neurology & Rehabilitation Medicine (H.J.K.), George Washington University, DC; Department of Pharmacology & Physiology (L.L.K.), and Department of Biostatistics (G.R.C.), University of Alabama at Birmingham; INSERM (R.L.P.), Institute of Myology, Center of Research in Myology, Sorbonne University, Paris, France; Department of Surgery (C.D.W.), Harvard Medical School, Boston, MA; and Department of Surgery (Y.P.), and Department of Neurology (G.I.W.), Jacobs School of Medicine and Biomedical Sciences, University at Buffalo/SUNY, NY
| | - Gary R Cutter
- From the Department of Neurology & Rehabilitation Medicine (H.J.K.), George Washington University, DC; Department of Pharmacology & Physiology (L.L.K.), and Department of Biostatistics (G.R.C.), University of Alabama at Birmingham; INSERM (R.L.P.), Institute of Myology, Center of Research in Myology, Sorbonne University, Paris, France; Department of Surgery (C.D.W.), Harvard Medical School, Boston, MA; and Department of Surgery (Y.P.), and Department of Neurology (G.I.W.), Jacobs School of Medicine and Biomedical Sciences, University at Buffalo/SUNY, NY
| | - Rozen Le Panse
- From the Department of Neurology & Rehabilitation Medicine (H.J.K.), George Washington University, DC; Department of Pharmacology & Physiology (L.L.K.), and Department of Biostatistics (G.R.C.), University of Alabama at Birmingham; INSERM (R.L.P.), Institute of Myology, Center of Research in Myology, Sorbonne University, Paris, France; Department of Surgery (C.D.W.), Harvard Medical School, Boston, MA; and Department of Surgery (Y.P.), and Department of Neurology (G.I.W.), Jacobs School of Medicine and Biomedical Sciences, University at Buffalo/SUNY, NY
| | - Cameron D Wright
- From the Department of Neurology & Rehabilitation Medicine (H.J.K.), George Washington University, DC; Department of Pharmacology & Physiology (L.L.K.), and Department of Biostatistics (G.R.C.), University of Alabama at Birmingham; INSERM (R.L.P.), Institute of Myology, Center of Research in Myology, Sorbonne University, Paris, France; Department of Surgery (C.D.W.), Harvard Medical School, Boston, MA; and Department of Surgery (Y.P.), and Department of Neurology (G.I.W.), Jacobs School of Medicine and Biomedical Sciences, University at Buffalo/SUNY, NY
| | - Yaron Perry
- From the Department of Neurology & Rehabilitation Medicine (H.J.K.), George Washington University, DC; Department of Pharmacology & Physiology (L.L.K.), and Department of Biostatistics (G.R.C.), University of Alabama at Birmingham; INSERM (R.L.P.), Institute of Myology, Center of Research in Myology, Sorbonne University, Paris, France; Department of Surgery (C.D.W.), Harvard Medical School, Boston, MA; and Department of Surgery (Y.P.), and Department of Neurology (G.I.W.), Jacobs School of Medicine and Biomedical Sciences, University at Buffalo/SUNY, NY
| | - Gil I Wolfe
- From the Department of Neurology & Rehabilitation Medicine (H.J.K.), George Washington University, DC; Department of Pharmacology & Physiology (L.L.K.), and Department of Biostatistics (G.R.C.), University of Alabama at Birmingham; INSERM (R.L.P.), Institute of Myology, Center of Research in Myology, Sorbonne University, Paris, France; Department of Surgery (C.D.W.), Harvard Medical School, Boston, MA; and Department of Surgery (Y.P.), and Department of Neurology (G.I.W.), Jacobs School of Medicine and Biomedical Sciences, University at Buffalo/SUNY, NY
| |
Collapse
|
7
|
Dinges SS, Amini K, Notarangelo LD, Delmonte OM. Primary and secondary defects of the thymus. Immunol Rev 2024; 322:178-211. [PMID: 38228406 PMCID: PMC10950553 DOI: 10.1111/imr.13306] [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: 01/18/2024]
Abstract
The thymus is the primary site of T-cell development, enabling generation, and selection of a diverse repertoire of T cells that recognize non-self, whilst remaining tolerant to self- antigens. Severe congenital disorders of thymic development (athymia) can be fatal if left untreated due to infections, and thymic tissue implantation is the only cure. While newborn screening for severe combined immune deficiency has allowed improved detection at birth of congenital athymia, thymic disorders acquired later in life are still underrecognized and assessing the quality of thymic function in such conditions remains a challenge. The thymus is sensitive to injury elicited from a variety of endogenous and exogenous factors, and its self-renewal capacity decreases with age. Secondary and age-related forms of thymic dysfunction may lead to an increased risk of infections, malignancy, and autoimmunity. Promising results have been obtained in preclinical models and clinical trials upon administration of soluble factors promoting thymic regeneration, but to date no therapy is approved for clinical use. In this review we provide a background on thymus development, function, and age-related involution. We discuss disease mechanisms, diagnostic, and therapeutic approaches for primary and secondary thymic defects.
Collapse
Affiliation(s)
- Sarah S. Dinges
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Kayla Amini
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Luigi D. Notarangelo
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Ottavia M. Delmonte
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
8
|
Xu X, Tao N, Sun C, Hoffman RD, Shi D, Ying Y, Dong S, Gao J. Ligustilide prevents thymic immune senescence by regulating Thymosin β15-dependent spatial distribution of thymic epithelial cells. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 123:155216. [PMID: 38061285 DOI: 10.1016/j.phymed.2023.155216] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 11/02/2023] [Accepted: 11/11/2023] [Indexed: 01/17/2024]
Abstract
BACKGROUND Thymus is the most crucial organ connecting immunity and aging. The progressive senescence of thymic epithelial cells (TECs) leads to the involution of thymus under aging, chronic stress and other factors. Ligustilide (LIG) is a major active component of the anti-aging Chinese herbal medicine Angelica sinensis (Oliv.) Diels, but its role in preventing TEC-based thymic aging remains elusive. PURPOSE This study explored the protective role of Ligustilide in alleviating ADM (adriamycin) -induced thymic immune senescence and its underlying molecular mechanisms. METHOD The protective effect of Ligustilide on ADM-induced thymic atrophy was examined by mouse and organotypic models, and conformed by SA-β-gal staining in TECs. The abnormal spatial distribution of TECs in the senescent thymus was analyzed using H&E, immunofluorescence and flow cytometry. The possible mechanisms of Ligustilide in ADM-induced thymic aging were elucidated by qPCR, fluorescence labeling and Western blot. The mechanism of Ligustilide was subsequently validated through actin polymerization inhibitor, genetic engineering to regulate Thymosin β15 (Tβ15) and Tβ4 expression, molecular docking and β Thymosin-G-actin cross-linking assay. RESULTS At a 5 mg/kg dose, Ligustilide markedly ameliorated ADM-induced weight loss and limb grip weakness in mice. It also reversed thymic damage and restored positive selection impaired by ADM. In vitro, ADM disrupted thymic structure, reduced TECs number and hindered double negative (DN) T cell differentiation. Ligustilide counteracted these effects, promoted TEC proliferation and reticular differentiation, leading to an increase in CD4+ single positive (CD4SP) T cell proportion. Mechanistically, ADM diminished the microfilament quantity in immortalized TECs (iTECs), and lowered the expression of cytoskeletal marker proteins. Molecular docking and cross-linking assay revealed that Ligustilide inhibited the protein binding between G-actin and Tβ15 by inhibiting the formation of the Tβ15-G-actin complex, thus enhancing the microfilament assembly capacity in TECs. CONCLUSION This study, for the first time, reveals that Ligustilide can attenuate actin depolymerization, protects TECs from ADM-induced acute aging by inhibiting the binding of Tβ15 to G-actin, thereby improving thymic immune function. Moreover, it underscores the interesting role of Ligustilide in maintaining cytoskeletal assembly and network structure of TECs, offering a novel perspective for deeper understanding of anti thymic aging.
Collapse
Affiliation(s)
- Xie Xu
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, China; Zhejiang Provincial Hospital of Chinese Medicine, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, China.
| | - Nana Tao
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, China.
| | - Caihua Sun
- Zhejiang Provincial Hospital of Chinese Medicine, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, China.
| | - Robert D Hoffman
- Yo San University of Traditional Chinese Medicine, Los Angeles, CA 90066, USA.
| | - Dongling Shi
- Academy of Chinese Medical Science, Zhejiang Chinese Medical University, Hangzhou 310006, Zhejiang, China.
| | - Yuyuan Ying
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, China.
| | - Shujie Dong
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, China.
| | - Jianli Gao
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, China; State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao 999078, China.
| |
Collapse
|
9
|
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
|
10
|
Yang J, Liu J, Liang J, Li F, Wang W, Chen H, Xie X. Epithelial-mesenchymal transition in age-associated thymic involution: Mechanisms and therapeutic implications. Ageing Res Rev 2023; 92:102115. [PMID: 37922996 DOI: 10.1016/j.arr.2023.102115] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 10/17/2023] [Accepted: 10/29/2023] [Indexed: 11/07/2023]
Abstract
The thymus is a critical immune organ with endocrine and immune functions that plays important roles in the physiological and pathological processes of the body. However, with aging, the thymus undergoes degenerative changes leading to decreased production and output of naive T cells and the secretion of thymic hormones and related cytokines, thereby promoting the occurrence and development of various age-associated diseases. Therefore, identifying essential processes that regulate age-associated thymic involution is crucial for long-term control of thymic involution and age-associated disease progression. Epithelial-mesenchymal transition (EMT) is a well-established process involved in organ aging and functional impairment through tissue fibrosis in several organs, such as the heart and kidney. In the thymus, EMT promotes fibrosis and potentially adipogenesis, leading to thymic involution. This review focuses on the factors involved in thymic involution, including oxidative stress, inflammation, and hormones, from the perspective of EMT. Furthermore, current interventions for reversing age-associated thymic involution by targeting EMT-associated processes are summarized. Understanding the key mechanisms of thymic involution through EMT as an entry point may promote the development of new therapies and clinical agents to reverse thymic involution and age-associated disease.
Collapse
Affiliation(s)
- Jiali Yang
- The School of Basic Medical Sciences, Southwest Medical University, Luzhou, China; Public Center of Experimental Technology, Model Animal and Human Disease Research of Luzhou Key Laboratory, Southwest Medical University, Luzhou, China
| | - Juan Liu
- The School of Basic Medical Sciences, Southwest Medical University, Luzhou, China; Public Center of Experimental Technology, Model Animal and Human Disease Research of Luzhou Key Laboratory, Southwest Medical University, Luzhou, China
| | - Jiayu Liang
- The School of Basic Medical Sciences, Southwest Medical University, Luzhou, China; Public Center of Experimental Technology, Model Animal and Human Disease Research of Luzhou Key Laboratory, Southwest Medical University, Luzhou, China
| | - Fan Li
- The School of Basic Medical Sciences, Southwest Medical University, Luzhou, China; Public Center of Experimental Technology, Model Animal and Human Disease Research of Luzhou Key Laboratory, Southwest Medical University, Luzhou, China
| | - Wenwen Wang
- The School of Basic Medical Sciences, Southwest Medical University, Luzhou, China; Public Center of Experimental Technology, Model Animal and Human Disease Research of Luzhou Key Laboratory, Southwest Medical University, Luzhou, China
| | - Huan Chen
- The School of Basic Medical Sciences, Southwest Medical University, Luzhou, China; Nucleic Acid Medicine of Luzhou Key Laboratory, Southwest Medical University, Luzhou, China.
| | - Xiang Xie
- The School of Basic Medical Sciences, Southwest Medical University, Luzhou, China; Public Center of Experimental Technology, Model Animal and Human Disease Research of Luzhou Key Laboratory, Southwest Medical University, Luzhou, China.
| |
Collapse
|
11
|
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: 1.0] [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
|
12
|
Sonar SA, Watanabe M, Nikolich JŽ. Disorganization of secondary lymphoid organs and dyscoordination of chemokine secretion as key contributors to immune aging. Semin Immunol 2023; 70:101835. [PMID: 37651849 PMCID: PMC10840697 DOI: 10.1016/j.smim.2023.101835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 08/18/2023] [Accepted: 08/18/2023] [Indexed: 09/02/2023]
Abstract
Aging is characterized by progressive loss of organ and tissue function, and the immune system is no exception to that inevitable principle. Of all the age-related changes in the body, reduction of the size of, and naïve T (Tn) cell output from, the thymus occurs earliest, being prominent already before or by the time of puberty. Therefore, to preserve immunity against new infections, over much of their lives, vertebrates dominantly rely on peripheral maintenance of the Tn cell pool in the secondary lymphoid organs (SLO). However, SLO structure and function subsequently also deteriorate with aging. Several recent studies have made a convincing case that this deterioration is of major importance to the erosion of protective immunity in the last third of life. Specifically, the SLO were found to accumulate multiple degenerative changes with aging. Importantly, the results from adoptive transfer and parabiosis studies teach us that the old microenvironment is the limiting factor for protective immunity in old mice. In this review, we discuss the extent, mechanisms, and potential role of stromal cell aging in the age-related alteration of T cell homeostatic maintenance and immune function decline. We use that discussion to frame the potential strategies to correct the SLO stromal aging defects - in the context of other immune rejuvenation approaches, - to improve functional immune responses and protective immunity in older adults.
Collapse
Affiliation(s)
- Sandip Ashok Sonar
- Department of Immunobiology, University of Arizona College of Medicine-Tucson, Tucson, AZ, USA; The University of Arizona Center on Aging, University of Arizona College of Medicine-Tucson, Tucson, AZ, USA
| | - Makiko Watanabe
- Department of Immunobiology, University of Arizona College of Medicine-Tucson, Tucson, AZ, USA; The University of Arizona Center on Aging, University of Arizona College of Medicine-Tucson, Tucson, AZ, USA
| | - Janko Ž Nikolich
- Department of Immunobiology, University of Arizona College of Medicine-Tucson, Tucson, AZ, USA; The University of Arizona Center on Aging, University of Arizona College of Medicine-Tucson, Tucson, AZ, USA; the Aegis Consortium for Pandemic-free Future, University of Arizona Health Sciences, USA; BIO5 Institute, University of Arizona, Tucson, AZ, USA.
| |
Collapse
|
13
|
Lee S, Song SG, Chung DH. Comprehensive Transcriptomic Analysis for Thymic Epithelial Cells of Aged Mice and Humans. Immune Netw 2023; 23:e36. [PMID: 37970235 PMCID: PMC10643332 DOI: 10.4110/in.2023.23.e36] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 08/04/2023] [Accepted: 08/07/2023] [Indexed: 11/17/2023] Open
Abstract
Thymic epithelial cells (TECs) play a critical role in thymic development and thymopoiesis. As individuals age, TECs undergo various changes that impact their functions, leading to a reduction in cell numbers and impaired thymic selection. These age-related alterations have been observed in both mice and humans. However, the precise mechanisms underlying age-related TEC dysfunction remain unclear. Furthermore, there is a lack of a comprehensive study that connects mouse and human biological processes in this area. To address this gap, we conducted an extensive transcriptome analysis of young and old TECs in mice, complemented by further analysis of publicly available human TEC single-cell RNA sequencing data. Our analysis revealed alterations in both known and unknown pathways that potentially contribute to age-related TEC dysfunction. Specifically, we observed downregulation of pathways related to cell proliferation, T cell development, metabolism, and cytokine signaling in old age TECs. Conversely, TGF-β, BMP, and Wnt signaling pathways were upregulated, which have been known to be associated with age-related TEC dysfunctions or newly discovered in this study. Importantly, we found that these age-related changes in mouse TECs were consistently present in human TECs as well. This cross-species validation further strengthens the significance of our findings. In conclusion, our comprehensive analysis provides valuable insight into the biological and immunological characteristics of aged TECs in both mice and humans. These findings contribute to a better understanding of thymic involution and age-induced immune dysfunction.
Collapse
Affiliation(s)
- Sangsin Lee
- Laboratory of Immune Regulation, Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Seung Geun Song
- Department of Pathology, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Doo Hyun Chung
- Laboratory of Immune Regulation, Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Korea
- Department of Pathology, Seoul National University College of Medicine, Seoul 03080, Korea
| |
Collapse
|
14
|
Han S, Georgiev P, Ringel AE, Sharpe AH, Haigis MC. Age-associated remodeling of T cell immunity and metabolism. Cell Metab 2023; 35:36-55. [PMID: 36473467 PMCID: PMC10799654 DOI: 10.1016/j.cmet.2022.11.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 10/14/2022] [Accepted: 11/09/2022] [Indexed: 12/12/2022]
Abstract
Aging results in remodeling of T cell immunity and is associated with poor clinical outcomes in age-related diseases such as cancer. Among the hallmarks of aging, changes in host and cellular metabolism critically affect the development, maintenance, and function of T cells. Although metabolic perturbations impact anti-tumor T cell responses, the link between age-associated metabolic dysfunction and anti-tumor immunity remains unclear. In this review, we summarize recent advances in our understanding of aged T cell metabolism, with a focus on the bioenergetic and immunologic features of T cell subsets unique to the aging process. We also survey insights into mechanisms of metabolic T cell dysfunction in aging and discuss the impacts of aging on the efficacy of cancer immunotherapy. As the average life expectancy continues to increase, understanding the interplay between age-related metabolic reprogramming and maladaptive T cell immunity will be instrumental for the development of therapeutic strategies for older patients.
Collapse
Affiliation(s)
- SeongJun Han
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA; Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA; Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Peter Georgiev
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA; Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA; Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Alison E Ringel
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Arlene H Sharpe
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA; Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA.
| | - Marcia C Haigis
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
15
|
Semwal MK, Hester AK, Xiao Y, Udeaja C, Cepeda S, Verschelde JS, Jones N, Wedemeyer SA, Emtage S, Wimberly K, Griffith AV. Redox status regulates autophagy in thymic stromal cells and promotes T cell tolerance. Proc Natl Acad Sci U S A 2022; 119:e2204296119. [PMID: 36161925 PMCID: PMC9549397 DOI: 10.1073/pnas.2204296119] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 08/30/2022] [Indexed: 11/18/2022] Open
Abstract
Thymic stromal cells (TSCs) are critical regulators of T cell tolerance, but their basic biology has remained under-characterized because they are relatively rare and difficult to isolate. Recent work has revealed that constitutive autophagy in TSCs is required for self-antigen presentation and central T cell tolerance induction; however, the mechanisms regulating constitutive autophagy in TSCs are not well understood. Hydrogen peroxide has been shown to increase autophagy flux in other tissues, and we previously identified conspicuously low expression of the hydrogen peroxide-quenching enzyme catalase in TSCs. We investigated whether the redox status of TSCs established by low catalase expression regulates their basal autophagy levels and their capacity to impose central T cell tolerance. Transgenic overexpression of catalase diminished autophagy in TSCs and impaired thymocyte clonal deletion, concomitant with increased frequencies of spontaneous lymphocytic infiltrates in lung and liver and of serum antinuclear antigen reactivity. Effects on clonal deletion and autoimmune indicators were diminished in catalase transgenic mice when autophagy was rescued by expression of the Becn1F121A/F121A knock-in allele. These results suggest a metabolic mechanism by which the redox status of TSCs may regulate central T cell tolerance.
Collapse
Affiliation(s)
- Manpreet K. Semwal
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Long School of Medicine, UT Health San Antonio, San Antonio, TX 78229
- Barshop Institute for Longevity and Aging Studies, UT Health San Antonio, San Antonio, TX 78229
| | - Allison K. Hester
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Long School of Medicine, UT Health San Antonio, San Antonio, TX 78229
- Department of Medicine, Blood and Marrow Transplantation Division, Stanford University, Stanford, CA 94305
| | - Yangming Xiao
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Long School of Medicine, UT Health San Antonio, San Antonio, TX 78229
| | - Chioma Udeaja
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Long School of Medicine, UT Health San Antonio, San Antonio, TX 78229
| | - Sergio Cepeda
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - John S. Verschelde
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Long School of Medicine, UT Health San Antonio, San Antonio, TX 78229
| | - Nicholas Jones
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Long School of Medicine, UT Health San Antonio, San Antonio, TX 78229
| | - Sarah A. Wedemeyer
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Long School of Medicine, UT Health San Antonio, San Antonio, TX 78229
| | - Simon Emtage
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Long School of Medicine, UT Health San Antonio, San Antonio, TX 78229
| | - Kymberly Wimberly
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Long School of Medicine, UT Health San Antonio, San Antonio, TX 78229
| | - Ann V. Griffith
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Long School of Medicine, UT Health San Antonio, San Antonio, TX 78229
| |
Collapse
|
16
|
da Silva GB, Yamauchi MA, Bagatini MD. Oxidative stress in Hashimoto's thyroiditis: possible adjuvant therapies to attenuate deleterious effects. Mol Cell Biochem 2022; 478:949-966. [PMID: 36168075 DOI: 10.1007/s11010-022-04564-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Accepted: 09/15/2022] [Indexed: 11/30/2022]
Abstract
A number of studies have shown that oxidative stress is related to the pathogenesis of several immunological diseases, such as Hashimoto's thyroiditis (HT), although there is no plausible mechanism to explain it. Thus, we aimed at hypothesizing and providing some possible mechanisms linking oxidative stress to autoimmunity aspects and its implications for HT, as well as adjuvant therapeutic proposals to mitigate the deleterious effects. Our hypothesis is that deficient eating habits, autoimmune regulator gene predisposing gene, dysbiosis and molecular mimicry, unfolded proteins and stress in the endoplasmic reticulum, and thymus involution appear to be the main potential factors leading to HT oxidative stress. Likewise, we show that the use of minerals selenium and zinc, vitamins D and C, as well as probiotics, can be interesting adjuvant therapies for the control of oxidative damage and poor prognosis of HT. Further clinical trials are needed to understand the real beneficial and side effects of these supplements.
Collapse
Affiliation(s)
- Gilnei Bruno da Silva
- Post Graduate Program in Biomedical Sciences, Universidade Federal da Fronteira Sul, Chapecó, Santa Catarina, 89815-899, Brazil
| | - Milena Ayumi Yamauchi
- Post Graduate Program in Biomedical Sciences, Universidade Federal da Fronteira Sul, Chapecó, Santa Catarina, 89815-899, Brazil
| | - Margarete Dulce Bagatini
- Post Graduate Program in Biomedical Sciences, Universidade Federal da Fronteira Sul, Chapecó, Santa Catarina, 89815-899, Brazil.
| |
Collapse
|
17
|
Liang Z, Dong X, Zhang Z, Zhang Q, Zhao Y. Age-related thymic involution: Mechanisms and functional impact. Aging Cell 2022; 21:e13671. [PMID: 35822239 PMCID: PMC9381902 DOI: 10.1111/acel.13671] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 06/16/2022] [Accepted: 07/01/2022] [Indexed: 11/28/2022] Open
Abstract
The thymus is the primary immune organ responsible for generating self‐tolerant and immunocompetent T cells. However, the thymus gradually involutes during early life resulting in declined naïve T‐cell production, a process known as age‐related thymic involution. Thymic involution has many negative impacts on immune function including reduced pathogen resistance, high autoimmunity incidence, and attenuated tumor immunosurveillance. Age‐related thymic involution leads to a gradual reduction in thymic cellularity and thymic stromal microenvironment disruption, including loss of definite cortical‐medullary junctions, reduction of cortical thymic epithelial cells and medullary thymic epithelial cells, fibroblast expansion, and an increase in perivascular space. The compromised thymic microenvironment in aged individuals substantially disturbs thymocyte development and differentiation. Age‐related thymic involution is regulated by many transcription factors, micro RNAs, growth factors, cytokines, and other factors. In this review, we summarize the current understanding of age‐related thymic involution mechanisms and effects.
Collapse
Affiliation(s)
- Zhanfeng Liang
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China.,Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China.,Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
| | - Xue Dong
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Zhaoqi Zhang
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Qian Zhang
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Yong Zhao
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China.,Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China.,Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
18
|
Mohtashami M, Li YR, Lee CR, Zúñiga-Pflücker JC. Thymus Reconstitution in Young and Aged Mice Is Facilitated by In Vitro-Generated Progenitor T Cells. Front Immunol 2022; 13:926773. [PMID: 35874726 PMCID: PMC9304753 DOI: 10.3389/fimmu.2022.926773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Accepted: 06/13/2022] [Indexed: 11/13/2022] Open
Abstract
The prolonged lag in T cell recovery seen in older patients undergoing hematopoietic stem cell transplant (HSCT), after chemo-/radiotherapy, can lead to immune dysfunction. As a result, recovering patients may experience a relapse in malignancies and opportunistic infections, leading to high mortality rates. The delay in T cell recovery is partly due to thymic involution, a natural collapse in the size and function of the thymus, as individuals age, and partly due to the damage sustained by the thymic stromal cells through exposure to chemo-/radiotherapy. There is a clear need for new strategies to accelerate intrathymic T cell reconstitution when treating aged patients to counter the effects of involution and cancer therapy regimens. Adoptive transfer of human progenitor T (proT) cells has been shown to accelerate T cell regeneration in radiation-treated young mice and to restore thymic architecture in immunodeficient mice. Here, we demonstrate that the adoptive transfer of in vitro-generated proT cells in aged mice (18-24 months) accelerated thymic reconstitution after treatment with chemotherapy and gamma irradiation compared to HSCT alone. We noted that aged mice appeared to have a more limited expansion of CD4-CD8- thymocytes and slower temporal kinetics in the development of donor proT cells into mature T cells, when compared to younger mice, despite following the same chemo/radiation regimen. This suggests a greater resilience of the young thymus compared to the aged thymus. Nevertheless, newly generated T cells from proT cell engrafted aged and young mice were readily present in the periphery accelerating the reappearance of new naïve T cells. Accelerated T cell recovery was also observed in both aged and young mice receiving both proT cells and HSCT. The strategy of transferring proT cells can potentially be used as an effective cellular therapy in aged patients to improve immune recovery and reduce the risk of opportunistic infections post-HSCT.
Collapse
Affiliation(s)
| | - Yue Ru Li
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Christina R. Lee
- Biological Sciences, Sunnybrook Research Institute, Toronto, ON, Canada
| | - Juan Carlos Zúñiga-Pflücker
- Biological Sciences, Sunnybrook Research Institute, Toronto, ON, Canada
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
19
|
Cosway EJ, White AJ, Parnell SM, Schweighoffer E, Jolin HE, Bacon A, Rodewald HR, Tybulewicz V, McKenzie ANJ, Jenkinson WE, Anderson G. Eosinophils are an essential element of a type 2 immune axis that controls thymus regeneration. Sci Immunol 2022; 7:eabn3286. [PMID: 35275754 PMCID: PMC7612579 DOI: 10.1126/sciimmunol.abn3286] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Therapeutic interventions used for cancer treatment provoke thymus damage and limit the recovery of protective immunity. Here, we show that eosinophils are an essential part of an intrathymic type 2 immune network that enables thymus recovery after ablative therapy. Within hours of damage, the thymus undergoes CCR3-dependent colonization by peripheral eosinophils, which reestablishes the epithelial microenvironments that control thymopoiesis. Eosinophil regulation of thymus regeneration occurs via the concerted action of NKT cells that trigger CCL11 production via IL4 receptor signaling in thymic stroma, and ILC2 that represent an intrathymic source of IL5, a cytokine that therapeutically boosts thymus regeneration after damage. Collectively, our findings identify an intrathymic network composed of multiple innate immune cells that restores thymus function during reestablishment of the adaptive immune system.
Collapse
Affiliation(s)
- Emilie J. Cosway
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - Andrea J. White
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - Sonia M. Parnell
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | | | | | - Andrea Bacon
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - Hans-Reimer Rodewald
- Division of Cellular Immunology, German Cancer Research Center, 69120 Heidelberg, Germany
| | - Victor Tybulewicz
- Francis Crick Institute, London NW1 1AT, UK,Department of Immunology and Inflammation, Imperial College London, London W12 0NN, UK
| | | | - W. E. Jenkinson
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - Graham Anderson
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK,Correspondence to: Professor Graham Anderson, Institute for Immunology and Immunotherapy, Floor 4 Institute for Biomedical Research, Medical School, University of Birmingham, B15 2TT, United Kingdom. Tel: (44)1214146817.
| |
Collapse
|
20
|
Hester AK, Semwal MK, Cepeda S, Xiao Y, Rueda M, Wimberly K, Venables T, Dileepan T, Kraig E, Griffith AV. Redox regulation of age-associated defects in generation and maintenance of T cell self-tolerance and immunity to foreign antigens. Cell Rep 2022; 38:110363. [PMID: 35172147 PMCID: PMC8898380 DOI: 10.1016/j.celrep.2022.110363] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 11/22/2021] [Accepted: 01/19/2022] [Indexed: 12/20/2022] Open
Abstract
Thymic atrophy reduces naive T cell production and contributes to increased susceptibility to viral infection with age. Expression of tissue-restricted antigen (TRA) genes also declines with age and has been thought to increase autoimmune disease susceptibility. We find that diminished expression of a model TRA gene in aged thymic stromal cells correlates with impaired clonal deletion of cognate T cells recognizing an autoantigen involved in atherosclerosis. Clonal deletion in the polyclonal thymocyte population is also perturbed. Distinct age-associated defects in the generation of antigen-specific T cells include a conspicuous decline in generation of T cells recognizing an immunodominant influenza epitope. Increased catalase activity delays thymic atrophy, and here, we show that it mitigates declining production of influenza-specific T cells and their frequency in lung after infection, but does not reverse declines in TRA expression or efficient negative selection. These results reveal important considerations for strategies to restore thymic function.
Collapse
Affiliation(s)
- Allison K Hester
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Long School of Medicine, UT Health San Antonio, San Antonio, TX 78229, USA
| | - Manpreet K Semwal
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Long School of Medicine, UT Health San Antonio, San Antonio, TX 78229, USA
| | - Sergio Cepeda
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Long School of Medicine, UT Health San Antonio, San Antonio, TX 78229, USA
| | - Yangming Xiao
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Long School of Medicine, UT Health San Antonio, San Antonio, TX 78229, USA
| | - Meghan Rueda
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Long School of Medicine, UT Health San Antonio, San Antonio, TX 78229, USA
| | - Kymberly Wimberly
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Long School of Medicine, UT Health San Antonio, San Antonio, TX 78229, USA
| | | | - Thamotharampillai Dileepan
- Center for Immunology, University of Minnesota Medical School, Minneapolis, MN 55455, USA; Department of Microbiology, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Ellen Kraig
- Department of Cell Systems and Anatomy, University of Texas Long School of Medicine, UT Health San Antonio, San Antonio, TX 78229, USA
| | - Ann V Griffith
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Long School of Medicine, UT Health San Antonio, San Antonio, TX 78229, USA.
| |
Collapse
|
21
|
Duah M, Li L, Shen J, Lan Q, Pan B, Xu K. Thymus Degeneration and Regeneration. Front Immunol 2021; 12:706244. [PMID: 34539637 PMCID: PMC8442952 DOI: 10.3389/fimmu.2021.706244] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Accepted: 08/16/2021] [Indexed: 01/08/2023] Open
Abstract
The immune system’s ability to resist the invasion of foreign pathogens and the tolerance to self-antigens are primarily centered on the efficient functions of the various subsets of T lymphocytes. As the primary organ of thymopoiesis, the thymus performs a crucial role in generating a self-tolerant but diverse repertoire of T cell receptors and peripheral T cell pool, with the capacity to recognize a wide variety of antigens and for the surveillance of malignancies. However, cells in the thymus are fragile and sensitive to changes in the external environment and acute insults such as infections, chemo- and radiation-therapy, resulting in thymic injury and degeneration. Though the thymus has the capacity to self-regenerate, it is often insufficient to reconstitute an intact thymic function. Thymic dysfunction leads to an increased risk of opportunistic infections, tumor relapse, autoimmunity, and adverse clinical outcome. Thus, exploiting the mechanism of thymic regeneration would provide new therapeutic options for these settings. This review summarizes the thymus’s development, factors causing thymic injury, and the strategies for improving thymus regeneration.
Collapse
Affiliation(s)
- Maxwell Duah
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou Medical University, Xuzhou, China.,Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China
| | - Lingling Li
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou Medical University, Xuzhou, China.,Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China
| | - Jingyi Shen
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou Medical University, Xuzhou, China.,Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China
| | - Qiu Lan
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou Medical University, Xuzhou, China.,Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China
| | - Bin Pan
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou Medical University, Xuzhou, China.,Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China
| | - Kailin Xu
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou Medical University, Xuzhou, China.,Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
22
|
Ducloux D, Legendre M, Bamoulid J, Saas P, Courivaud C, Crepin T. End-Stage Renal Disease-Related Accelerated Immune Senescence: Is Rejuvenation of the Immune System a Therapeutic Goal? Front Med (Lausanne) 2021; 8:720402. [PMID: 34540869 PMCID: PMC8446427 DOI: 10.3389/fmed.2021.720402] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 08/17/2021] [Indexed: 02/05/2023] Open
Abstract
End-stage renal disease (ESRD) patients exhibit clinical features of premature ageing, including frailty, cardiovascular disease, and muscle wasting. Accelerated ageing also concerns the immune system. Patients with ESRD have both immune senescence and chronic inflammation that are resumed in the so-called inflammaging syndrome. Immune senescence is particularly characterised by premature loss of thymic function that is associated with hyporesponsiveness to vaccines, susceptibility to infections, and death. ESRD-related chronic inflammation has multiple causes and participates to accelerated cardiovascular disease. Although, both characterisation of immune senescence and its consequences are relatively well-known, mechanisms are more uncertain. However, prevention of immune senescence/inflammation or/and rejuvenation of the immune system are major goal to ameliorate clinical outcomes of ESRD patients.
Collapse
Affiliation(s)
- Didier Ducloux
- Inserm, UMR1098, Federation Hospitalo-Universitaire INCREASE, Besançon, France.,University Bourgogne Franche-Comté, Faculté de Médecine et de Pharmacie, LabEx LipSTIC, Besançon, France.,Structure Fédérative de Recherche, SFR FED4234, Besançon, France.,CHU Besançon, Department of Nephrology, Dialysis, and Renal Transplantation, Besançon, France
| | - Mathieu Legendre
- Inserm, UMR1098, Federation Hospitalo-Universitaire INCREASE, Besançon, France.,University Bourgogne Franche-Comté, Faculté de Médecine et de Pharmacie, LabEx LipSTIC, Besançon, France
| | - Jamal Bamoulid
- Inserm, UMR1098, Federation Hospitalo-Universitaire INCREASE, Besançon, France.,University Bourgogne Franche-Comté, Faculté de Médecine et de Pharmacie, LabEx LipSTIC, Besançon, France.,Structure Fédérative de Recherche, SFR FED4234, Besançon, France.,CHU Besançon, Department of Nephrology, Dialysis, and Renal Transplantation, Besançon, France
| | - Philippe Saas
- Inserm, UMR1098, Federation Hospitalo-Universitaire INCREASE, Besançon, France.,University Bourgogne Franche-Comté, Faculté de Médecine et de Pharmacie, LabEx LipSTIC, Besançon, France.,Structure Fédérative de Recherche, SFR FED4234, Besançon, France.,EFS Bourgogne Franche-Comté, Plateforme de Biomonitoring, CIC 1431/UMR1098, Besançon, France
| | - Cécile Courivaud
- Inserm, UMR1098, Federation Hospitalo-Universitaire INCREASE, Besançon, France.,University Bourgogne Franche-Comté, Faculté de Médecine et de Pharmacie, LabEx LipSTIC, Besançon, France.,Structure Fédérative de Recherche, SFR FED4234, Besançon, France.,CHU Besançon, Department of Nephrology, Dialysis, and Renal Transplantation, Besançon, France
| | - Thomas Crepin
- Inserm, UMR1098, Federation Hospitalo-Universitaire INCREASE, Besançon, France.,CHU Besançon, Department of Nephrology, Dialysis, and Renal Transplantation, Besançon, France
| |
Collapse
|
23
|
Budamagunta V, Foster TC, Zhou D. Cellular senescence in lymphoid organs and immunosenescence. Aging (Albany NY) 2021; 13:19920-19941. [PMID: 34382946 PMCID: PMC8386533 DOI: 10.18632/aging.203405] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Accepted: 08/02/2021] [Indexed: 02/07/2023]
Abstract
Immunosenescence is a multi-faceted phenomenon at the root of age-associated immune dysfunction. It can lead to an array of pathological conditions, including but not limited to a decreased capability to surveil and clear senescent cells (SnCs) and cancerous cells, an increased autoimmune responses leading to tissue damage, a reduced ability to tackle pathogens, and a decreased competence to illicit a robust response to vaccination. Cellular senescence is a phenomenon by which oncogene-activated, stressed or damaged cells undergo a stable cell cycle arrest. Failure to efficiently clear SnCs results in their accumulation in an organism as it ages. SnCs actively secrete a myriad of molecules, collectively called senescence-associated secretory phenotype (SASP), which are factors that cause dysfunction in the neighboring tissue. Though both cellular senescence and immunosenescence have been studied extensively and implicated in various pathologies, their relationship has not been greatly explored. In the wake of an ongoing pandemic (COVID-19) that disproportionately affects the elderly, immunosenescence as a function of age has become a topic of great importance. The goal of this review is to explore the role of cellular senescence in age-associated lymphoid organ dysfunction and immunosenescence, and provide a framework to explore therapies to rejuvenate the aged immune system.
Collapse
Affiliation(s)
- Vivekananda Budamagunta
- Genetics and Genomics Graduate Program, Genetics Institute, College of Medicine, University of Florida, Gainesville, FL 32610, USA.,Department of Neuroscience, McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, FL 32610, USA.,Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL 32610, USA
| | - Thomas C Foster
- Genetics and Genomics Graduate Program, Genetics Institute, College of Medicine, University of Florida, Gainesville, FL 32610, USA.,Department of Neuroscience, McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Daohong Zhou
- Genetics and Genomics Graduate Program, Genetics Institute, College of Medicine, University of Florida, Gainesville, FL 32610, USA.,Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL 32610, USA
| |
Collapse
|
24
|
Reis M, Willis GR, Fernandez-Gonzalez A, Yeung V, Taglauer E, Magaletta M, Parsons T, Derr A, Liu X, Maehr R, Kourembanas S, Mitsialis SA. Mesenchymal Stromal Cell-Derived Extracellular Vesicles Restore Thymic Architecture and T Cell Function Disrupted by Neonatal Hyperoxia. Front Immunol 2021; 12:640595. [PMID: 33936055 PMCID: PMC8082426 DOI: 10.3389/fimmu.2021.640595] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 03/24/2021] [Indexed: 11/28/2022] Open
Abstract
Treating premature infants with high oxygen is a routine intervention in the context of neonatal intensive care. Unfortunately, the increase in survival rates is associated with various detrimental sequalae of hyperoxia exposure, most notably bronchopulmonary dysplasia (BPD), a disease of disrupted lung development. The effects of high oxygen exposure on other developing organs of the infant, as well as the possible impact such disrupted development may have on later life remain poorly understood. Using a neonatal mouse model to investigate the effects of hyperoxia on the immature immune system we observed a dramatic involution of the thymic medulla, and this lesion was associated with disrupted FoxP3+ regulatory T cell generation and T cell autoreactivity. Significantly, administration of mesenchymal stromal cell-derived extracellular vesicles (MEx) restored thymic medullary architecture and physiological thymocyte profiles. Using single cell transcriptomics, we further demonstrated preferential impact of MEx treatment on the thymic medullary antigen presentation axis, as evidenced by enrichment of antigen presentation and antioxidative-stress related genes in dendritic cells (DCs) and medullary epithelial cells (mTECs). Our study demonstrates that MEx treatment represents a promising restorative therapeutic approach for oxygen-induced thymic injury, thus promoting normal development of both central tolerance and adaptive immunity.
Collapse
Affiliation(s)
- Monica Reis
- Division of Newborn Medicine & Department of Pediatrics, Boston Children's Hospital, Boston, MA, United States.,Department of Pediatrics, Harvard Medical School, Boston, MA, United States
| | - Gareth R Willis
- Division of Newborn Medicine & Department of Pediatrics, Boston Children's Hospital, Boston, MA, United States.,Department of Pediatrics, Harvard Medical School, Boston, MA, United States
| | - Angeles Fernandez-Gonzalez
- Division of Newborn Medicine & Department of Pediatrics, Boston Children's Hospital, Boston, MA, United States.,Department of Pediatrics, Harvard Medical School, Boston, MA, United States
| | - Vincent Yeung
- Division of Newborn Medicine & Department of Pediatrics, Boston Children's Hospital, Boston, MA, United States.,Department of Pediatrics, Harvard Medical School, Boston, MA, United States
| | - Elizabeth Taglauer
- Division of Newborn Medicine & Department of Pediatrics, Boston Children's Hospital, Boston, MA, United States.,Department of Pediatrics, Harvard Medical School, Boston, MA, United States
| | - Margaret Magaletta
- Program in Molecular Medicine, Diabetes Center of Excellence, University of Massachusetts Medical School, Worcester, MA, United States
| | - Teagan Parsons
- Program in Molecular Medicine, Diabetes Center of Excellence, University of Massachusetts Medical School, Worcester, MA, United States
| | - Alan Derr
- Program in Molecular Medicine, Diabetes Center of Excellence, University of Massachusetts Medical School, Worcester, MA, United States
| | - Xianlan Liu
- Division of Newborn Medicine & Department of Pediatrics, Boston Children's Hospital, Boston, MA, United States.,Department of Pediatrics, Harvard Medical School, Boston, MA, United States
| | - Rene Maehr
- Program in Molecular Medicine, Diabetes Center of Excellence, University of Massachusetts Medical School, Worcester, MA, United States
| | - Stella Kourembanas
- Division of Newborn Medicine & Department of Pediatrics, Boston Children's Hospital, Boston, MA, United States.,Department of Pediatrics, Harvard Medical School, Boston, MA, United States
| | - S Alex Mitsialis
- Division of Newborn Medicine & Department of Pediatrics, Boston Children's Hospital, Boston, MA, United States.,Department of Pediatrics, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
25
|
Han J, Zúñiga-Pflücker JC. High-Oxygen Submersion Fetal Thymus Organ Cultures Enable FOXN1-Dependent and -Independent Support of T Lymphopoiesis. Front Immunol 2021; 12:652665. [PMID: 33859647 PMCID: PMC8043069 DOI: 10.3389/fimmu.2021.652665] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 03/15/2021] [Indexed: 01/19/2023] Open
Abstract
T cell development is effectively supported in fetal thymus organ cultures (FTOCs), which places thymus lobes atop an air-liquid interface (ALI) culture system. The direct exposure to air is critical for its success, as fetal thymus lobes placed in low oxygen submersion (LOS)-FTOCs fail to support thymocyte development. However, submersion cultures performed in the presence of high concentration of ambient oxygen (60~80%) allow for normal thymocyte development, but the underlying mechanism for this rescue has remained elusive. Here, we show that FOXN1 expression in thymic epithelial cells (TECs) from LOS-FTOCs was greatly reduced compared to conventional ALI-FTOCs. Consequently, the expression of important FOXN1 target genes, including Dll4 and Ccl25, in TECs was extinguished. The loss of DLL4 and CCL25 interrupted thymocyte differentiation and led to CD4+CD8+ cells exiting the lobes, respectively. High oxygen submersion (HOS)-FTOCs restored the expression of FOXN1 and its target genes, as well as maintained high levels of MHCII expression in TECs. In addition, HOS-FTOCs promoted the self-renewal of CD4−CD8−CD44−CD25+ cells, allowing for the continuous generation of later stage thymocytes. Forced FOXN1 expression in TECs rescued thymocyte developmental progression, but not cellularity, in LOS-FTOCs. Given that oxidative stress has been reported to accelerate the onset of age-associated thymic involution, we postulate that regulation of FOXN1 by oxygen and antioxidants may underpin this biological process.
Collapse
Affiliation(s)
- Jianxun Han
- Department of Biological Sciences, Sunnybrook Research Institute, Toronto, ON, Canada
| | - Juan Carlos Zúñiga-Pflücker
- Department of Biological Sciences, Sunnybrook Research Institute, Toronto, ON, Canada.,Department of Immunology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
26
|
Semwal MK, Jones NE, Griffith AV. Metabolic Regulation of Thymic Epithelial Cell Function. Front Immunol 2021; 12:636072. [PMID: 33746975 PMCID: PMC7968369 DOI: 10.3389/fimmu.2021.636072] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 02/09/2021] [Indexed: 12/28/2022] Open
Abstract
The thymus is the primary site of T lymphocyte development, where mutually inductive signaling between lymphoid progenitors and thymic stromal cells directs the progenitors along a well-characterized program of differentiation. Although thymic stromal cells, including thymic epithelial cells (TECs) are critical for the development of T cell-mediated immunity, many aspects of their basic biology have been difficult to resolve because they represent a small fraction of thymus cellularity, and because their isolation requires enzymatic digestion that induces broad physiological changes. These obstacles are especially relevant to the study of metabolic regulation of cell function, since isolation procedures necessarily disrupt metabolic homeostasis. In contrast to the well-characterized relationships between metabolism and intracellular signaling in T cell function during an immune response, metabolic regulation of thymic stromal cell function represents an emerging area of study. Here, we review recent advances in three distinct, but interconnected areas: regulation of mTOR signaling, reactive oxygen species (ROS), and autophagy, with respect to their roles in the establishment and maintenance of the thymic stromal microenvironment.
Collapse
Affiliation(s)
- Manpreet K Semwal
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Joe R. and Teresa Lozano Long School of Medicine, UT Health San Antonio, San Antonio, TX, United States
| | - Nicholas E Jones
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Joe R. and Teresa Lozano Long School of Medicine, UT Health San Antonio, San Antonio, TX, United States
| | - Ann V Griffith
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Joe R. and Teresa Lozano Long School of Medicine, UT Health San Antonio, San Antonio, TX, United States
| |
Collapse
|
27
|
Du H, Wang Y, Liu X, Wang S, Wu S, Yuan Z, Zhu X. miRNA-146a-5p mitigates stress-induced premature senescence of D-galactose-induced primary thymic stromal cells. Cytokine 2021; 137:155314. [PMID: 33002743 DOI: 10.1016/j.cyto.2020.155314] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 09/08/2020] [Accepted: 09/22/2020] [Indexed: 12/12/2022]
Abstract
Senescent thymic stromal cells (TSCs) producing senescence-associated secretory phenotype (SASP) may play a role at later phases of thymic involution. However, the etiology and mechanisms responsible for TSC senescence remain to be elucidated. In the present study, the effects of oxidative stress on TSCs and role of miRNA-146a-5p in stress-induced premature senescence (SIPS) were identified. D-galactose (D-gal) induced oxidative stress in primary TSCs and a limited cumulative oxidative stress induced premature senescence but not apoptosis of TSCs. miRNA-146a-5p overexpression can mitigate the SIPS by targeting tumor necrosis factor receptor-associated factor 6 (TRAF6) instead of increasing autophagy clearance. Furthermore, exogenous miRNA-146a-5p reversed the upregulation of chemokines including Cxcl5, pro-inflammatory cytokines, and antimicrobial peptides in TSCs with SIPS. In conclusion, the accumulated oxidative stress may be partially responsible for senescence in TSCs and modulation of miRNA-146a-5p may attenuate this process.
Collapse
Affiliation(s)
- Hongmei Du
- Research Center, Shengjing Hospital of China Medical University, 7 Mulan Road, Economic Development Zone, Benxi, China; Department of Oncology, Shengjing Hospital of China Medical University, 39 Huaxiang Road, Shenyang, China
| | - Yajun Wang
- Research Center, Shengjing Hospital of China Medical University, 7 Mulan Road, Economic Development Zone, Benxi, China
| | - Xue Liu
- Research Center, Shengjing Hospital of China Medical University, 7 Mulan Road, Economic Development Zone, Benxi, China
| | - Siliang Wang
- Department of Oncology, Shengjing Hospital of China Medical University, 39 Huaxiang Road, Shenyang, China
| | - Simeng Wu
- Department of Blood Transfusion, Shengjing Hospital of China Medical University, 39 Huaxiang Road, Shenyang, China
| | - Zhe Yuan
- Department of Blood Transfusion, Shengjing Hospital of China Medical University, 39 Huaxiang Road, Shenyang, China
| | - Xike Zhu
- Research Center, Shengjing Hospital of China Medical University, 7 Mulan Road, Economic Development Zone, Benxi, China.
| |
Collapse
|
28
|
The Teleost Thymus in Health and Disease: New Insights from Transcriptomic and Histopathological Analyses of Turbot, Scophthalmus maximus. BIOLOGY 2020; 9:biology9080221. [PMID: 32823553 PMCID: PMC7465915 DOI: 10.3390/biology9080221] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 08/08/2020] [Accepted: 08/11/2020] [Indexed: 02/07/2023]
Abstract
The thymus is a primary lymphoid organ that plays a pivotal role in the adaptive immune system. The immunobiology of the thymus in fish is considered to be similar to that of mammals, but it is actually poorly characterized in several cultured teleost species. In particular, while investigations in human and veterinary medicine have highlighted that the thymus can be affected by different pathological conditions, little is known about its response during disease in fish. To better understand the role of the thymus under physiological and pathological conditions, we conducted a study in turbot (Scophthalmus maximus), a commercially valuable flatfish species, combining transcriptomic and histopathological analyses. The myxozoan parasite Enteromyxum scophthalmi, which represents a major challenge to turbot production, was used as a model of infection. The thymus tissues of healthy fish showed overrepresented functions related to its immunological role in T-cell development and maturation. Large differences were observed between the transcriptomes of control and severely infected fish. Evidence of inflammatory response, apoptosis modulation, and declined thymic function associated with loss of cellularity was revealed by both genomic and morphopathological analyses. This study presents the first description of the turbot thymus transcriptome and provides novel insights into the role of this organ in teleosts’ immune responses.
Collapse
|
29
|
Blood autophagy defect causes accelerated non-hematopoietic organ aging. Aging (Albany NY) 2020; 11:4910-4922. [PMID: 31327762 PMCID: PMC6682532 DOI: 10.18632/aging.102086] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2019] [Accepted: 07/01/2019] [Indexed: 02/06/2023]
Abstract
Autophagy has been well studied in regulating aging; however, the impact of autophagy in one organ on the aging of other organs has not been documented. In this study, we used a mouse model with deletion of an autophagy-essential gene Atg7 in hematopoietic system to evaluate the intrinsic role of hematopoietic autophagy on the aging of non-hematopoietic organs. We found that autophagy defect in hematopoietic system causes growth retardation and shortened lifespan, along with aging-like phenotypes including hypertrophic heart, lung and spleen, but atrophic thymus and reduced bone mineral density at organismal level. Hematopoietic autophagy defect also causes increased oxidative stress and mitochondrial mass or aging gene expression at cellular level in multiple non-hematopoietic organs. The organ aging in the Atg7-deleted mice was reversed by anatomic connection to wild-type mice with intact blood autophagy via parabiosis, but not by injection of blood cell-free plasma. Our finding thus highlights an essential role of hematopoietic autophagy for decelerating aging in non-hematopoietic organs.
Collapse
|
30
|
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: 12] [Impact Index Per Article: 3.0] [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
|
31
|
Implications of Oxidative Stress and Cellular Senescence in Age-Related Thymus Involution. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:7986071. [PMID: 32089780 PMCID: PMC7025075 DOI: 10.1155/2020/7986071] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 01/20/2020] [Accepted: 01/23/2020] [Indexed: 02/07/2023]
Abstract
The human thymus is a primary lymphoepithelial organ which supports the production of self-tolerant T cells with competent and regulatory functions. Paradoxically, despite the crucial role that it exerts in T cell-mediated immunity and prevention of systemic autoimmunity, the thymus is the first organ of the body that exhibits age-associated degeneration/regression, termed “thymic involution.” A hallmark of this early phenomenon is a progressive decline of thymic mass as well as a decreased output of naïve T cells, thus resulting in impaired immune response. Importantly, thymic involution has been recently linked with cellular senescence which is a stress response induced by various stimuli. Accumulation of senescent cells in tissues has been implicated in aging and a plethora of age-related diseases. In addition, several lines of evidence indicate that oxidative stress, a well-established trigger of senescence, is also involved in thymic involution, thus highlighting a possible interplay between oxidative stress, senescence, and thymic involution.
Collapse
|
32
|
Dynamic changes in epithelial cell morphology control thymic organ size during atrophy and regeneration. Nat Commun 2019; 10:4402. [PMID: 31562306 PMCID: PMC6765001 DOI: 10.1038/s41467-019-11879-2] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Accepted: 08/07/2019] [Indexed: 12/31/2022] Open
Abstract
T lymphocytes must be produced throughout life, yet the thymus, where T lymphocytes are made, exhibits accelerated atrophy with age. Even in advanced atrophy, however, the thymus remains plastic, and can be regenerated by appropriate stimuli. Logically, thymic atrophy is thought to reflect senescent cell death, while regeneration requires proliferation of stem or progenitor cells, although evidence is scarce. Here we use conditional reporters to show that accelerated thymic atrophy reflects contraction of complex cell projections unique to cortical epithelial cells, while regeneration requires their regrowth. Both atrophy and regeneration are independent of changes in epithelial cell number, suggesting that the size of the thymus is regulated primarily by rate-limiting morphological changes in cortical stroma, rather than by their cell death or proliferation. Our data also suggest that cortical epithelial morphology is under the control of medullary stromal signals, revealing a previously unrecognized endocrine-paracrine signaling axis in the thymus.
Collapse
|
33
|
Du HM, Wang YJ, Liu X, Wang SL, Wu SM, Yuan Z, Zhu XK. Defective Central Immune Tolerance Induced by High-Dose D-Galactose Resembles Aging. BIOCHEMISTRY (MOSCOW) 2019; 84:617-626. [DOI: 10.1134/s000629791906004x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
34
|
Nacka-Aleksić M, Pilipović I, Kotur-Stevuljević J, Petrović R, Sopta J, Leposavić G. Sexual dimorphism in rat thymic involution: a correlation with thymic oxidative status and inflammation. Biogerontology 2019; 20:545-569. [DOI: 10.1007/s10522-019-09816-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Accepted: 05/12/2019] [Indexed: 01/05/2023]
|
35
|
RNA-Seq-Based Gene Expression Pattern and Morphological Alterations in Chick Thymus during Postnatal Development. Int J Genomics 2019; 2019:6905194. [PMID: 31179312 PMCID: PMC6501151 DOI: 10.1155/2019/6905194] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2018] [Revised: 12/20/2018] [Accepted: 02/25/2019] [Indexed: 12/23/2022] Open
Abstract
The thymus is a lobulated unique lymphoid immune organ that plays a critical role in the selection, development, proliferation, and differentiation of T cells. The thymus of developing chickens undergoes continued morphological alterations; however, the biomolecular and transcriptional dynamics of the postnatal thymus in avian species is not clear yet. Therefore, the thymuses from chickens at different stages of development (at weeks 0, 1, 5, 9, 18, and 27) were used in the present study. The RNA-seq method was used to study the gene expression patterns. On average, 24120819 clean reads were mapped, differentially expressed genes (DEGs) were identified on the basis of log values (fold change), including 744 upregulated and 425 downregulated genes. The expression pattern revealed by RNA-seq was validated by quantitative real-time PCR (qPCR) analysis of four important genes, which are PCNA, CCNA2, CCNB2, and CDK1. Thus, the current study revealed that during postnatal development, the thymus undergoes severe atrophy. Thymus structure was damaged and gene expression changed dramatically, especially at the 27th week of age. Moreover, we found significant changes of several signaling pathways such as the cytokine-cytokine receptor interaction and cell cycle signaling pathways. Hence, it may be inferred that those signaling pathways might be closely related to the postnatal chicken thymus development.
Collapse
|
36
|
Cepeda S, Griffith AV. Thymic stromal cells: Roles in atrophy and age-associated dysfunction of the thymus. Exp Gerontol 2018; 105:113-117. [PMID: 29278750 PMCID: PMC5869099 DOI: 10.1016/j.exger.2017.12.022] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2017] [Revised: 12/21/2017] [Accepted: 12/22/2017] [Indexed: 11/25/2022]
Abstract
Atrophy of the thymus, the primary site of T lymphocyte generation, is a hallmark of the aging immune system. Age-associated thymic atrophy results in diminished output of new, naïve T cells, with immune sequelae that include diminished responses to novel pathogenic challenge and vaccines, as well as diminished tumor surveillance. Although a variety of stimuli are known to regulate transient thymic atrophy, mechanisms governing progressive age-associated atrophy have been difficult to resolve. This has been due in part to the fact that one of the primary targets of age-associated thymic atrophy is a relatively rare population, thymic stromal cells. This review focuses on changes in thymic stromal cells during aging and on the contributions of periodic, stochastic, and progressive causes of thymic atrophy.
Collapse
Affiliation(s)
- Sergio Cepeda
- Microbiology, Immunology, and Molecular Genetics, School of Medicine, UT Health San Antonio, San Antonio, TX, United States
| | - Ann V Griffith
- Microbiology, Immunology, and Molecular Genetics, School of Medicine, UT Health San Antonio, San Antonio, TX, United States.
| |
Collapse
|
37
|
Skulachev MV, Skulachev VP. Programmed aging of mammals: Proof of concept and prospects of biochemical approaches for anti-aging therapy. BIOCHEMISTRY (MOSCOW) 2017; 82:1403-1422. [DOI: 10.1134/s000629791712001x] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
38
|
Majumdar S, Nandi D. Thymic Atrophy: Experimental Studies and Therapeutic Interventions. Scand J Immunol 2017; 87:4-14. [PMID: 28960415 DOI: 10.1111/sji.12618] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Revised: 09/01/2017] [Accepted: 09/25/2017] [Indexed: 12/13/2022]
Abstract
The thymus is essential for T cell development and maturation. It is extremely sensitive to atrophy, wherein loss in cellularity of the thymus and/or disruption of the thymic architecture occur. This may lead to lower naïve T cell output and limited TCR diversity. Thymic atrophy is often associated with ageing. What is less appreciated is that proper functioning of the thymus is critical for reduction in morbidity and mortality associated with various clinical conditions including infections and transplantation. Therefore, therapeutic interventions which possess thymopoietic potential and lower thymic atrophy are required. These treatments enhance thymic output, which is a vital factor in generating favourable outcomes in clinical conditions. In this review, experimental studies on thymic atrophy in rodents and clinical cases where the thymus atrophies are discussed. In addition, mechanisms leading to thymic atrophy during ageing as well as during various stress conditions are reviewed. Therapies such as zinc supplementation, IL7 administration, leptin treatment, keratinocyte growth factor administration and sex steroid ablation during thymic atrophy involving experiments in animals and various clinical scenarios are reviewed. Interventions that have been used across different scenarios to reduce the extent of thymic atrophy and enhance its output are discussed. This review aims to speculate on the roles of combination therapies, which by acting additively or synergistically may further alleviate thymic atrophy and boost its function, thereby strengthening cellular T cell responses.
Collapse
Affiliation(s)
- S Majumdar
- Department of Biochemistry & Centre for Infectious Diseases Research, Indian Institute of Science, Bangalore, India
| | - D Nandi
- Department of Biochemistry & Centre for Infectious Diseases Research, Indian Institute of Science, Bangalore, India
| |
Collapse
|
39
|
Yan F, Mo X, Liu J, Ye S, Zeng X, Chen D. Thymic function in the regulation of T cells, and molecular mechanisms underlying the modulation of cytokines and stress signaling (Review). Mol Med Rep 2017; 16:7175-7184. [PMID: 28944829 PMCID: PMC5865843 DOI: 10.3892/mmr.2017.7525] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Accepted: 05/12/2017] [Indexed: 01/08/2023] Open
Abstract
The thymus is critical in establishing and maintaining the appropriate microenvironment for promoting the development and selection of T cells. The function and structure of the thymus gland has been extensively studied, particularly as the thymus serves an important physiological role in the lymphatic system. Numerous studies have investigated the morphological features of thymic involution. Recently, research attention has increasingly been focused on thymic proteins as targets for drug intervention. Omics approaches have yielded novel insights into the thymus and possible drug targets. The present review addresses the signaling and transcriptional functions of the thymus, including the molecular mechanisms underlying the regulatory functions of T cells and their role in the immune system. In addition, the levels of cytokines secreted in the thymus have a significant effect on thymic functions, including thymocyte migration and development, thymic atrophy and thymic recovery. Furthermore, the regulation and molecular mechanisms of stress-mediated thymic atrophy and involution were investigated, with particular emphasis on thymic function as a potential target for drug development and discovery using proteomics.
Collapse
Affiliation(s)
- Fenggen Yan
- Department of Dermatology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong 510120, P.R. China
| | - Xiumei Mo
- Department of Dermatology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong 510120, P.R. China
| | - Junfeng Liu
- Department of Dermatology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong 510120, P.R. China
| | - Siqi Ye
- Department of Dermatology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong 510120, P.R. China
| | - Xing Zeng
- Department of Dermatology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong 510120, P.R. China
| | - Dacan Chen
- Department of Dermatology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong 510120, P.R. China
| |
Collapse
|
40
|
Abstract
Intrathymic T cell development is a complex process that depends upon continuous guidance from thymus stromal cell microenvironments. The thymic epithelium within the thymic stroma comprises highly specialized cells with a high degree of anatomic, phenotypic, and functional heterogeneity. These properties are collectively required to bias thymocyte development toward production of self-tolerant and functionally competent T cells. The importance of thymic epithelial cells (TECs) is evidenced by clear links between their dysfunction and multiple diseases where autoimmunity and immunodeficiency are major components. Consequently, TECs are an attractive target for cell therapies to restore effective immune system function. The pathways and molecular regulators that control TEC development are becoming clearer, as are their influences on particular stages of T cell development. Here, we review both historical and the most recent advances in our understanding of the cellular and molecular mechanisms controlling TEC development, function, dysfunction, and regeneration.
Collapse
Affiliation(s)
- Jakub Abramson
- Department of Immunology, Weizmann Institute of Science, Rehovot 76100, Israel;
| | - Graham Anderson
- MRC Centre for Immune Regulation, Institute for Immunology and Immunotherapy, University of Birmingham, Birmingham B15 2TT, United Kingdom;
| |
Collapse
|
41
|
Skulachev VP, Holtze S, Vyssokikh MY, Bakeeva LE, Skulachev MV, Markov AV, Hildebrandt TB, Sadovnichii VA. Neoteny, Prolongation of Youth: From Naked Mole Rats to “Naked Apes” (Humans). Physiol Rev 2017; 97:699-720. [DOI: 10.1152/physrev.00040.2015] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
It has been suggested that highly social mammals, such as naked mole rats and humans, are long-lived due to neoteny (the prolongation of youth). In both species, aging cannot operate as a mechanism facilitating natural selection because the pressure of this selection is strongly reduced due to 1) a specific social structure where only the “queen” and her “husband(s)” are involved in reproduction (naked mole rats) or 2) substituting fast technological progress for slow biological evolution (humans). Lists of numerous traits of youth that do not disappear with age in naked mole rats and humans are presented and discussed. A high resistance of naked mole rats to cancer, diabetes, cardiovascular and brain diseases, and many infections explains why their mortality rate is very low and almost age-independent and why their lifespan is more than 30 years, versus 3 years in mice. In young humans, curves of mortality versus age start at extremely low values. However, in the elderly, human mortality strongly increases. High mortality rates in other primates are observed at much younger ages than in humans. The inhibition of the aging process in humans by specific drugs seems to be a promising approach to prolong our healthspan. This might be a way to retard aging, which is already partially accomplished via the natural physiological phenomenon neoteny.
Collapse
Affiliation(s)
- Vladimir P. Skulachev
- Lomonosov Moscow State University, Belozersky Institute of Physico-Chemical Biology, Moscow, Russia; Lomonosov Moscow State University, Institute of Mitoengineering, Moscow, Russia; Leibniz Institute for Zoo and Wildlife Research, Berlin, Germany; Lomonosov Moscow State University, Biological Faculty, Moscow, Russia; Lomonosov Moscow State University, Faculty of Mechanics and Mathematics, Moscow, Russia
| | - Susanne Holtze
- Lomonosov Moscow State University, Belozersky Institute of Physico-Chemical Biology, Moscow, Russia; Lomonosov Moscow State University, Institute of Mitoengineering, Moscow, Russia; Leibniz Institute for Zoo and Wildlife Research, Berlin, Germany; Lomonosov Moscow State University, Biological Faculty, Moscow, Russia; Lomonosov Moscow State University, Faculty of Mechanics and Mathematics, Moscow, Russia
| | - Mikhail Y. Vyssokikh
- Lomonosov Moscow State University, Belozersky Institute of Physico-Chemical Biology, Moscow, Russia; Lomonosov Moscow State University, Institute of Mitoengineering, Moscow, Russia; Leibniz Institute for Zoo and Wildlife Research, Berlin, Germany; Lomonosov Moscow State University, Biological Faculty, Moscow, Russia; Lomonosov Moscow State University, Faculty of Mechanics and Mathematics, Moscow, Russia
| | - Lora E. Bakeeva
- Lomonosov Moscow State University, Belozersky Institute of Physico-Chemical Biology, Moscow, Russia; Lomonosov Moscow State University, Institute of Mitoengineering, Moscow, Russia; Leibniz Institute for Zoo and Wildlife Research, Berlin, Germany; Lomonosov Moscow State University, Biological Faculty, Moscow, Russia; Lomonosov Moscow State University, Faculty of Mechanics and Mathematics, Moscow, Russia
| | - Maxim V. Skulachev
- Lomonosov Moscow State University, Belozersky Institute of Physico-Chemical Biology, Moscow, Russia; Lomonosov Moscow State University, Institute of Mitoengineering, Moscow, Russia; Leibniz Institute for Zoo and Wildlife Research, Berlin, Germany; Lomonosov Moscow State University, Biological Faculty, Moscow, Russia; Lomonosov Moscow State University, Faculty of Mechanics and Mathematics, Moscow, Russia
| | - Alexander V. Markov
- Lomonosov Moscow State University, Belozersky Institute of Physico-Chemical Biology, Moscow, Russia; Lomonosov Moscow State University, Institute of Mitoengineering, Moscow, Russia; Leibniz Institute for Zoo and Wildlife Research, Berlin, Germany; Lomonosov Moscow State University, Biological Faculty, Moscow, Russia; Lomonosov Moscow State University, Faculty of Mechanics and Mathematics, Moscow, Russia
| | - Thomas B. Hildebrandt
- Lomonosov Moscow State University, Belozersky Institute of Physico-Chemical Biology, Moscow, Russia; Lomonosov Moscow State University, Institute of Mitoengineering, Moscow, Russia; Leibniz Institute for Zoo and Wildlife Research, Berlin, Germany; Lomonosov Moscow State University, Biological Faculty, Moscow, Russia; Lomonosov Moscow State University, Faculty of Mechanics and Mathematics, Moscow, Russia
| | - Viktor A. Sadovnichii
- Lomonosov Moscow State University, Belozersky Institute of Physico-Chemical Biology, Moscow, Russia; Lomonosov Moscow State University, Institute of Mitoengineering, Moscow, Russia; Leibniz Institute for Zoo and Wildlife Research, Berlin, Germany; Lomonosov Moscow State University, Biological Faculty, Moscow, Russia; Lomonosov Moscow State University, Faculty of Mechanics and Mathematics, Moscow, Russia
| |
Collapse
|
42
|
Tan J, Wang Y, Wang S, Zhang N, Wu S, Yuan Z, Zhu X. Untargeted metabolomics analysis of adipogenic transformation in OP9-DL1 cells using liquid chromatography-mass spectrometry: Implications for thymic adipogenesis. Cell Biol Int 2017; 41:447-456. [PMID: 28185342 DOI: 10.1002/cbin.10740] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Accepted: 02/04/2017] [Indexed: 01/07/2023]
Abstract
Adipocyte deposition is a key feature of age-related thymic involution, but the underlying mechanisms responsible for thymic adiposity remain to be elucidated. In the present study, we utilized rosiglitazone, a potent peroxisome proliferator-activated receptor γ agonist, to induce adipogenic differentiation of OP9-DL1 cells, and detected the metabolomics alterations during adipogenic differentiation by using liquid chromatography-mass spectrometry. The obtained metabolites were further processed by multivariate statistical analysis, including principal component analysis, partial least squares discriminant analysis, and orthogonal projection on latent-structures discriminant analysis. As a result, we identified a total of 33 significantly differential metabolites between dimethyl sulphoxide- and rosiglitazone-treated OP9-DL1 cells, which were closely related to the dysregulation of phospholipid metabolism pathway, oxidative stress, and associated amino acid metabolism. Meanwhile, two pathways including glycerophospholipid metabolism and nitrogen metabolism were significantly perturbed (P < 0.05). Collectively, our results may provide some heuristic guidance for addressing the underlying mechanism of thymic adipogenesis, and future studies are warranted to unravel the functions of these altered metabolites in thymic adipogenesis.
Collapse
Affiliation(s)
- Jianxin Tan
- Research Center, Shengjing Hospital of China Medical University, Shenyang, 110004, People's Republic of China
| | - Yajun Wang
- Research Center, Shengjing Hospital of China Medical University, Shenyang, 110004, People's Republic of China
| | - Siliang Wang
- Department of Medical Oncology, Shengjing Hospital of China Medical University, Shenyang, 110022, People's Republic of China
| | - Nannan Zhang
- Department of Nerve Function, Shengjing Hospital of China Medical University, Shenyang, 110004, People's Republic of China
| | - Simeng Wu
- Department of Blood Transfusion, Shengjing Hospital of China Medical University, Shenyang, 110022, People's Republic of China
| | - Zhe Yuan
- Department of Blood Transfusion, Shengjing Hospital of China Medical University, Shenyang, 110022, People's Republic of China
| | - Xike Zhu
- Research Center, Shengjing Hospital of China Medical University, Shenyang, 110004, People's Republic of China
| |
Collapse
|
43
|
Shilovsky GA, Feniouk BA, Skulachev VP. Thymic Involution in Ontogenesis: Role in Aging Program. BIOCHEMISTRY (MOSCOW) 2016; 80:1629-31. [PMID: 26638690 DOI: 10.1134/s0006297915120135] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
In most mammals, involution of the thymus occurs with aging. In this issue of Biochemistry (Moscow) devoted to phenoptosis, A. V. Khalyavkin considered involution of a thymus as an example of the program of development and further--of proliferation control and prevention of tumor growth. However, in animals devoid of a thymus (e.g. naked mice), stimulation of carcinogenesis, but not its prevention was observed. In this report, we focus on the involution of the thymus as a manifestation of the aging program (slow phenoptosis). We also consider methods of reversal/arrest of this program at different levels of organization of life (cell, tissue, and organism) including surgical manipulations, hormonal effects, genetic techniques, as well as the use of conventional and mitochondria-targeted antioxidants. We conclude that programmed aging (at least on the model of age-dependent thymic atrophy) can be inhibited.
Collapse
Affiliation(s)
- G A Shilovsky
- Lomonosov Moscow State University, Belozersky Institute of Physico-Chemical Biology, Moscow, 119991, Russia.
| | | | | |
Collapse
|
44
|
Masters AR, Haynes L, Su DM, Palmer DB. Immune senescence: significance of the stromal microenvironment. Clin Exp Immunol 2016; 187:6-15. [PMID: 27529161 DOI: 10.1111/cei.12851] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/09/2016] [Indexed: 12/12/2022] Open
Abstract
The immune system undergoes age-associated changes known as immunosenescence, resulting in increased susceptibility to infections, cancers and autoimmunity in the aged. The basis of our understanding of immunosenescence has been derived primarily from studies examining intrinsic defects within many of the cells of the immune system. While these studies have provided insight into the mechanisms of immunosenescence, a picture is now emerging that the stromal microenvironment within lymphoid organs also contributes significantly to the age-associated decline of immune function. These extrinsic defects appear to impact the functional activity of immune cells and may offer a potential target to recover immune activity. Indeed, rejuvenation studies which have targeted the stromal niche have restored immune function in aged successfully, highlighting the impact of the microenvironment towards the aetiology of immunosenescence.
Collapse
Affiliation(s)
- A R Masters
- UConn Center on Aging, University of Connecticut School of Medicine, Farmington, CT, USA.,Department of Immunology, University of Connecticut School of Medicine, Farmington, CT, USA
| | - L Haynes
- UConn Center on Aging, University of Connecticut School of Medicine, Farmington, CT, USA.,Department of Immunology, University of Connecticut School of Medicine, Farmington, CT, USA
| | - D-M Su
- Department of Molecular Biology and Immunology, University of North Texas Health Science Center at Fort Worth, Fort Worth, TX, USA
| | - D B Palmer
- Department of Comparative Biomedical Sciences, Royal Veterinary College, University of London, UK
| |
Collapse
|
45
|
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
|
46
|
Ohigashi I, Kozai M, Takahama Y. Development and developmental potential of cortical thymic epithelial cells. Immunol Rev 2016; 271:10-22. [DOI: 10.1111/imr.12404] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Izumi Ohigashi
- Division of Experimental Immunology; Institute for Genome Research; University of Tokushima; Tokushima Japan
| | - Mina Kozai
- Division of Experimental Immunology; Institute for Genome Research; University of Tokushima; Tokushima Japan
| | - Yousuke Takahama
- Division of Experimental Immunology; Institute for Genome Research; University of Tokushima; Tokushima Japan
| |
Collapse
|
47
|
Yang KE, Jang H, Hwang I, Chung Y, Choi J, Lee T, Chung Y, Lee M, Lee MY, Yeo E, Jang I. Phenyl 2-pyridyl ketoxime induces cellular senescence-like alterations via nitric oxide production in human diploid fibroblasts. Aging Cell 2016; 15:245-55. [PMID: 26696133 PMCID: PMC4783342 DOI: 10.1111/acel.12429] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/08/2015] [Indexed: 12/15/2022] Open
Abstract
Phenyl-2-pyridyl ketoxime (PPKO) was found to be one of the small molecules enriched in the extracellular matrix of near-senescent human diploid fibroblasts (HDFs). Treatment of young HDFs with PPKO reduced the viability of young HDFs in a dose- and time-dependent manner and resulted in senescence-associated β-galactosidase (SA-β-gal) staining and G2/M cell cycle arrest. In addition, the levels of some senescence-associated proteins, such as phosphorylated ERK1/2, caveolin-1, p53, p16(ink4a), and p21(waf1), were elevated in PPKO-treated cells. To monitor the effect of PPKO on cell stress responses, reactive oxygen species (ROS) production was examined by flow cytometry. After PPKO treatment, ROS levels transiently increased at 30 min but then returned to baseline at 60 min. The levels of some antioxidant enzymes, such as catalase, peroxiredoxin II and glutathione peroxidase I, were transiently induced by PPKO treatment. SOD II levels increased gradually, whereas the SOD I and III levels were biphasic during the experimental periods after PPKO treatment. Cellular senescence induced by PPKO was suppressed by chemical antioxidants, such as N-acetylcysteine, 2,2,6,6-tetramethylpiperidinyloxy, and L-buthionine-(S,R)-sulfoximine. Furthermore, PPKO increased nitric oxide (NO) production via inducible NO synthase (iNOS) in HDFs. In the presence of NOS inhibitors, such as L-NG-nitroarginine methyl ester and L-NG-monomethylarginine, PPKO-induced transient NO production and SA-β-gal staining were abrogated. Taken together, these results suggest that PPKO induces cellular senescence in association with transient ROS and NO production and the subsequent induction of senescence-associated proteins.
Collapse
Affiliation(s)
- Kyeong Eun Yang
- Drug & Disease Target Group Division of Bioconvergence Analysis Korea Basic Science Institute Daejeon 305‐333 Korea
| | - Hyun‐Jin Jang
- Drug & Disease Target Group Division of Bioconvergence Analysis Korea Basic Science Institute Daejeon 305‐333 Korea
| | - In‐Hu Hwang
- Department of Physiology Korea University College of Medicine Seoul 02841 Korea
| | - Young‐Ho Chung
- Drug & Disease Target Group Division of Bioconvergence Analysis Korea Basic Science Institute Daejeon 305‐333 Korea
| | - Jong‐Soon Choi
- Drug & Disease Target Group Division of Bioconvergence Analysis Korea Basic Science Institute Daejeon 305‐333 Korea
| | - Tae‐Hoon Lee
- Department of Oral Biochemistry Dental Science Research Institute Chonnam National University Gwangju 500‐757 Korea
| | - Yun‐Jo Chung
- Center for University‐Wide Research Facilities Chonbuk National University Jeonju Korea
| | - Min‐Seung Lee
- Department of Biochemistry College of Medicine Gachon University Inchon 406‐799 Korea
| | - Mi Young Lee
- KM Convergence Research Division Korea Institute of Oriental Medicine Daejeon 305‐811 Korea
| | - Eui‐Ju Yeo
- Department of Biochemistry College of Medicine Gachon University Inchon 406‐799 Korea
| | - Ik‐Soon Jang
- Drug & Disease Target Group Division of Bioconvergence Analysis Korea Basic Science Institute Daejeon 305‐333 Korea
| |
Collapse
|