1
|
Li L, Xu F, Han Y, Zeng J, Du S, Wang C. Thymic microenvironment's impact on immunosenescence. Immunol Res 2024; 72:1161-1173. [PMID: 39042204 DOI: 10.1007/s12026-024-09519-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 07/15/2024] [Indexed: 07/24/2024]
Abstract
Age-related thymic involution is characterized by the loss of T cell development and the supporting epithelial network, which are replaced by adipose tissue. We previously showed that aging functionally impairs lymphohematopoietic progenitor cells, including thymic early T cell progenitors (ETPs), contributing to thymic involution. Considering that the thymic microenvironment is essential for thymocyte incubation, we aimed to investigate its role in age-related thymic involution and the mechanisms underlying these changes. The challenge in studying these processes led us to transplant T cell-depleted fetal thymus tissue into the kidney capsule of aged mice. This model allowed us to identify the mechanisms driving age-related changes in the thymic microenvironment and to assess whether these changes could be reversed. Flow cytometry was used to detect naïve T cells (CD62L+CD44-), including CD4 CD8 double-negative, double-positive, and single-positive T cells. Real-time PCR was used to detect and quantify signal-joint T cell receptor excision circles. We rearranged δRec-ΨJα in murine peripheral blood leukocytes to evaluate the thymic output of newly developed naïve T cells in the mice and gene expression in the thymus. Age-related thymic involution decreased naïve T cells and increased memory T cells, while fetal thymus transplantation improved thymic output and T cell production and reversed the impairment of thymopoiesis due to thymic involution in aged mice. Furthermore, the expression of key cytokines was restored and ETPs in the aged mice showed normal thymic T cell development. Our study suggests that degenerative changes in the thymic microenvironment are the primary cause of thymic dysfunction, leading to immunosenescence associated with age-related thymic involution.
Collapse
Affiliation(s)
- Li Li
- Shenzhen Guangming District People's Hospital, 4253 Songbai Road, Matian Street, Guangming District, Shenzhen, 518106, Guangdong, China
| | - Feng Xu
- Shenzhen Guangming District People's Hospital, 4253 Songbai Road, Matian Street, Guangming District, Shenzhen, 518106, Guangdong, China
| | - Yi Han
- Shenzhen Guangming District People's Hospital, 4253 Songbai Road, Matian Street, Guangming District, Shenzhen, 518106, Guangdong, China
| | - Jun Zeng
- Shenzhen Guangming District People's Hospital, 4253 Songbai Road, Matian Street, Guangming District, Shenzhen, 518106, Guangdong, China
| | - Shan Du
- Shenzhen Guangming District People's Hospital, 4253 Songbai Road, Matian Street, Guangming District, Shenzhen, 518106, Guangdong, China
| | - Changshan Wang
- Shenzhen Guangming District People's Hospital, 4253 Songbai Road, Matian Street, Guangming District, Shenzhen, 518106, Guangdong, China.
| |
Collapse
|
2
|
Kousa AI, Jahn L, Zhao K, Flores AE, Acenas D, Lederer E, Argyropoulos KV, Lemarquis AL, Granadier D, Cooper K, D'Andrea M, Sheridan JM, Tsai J, Sikkema L, Lazrak A, Nichols K, Lee N, Ghale R, Malard F, Andrlova H, Velardi E, Youssef S, Burgos da Silva M, Docampo M, Sharma R, Mazutis L, Wimmer VC, Rogers KL, DeWolf S, Gipson B, Gomes ALC, Setty M, Pe'er D, Hale L, Manley NR, Gray DHD, van den Brink MRM, Dudakov JA. Age-related epithelial defects limit thymic function and regeneration. Nat Immunol 2024; 25:1593-1606. [PMID: 39112630 PMCID: PMC11362016 DOI: 10.1038/s41590-024-01915-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 07/03/2024] [Indexed: 09/01/2024]
Abstract
The thymus is essential for establishing adaptive immunity yet undergoes age-related involution that leads to compromised immune responsiveness. The thymus is also extremely sensitive to acute insult and although capable of regeneration, this capacity declines with age for unknown reasons. We applied single-cell and spatial transcriptomics, lineage-tracing and advanced imaging to define age-related changes in nonhematopoietic stromal cells and discovered the emergence of two atypical thymic epithelial cell (TEC) states. These age-associated TECs (aaTECs) formed high-density peri-medullary epithelial clusters that were devoid of thymocytes; an accretion of nonproductive thymic tissue that worsened with age, exhibited features of epithelial-to-mesenchymal transition and was associated with downregulation of FOXN1. Interaction analysis revealed that the emergence of aaTECs drew tonic signals from other functional TEC populations at baseline acting as a sink for TEC growth factors. Following acute injury, aaTECs expanded substantially, further perturbing trophic regeneration pathways and correlating with defective repair of the involuted thymus. These findings therefore define a unique feature of thymic involution linked to immune aging and could have implications for developing immune-boosting therapies in older individuals.
Collapse
Grants
- T32-GM007270 U.S. Department of Health & Human Services | NIH | National Institute of General Medical Sciences (NIGMS)
- 1187367 Department of Health | National Health and Medical Research Council (NHMRC)
- R01 CA228308 NCI NIH HHS
- 1158024 Department of Health | National Health and Medical Research Council (NHMRC)
- R01-HL145276 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01-HL147584 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01-HL165673 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01 HL123340 NHLBI NIH HHS
- R01 HL145276 NHLBI NIH HHS
- R01-CA228308 U.S. Department of Health & Human Services | NIH | National Cancer Institute (NCI)
- T32 GM007103 NIGMS NIH HHS
- P30 CA015704 NCI NIH HHS
- P01 CA023766 NCI NIH HHS
- R01 HL165673 NHLBI NIH HHS
- R01 HL147584 NHLBI NIH HHS
- P01-AG052359 U.S. Department of Health & Human Services | NIH | National Institute on Aging (U.S. National Institute on Aging)
- P30-CA015704 U.S. Department of Health & Human Services | NIH | National Cancer Institute (NCI)
- 1090236 Department of Health | National Health and Medical Research Council (NHMRC)
- P30 CA008748 NCI NIH HHS
- P01 AG052359 NIA NIH HHS
- T32 GM007270 NIGMS NIH HHS
- 1102104 Cancer Council Victoria
- 1078763 Department of Health | National Health and Medical Research Council (NHMRC)
- 1146518 Cancer Council Victoria
- U01-AI70035 U.S. Department of Health & Human Services | NIH | National Institute of Allergy and Infectious Diseases (NIAID)
- R35 HL171556 NHLBI NIH HHS
- ALTF-431-2017 European Molecular Biology Organization (EMBO)
- R01 CA228358 NCI NIH HHS
- F30-HL165761 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01-HL123340 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R35-HL-171556 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- 1121325 Department of Health | National Health and Medical Research Council (NHMRC)
- F30 HL165761 NHLBI NIH HHS
- U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- U.S. Department of Health & Human Services | NIH | National Institute of Allergy and Infectious Diseases (NIAID)
- U.S. Department of Health & Human Services | NIH | National Cancer Institute (NCI)
- U.S. Department of Health & Human Services | NIH | National Institute on Aging (U.S. National Institute on Aging)
- U.S. Department of Health & Human Services | NIH | National Institute of General Medical Sciences (NIGMS)
Collapse
Affiliation(s)
- Anastasia I Kousa
- Program in Immunology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Translational Science and Therapeutics Division, and Immunotherapy Integrated Research Center, Fred Hutchinson Cancer Center, Seattle, WA, USA
- City of Hope Los Angeles and National Medical Center, Duarte, CA, USA
| | - Lorenz Jahn
- Program in Immunology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Kelin Zhao
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
| | - Angel E Flores
- Department of Genetics, University of Georgia, Athens, GA, USA
| | - Dante Acenas
- Translational Science and Therapeutics Division, and Immunotherapy Integrated Research Center, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Department of Immunology, University of Washington, Seattle, WA, USA
| | - Emma Lederer
- Translational Science and Therapeutics Division, and Immunotherapy Integrated Research Center, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Department of Immunology, University of Washington, Seattle, WA, USA
| | - Kimon V Argyropoulos
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Andri L Lemarquis
- Program in Immunology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- City of Hope Los Angeles and National Medical Center, Duarte, CA, USA
| | - David Granadier
- Translational Science and Therapeutics Division, and Immunotherapy Integrated Research Center, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Kirsten Cooper
- Translational Science and Therapeutics Division, and Immunotherapy Integrated Research Center, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Michael D'Andrea
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
| | - Julie M Sheridan
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
| | - Jennifer Tsai
- Program in Immunology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Lisa Sikkema
- Computational and Systems Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Institute of Computational Biology, Helmholtz Center Munich, Munich, Germany
| | - Amina Lazrak
- Program in Immunology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Katherine Nichols
- Program in Immunology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Nichole Lee
- Program in Immunology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Romina Ghale
- Program in Immunology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Florent Malard
- Program in Immunology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Sorbonne Université, Centre de Recherche Saint-Antoine INSERM UMRs938, Service d'Hématologie Clinique et de Thérapie Cellulaire, Hôpital Saint Antoine, AP-HP, Paris, France
| | - Hana Andrlova
- Program in Immunology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Enrico Velardi
- Division of Pediatric Hematology and Oncology, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Salma Youssef
- Program in Immunology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | - Melissa Docampo
- Program in Immunology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Roshan Sharma
- Computational and Systems Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Linas Mazutis
- Computational and Systems Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Verena C Wimmer
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
| | - Kelly L Rogers
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
| | - Susan DeWolf
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Brianna Gipson
- Program in Immunology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Antonio L C Gomes
- Program in Immunology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Manu Setty
- Computational and Systems Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Basic Sciences Division & Translational Data Science Integrated Research Center, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Dana Pe'er
- Computational and Systems Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Laura Hale
- Human Vaccine Institute, Duke University, Durham, NC, USA
| | - Nancy R Manley
- Department of Genetics, University of Georgia, Athens, GA, USA
- School of Life Sciences, Arizona State University, Phoenix, AZ, USA
| | - Daniel H D Gray
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia.
- Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia.
| | - Marcel R M van den Brink
- Program in Immunology, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- City of Hope Los Angeles and National Medical Center, Duarte, CA, USA.
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| | - Jarrod A Dudakov
- Translational Science and Therapeutics Division, and Immunotherapy Integrated Research Center, Fred Hutchinson Cancer Center, Seattle, WA, USA.
- Department of Immunology, University of Washington, Seattle, WA, USA.
| |
Collapse
|
3
|
Liu T, Xia S. The Proteostasis of Thymic Stromal Cells in Health and Diseases. Protein J 2024; 43:447-463. [PMID: 38622349 DOI: 10.1007/s10930-024-10197-x] [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] [Accepted: 04/05/2024] [Indexed: 04/17/2024]
Abstract
The thymus is the key immune organ for the development of T cells. Different populations of thymic stromal cells interact with T cells, thereby controlling the dynamic development of T cells through their differentiation and function. Proteostasis represents a balance between protein expression, folding, and modification and protein clearance, and its fluctuation usually depends at least partially on related protein regulatory systems for further survival and effects. However, in terms of the substantial requirement for self-antigens and their processing burden, increasing evidence highlights that protein regulation contributes to the physiological effects of thymic stromal cells. Impaired proteostasis may expedite the progression of thymic involution and dysfunction, accompanied by the development of autoimmune diseases or thymoma. Hence, in this review, we summarize the regulation of proteostasis within different types of thymic stromal cells under physiological and pathological conditions to identify potential targets for thymic regeneration and immunotherapy.
Collapse
Affiliation(s)
- Ting Liu
- Department of Immunology, School of Medicine, Jiangsu University, 301, Xuefu Road, Zhenjiang, Jiangsu, 212013, China
| | - Sheng Xia
- Department of Immunology, School of Medicine, Jiangsu University, 301, Xuefu Road, Zhenjiang, Jiangsu, 212013, China.
| |
Collapse
|
4
|
Bushmeleva K, Vyshtakalyuk A, Terenzhev D, Belov T, Nikitin E, Zobov V. Effect of Flavonols of Aronia melanocarpa Fruits on Morphofunctional State of Immunocompetent Organs of Rats under Cyclophosphamide-Induced Immunosuppression. Biomolecules 2024; 14:578. [PMID: 38785985 PMCID: PMC11117470 DOI: 10.3390/biom14050578] [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: 03/25/2024] [Revised: 05/02/2024] [Accepted: 05/10/2024] [Indexed: 05/25/2024] Open
Abstract
Aronia melanocarpa berries contain many compounds with potential benefits for human health. The food flavonoids quercetin and rutin, found in significant amounts in the fruits of A. melanocarpa, are known to have favourable effects on animal and human organisms. However, data on the effect of flavonols isolated from black chokeberry on immune functions during immunosuppression are not available in the literature. Thus, the aim of this study was to evaluate the effect of flavonol fraction isolated from A. melanocarpa fruits, in comparison with pure quercetin and rutin substances, on the dysfunctional state of rat thymus and spleen in immunodeficiency. The study was performed on Wistar rats. The animals were orally administered solutions of the investigated substances for 7 days: water, a mixture of quercetin and rutin and flavonol fraction of A. melanocarpa. For induction of immunosuppression, the animals were injected once intraperitoneally with cyclophosphamide. Substance administration was then continued for another 7 days. The results showed that under the influence of flavonols, there was a decrease in cyclophosphamide-mediated reaction of lipid peroxidation enhancement and stimulation of proliferation of lymphocytes of thymus and spleen in rats. At that, the effect of the flavonol fraction of aronia was more pronounced.
Collapse
Affiliation(s)
- Kseniya Bushmeleva
- A.E. Arbuzov Institute of Organic and Physical Chemistry, Kazan Scientific Center, Russian Academy of Sciences, Arbuzov Str. 8, Kazan 420088, Russia; (A.V.); (D.T.); (T.B.); (E.N.); (V.Z.)
| | | | | | | | | | | |
Collapse
|
5
|
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
|
6
|
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: 1.0] [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
|
7
|
Li YR, Zúñiga-Pflücker JC. Thymus aging and immune reconstitution, progresses and challenges. Semin Immunol 2023; 70:101837. [PMID: 37659170 DOI: 10.1016/j.smim.2023.101837] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 08/25/2023] [Accepted: 08/25/2023] [Indexed: 09/04/2023]
Abstract
Thymus is a primary lymphoid organ essential for the development of T lymphocytes. Age-related thymic involution is a prominent feature of immune senescence. The thymus undergoes rapid growth during fetal and neonatal development, peaks in size before puberty and then begins to undergo a decrease in cellularity with age. Dramatic changes occur with age-associated thymic involution. The most prominent features of thymic involution include: (i) epithelial structure disruption, (ii) adipogenesis, and (iii) thymocyte development arrest. There is a sex disparity in thymus aging. It is a multifactorial process controlled and regulated by a series of molecules, including the transcription factor FOXN1, fibroblast and keratinocyte growth factors (FGF and KGF, respectively), sex steroids, Notch signaling, WNT signaling, and microRNAs. Nevertheless, there is still no satisfactory evolutionary or physiological explanation for age-associated thymic involution, and understanding the precise mechanism(s) for thymus aging remains challenging. Sustained thymic regeneration has yet to be achieved by sex steroid ablation. Recent preclinical studies indicate that long-term thymic reconstitution can be achieved via adoptive transfer of in vitro-generated progenitor T (proT) cells, and improvements in the methods for the generation of human proT cells make this an attractive approach. Future clinical applications may rely on new applications integrating proT cells, cytokine support and sex-steroid inhibition treatments.
Collapse
Affiliation(s)
- Yue Ru Li
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Juan Carlos Zúñiga-Pflücker
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada; Biological Sciences, Sunnybrook Research Institute, Toronto, Ontario, Canada.
| |
Collapse
|
8
|
Wang H, Zhang Y, Wang Z, Zhang L, Guo M, Cao C, Xiao H. Deciphering Nucleic Acid Binding Proteome of Mouse Immune Organs Reveals Hub Proteins for Aging. Mol Cell Proteomics 2023; 22:100611. [PMID: 37391046 PMCID: PMC10412848 DOI: 10.1016/j.mcpro.2023.100611] [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: 02/23/2023] [Revised: 05/24/2023] [Accepted: 06/26/2023] [Indexed: 07/02/2023] Open
Abstract
Profiling the nucleic acid-binding proteins (NABPs) during aging process is critical to elucidate its roles in biological systems as well as transcriptional and translational regulation. Here, we developed a comprehensive strategy to survey the NABPs of mouse immune organs by using single cell preparation and selective capture technology-based proteomics. Our approach provided a global view of tissue NABPs from different organs under normal physiological conditions with extraction specificity of 70 to 90%. Through quantitative proteomics analysis of mouse spleen and thymus at 1, 4, 12, 24, 48, and 72 weeks, we investigated the molecular features of aging-related NABPs. A total of 2674 proteins were quantified in all six stages, demonstrating distinct and time-specific expression pattern of NABPs. Thymus and spleen exhibited unique aging signatures, and differential proteins and pathways were enriched across the mouse lifespan. Three core modules and 16 hub proteins associated with aging were revealed through weighted gene correlation network analysis. Significant candidates were screened for immunoassay verification, and six hub proteins were confirmed. The integrated strategy pertains the capability to decipher the dynamic functions of NABPs in aging physiology and benefit further mechanism research.
Collapse
Affiliation(s)
- Huiyu Wang
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory of Metabolic & Developmental Sciences, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Yan Zhang
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
| | - Zeyuan Wang
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory of Metabolic & Developmental Sciences, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Lu Zhang
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory of Metabolic & Developmental Sciences, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Miao Guo
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory of Metabolic & Developmental Sciences, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Chengxi Cao
- Department of Instrument Science and Engineering, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Hua Xiao
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory of Metabolic & Developmental Sciences, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
9
|
Zhang X, Schalke B, Kvell K, Kriegsmann K, Kriegsmann M, Graeter T, Preissler G, Ott G, Kurz K, Bulut E, Ströbel P, Marx A, Belharazem D. WNT4 overexpression and secretion in thymic epithelial tumors drive an autocrine loop in tumor cells in vitro. Front Oncol 2022; 12:920871. [PMID: 35965500 PMCID: PMC9372913 DOI: 10.3389/fonc.2022.920871] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 07/01/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundWNT4-driven non-canonical signaling is crucial for homeostasis and age-related involution of the thymus. Abnormal WNT signaling is important in many cancers, but the role of WNT signaling in thymic tumors is largely unknown.Materials & MethodsExpression and function of WNT4 and FZD6 were analyzed using qRT–PCR, Western blot, ELISA, in biopsies of non-neoplastic thymi (NT), thymoma and thymic carcinomas. ShRNA techniques and functional assays were used in primary thymic epithelial cells (pTECs) and TC cell line 1889c. Cells were conventionally (2D) grown and in three-dimensional (3D) spheroids.ResultsIn biopsy, WHO classified B3 thymomas and TCs showed increased WNT4 expression compared with NTs. During short-term 2D culture, WNT4 expression and secretion declined in neoplastic pTECs but not in 3D spheroids or medium supplemented with recombinant WNT4 cultures. Under the latter condition, the growth of pTECs was accompanied by increased expression of non-canonical targets RAC1 and JNK. Down-regulation of WNT4 by shRNA induced cell death in pTECs derived from B3 thymomas and led to decreased RAC1, but not JNK protein phosphorylation. Pharmacological inhibition of NF-κB decreased both RAC1 and JNK phosphorylation in neoplastic pTECs.ConclusionsLack of the age-related decline of non-canonical WNT4 expression in TETs and restoration of declining WNT4 expression through exogeneous WNT4 or 3D culture of pTECs hints at an oncogenic role of WNT4 in TETs and is compatible with the WNT4 autocrine loop model. Crosstalk between WNT4 and NF-κB signaling may present a promising target for combined interventions in TETs.
Collapse
Affiliation(s)
- Xiaonan Zhang
- Institute of Pathology and Medical Research Center, University Medical Centre Mannheim, University of Heidelberg, Mannheim, Germany
| | - Berthold Schalke
- Department of Neurology, University of Regensburg, Regensburg, Germany
| | - Krisztian Kvell
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, University of Pecs, Pecs, Hungary
| | - Katharina Kriegsmann
- Department of Hematology, Oncology and Rheumatology, University Hospital Heidelberg, Heidelberg, Germany
| | - Mark Kriegsmann
- Translational Lung Research Centre Heidelberg, German Centre for Lung Research, Heidelberg, Germany
- Institute of Pathology, University Hospital Heidelberg, Heidelberg University, Heidelberg, Germany
| | - Thomas Graeter
- Department of Thoracic Surgery, University Medical Centre Erlangen, Erlangen, Germany
| | - Gerhard Preissler
- Department of Thoraxic Surgery, Clinic Schillerhöhe, Robert-Bosch-Hospital, Gerlingen, Löwenstein, Germany
| | - German Ott
- Department of Clinical Pathology, Robert-Bosch-Hospital, Stuttgart, Germany
- Dr. Margarete Fischer-Bosch Institute for Clinical Pharmacology, Robert-Bosch-Hospital, Stuttgart, Germany
| | - Katrin Kurz
- Department of Clinical Pathology, Robert-Bosch-Hospital, Stuttgart, Germany
- Dr. Margarete Fischer-Bosch Institute for Clinical Pharmacology, Robert-Bosch-Hospital, Stuttgart, Germany
| | - Elena Bulut
- Department of Thoraxic Surgery, Thoraxklinik at Heidelberg University Hospital, Heidelberg, Germany
| | - Philipp Ströbel
- Institute of Pathology, University Medical Center Göttingen, University of Göttingen, Göttingen, Germany
| | - Alexander Marx
- Institute of Pathology and Medical Research Center, University Medical Centre Mannheim, University of Heidelberg, Mannheim, Germany
| | - Djeda Belharazem
- Institute of Pathology and Medical Research Center, University Medical Centre Mannheim, University of Heidelberg, Mannheim, Germany
- *Correspondence: Djeda Belharazem,
| |
Collapse
|
10
|
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: 54] [Impact Index Per Article: 18.0] [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
|
11
|
Rosichini M, Catanoso M, Screpanti I, Felli MP, Locatelli F, Velardi E. Signaling Crosstalks Drive Generation and Regeneration of the Thymus. Front Immunol 2022; 13:920306. [PMID: 35734178 PMCID: PMC9207182 DOI: 10.3389/fimmu.2022.920306] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 05/17/2022] [Indexed: 12/19/2022] Open
Abstract
Optimal recovery of immune competence after periods of hematopoietic insults or stress is crucial to re-establish patient response to vaccines, pathogens and tumor antigens. This is particularly relevant for patients receiving high doses of chemotherapy or radiotherapy, who experience prolonged periods of lymphopenia, which can be associated with an increased risk of infections, malignant relapse, and adverse clinical outcome. While the thymus represents the primary organ responsible for the generation of a diverse pool of T cells, its function is profoundly impaired by a range of acute insults (including those caused by cytoreductive chemo/radiation therapy, infections and graft-versus-host disease) and by the chronic physiological deterioration associated with aging. Impaired thymic function increases the risk of infections and tumor antigen escape due to a restriction in T-cell receptor diversity and suboptimal immune response. Therapeutic approaches that can promote the renewal of the thymus have the potential to restore immune competence in patients. Previous work has documented the importance of the crosstalk between thymocytes and thymic epithelial cells in establishing correct architecture and function of thymic epithelium. This crosstalk is relevant not only during thymus organogenesis, but also to promote the recovery of its function after injuries. In this review, we will analyze the signals involved in the crosstalk between TECs and hematopoietic cells. We will focus in particular on how signals from T-cells can regulate TEC function and discuss the relevance of these pathways in restoring thymic function and T-cell immunity in experimental models, as well as in the clinical setting.
Collapse
Affiliation(s)
- Marco Rosichini
- Department of Pediatric Hematology and Oncology, Cell and Gene Therapy, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Marialuigia Catanoso
- Department of Pediatric Hematology and Oncology, Cell and Gene Therapy, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Isabella Screpanti
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Maria Pia Felli
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Franco Locatelli
- Department of Pediatric Hematology and Oncology, Cell and Gene Therapy, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
- Department of Maternal and Child Health, Sapienza University of Rome, Rome, Italy
| | - Enrico Velardi
- Department of Pediatric Hematology and Oncology, Cell and Gene Therapy, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
- *Correspondence: Enrico Velardi,
| |
Collapse
|
12
|
Hu C, Zhang K, Jiang F, Wang H, Shao Q. Epigenetic modifications in thymic epithelial cells: an evolutionary perspective for thymus atrophy. Clin Epigenetics 2021; 13:210. [PMID: 34819170 PMCID: PMC8612001 DOI: 10.1186/s13148-021-01197-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 11/08/2021] [Indexed: 02/06/2023] Open
Abstract
Background The thymic microenvironment is mainly comprised of thymic epithelial cells, the cytokines, exosomes, surface molecules, and hormones from the cells, and plays a vital role in the development, differentiation, maturation and homeostasis of T lymphocytes. However, the thymus begins to degenerate as early as the second year of life and continues through aging in human beings, leading to a decreased output of naïve T cells, the limited TCR diversity and an expansion of monoclonal memory T cells in the periphery organs. These alternations will reduce the adaptive immune response to tumors and emerging infectious diseases, such as COVID-19, also it is easier to suffer from autoimmune diseases in older people. In the context of global aging, it is important to investigate and clarify the causes and mechanisms of thymus involution. Main body Epigenetics include histone modification, DNA methylation, non-coding RNA effects, and chromatin remodeling. In this review, we discuss how senescent thymic epithelial cells determine and control age-related thymic atrophy, how this process is altered by epigenetic modification. How the thymus adipose influences the dysfunctions of the thymic epithelial cells, and the prospects of targeting thymic epithelial cells for the treatment of thymus atrophy. Conclusion Epigenetic modifications are emerging as key regulators in governing the development and senescence of thymic epithelial cells. It is beneficial to re-establish effective thymopoiesis, identify the potential therapeutic strategy and rejuvenate the immune function in the elderly.
Collapse
Affiliation(s)
- Cexun Hu
- Department of Immunology, School of Medicine, Jiangsu University, Zhenjiang, 212013, Jiangsu, People's Republic of China.,Department of Immunology, Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, No. 301 Xuefu Road, Zhenjiang, 212013, Jiangsu, People's Republic of China
| | - Keyu Zhang
- Department of Immunology, School of Medicine, Jiangsu University, Zhenjiang, 212013, Jiangsu, People's Republic of China.,Department of Immunology, Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, No. 301 Xuefu Road, Zhenjiang, 212013, Jiangsu, People's Republic of China
| | - Feng Jiang
- Department of Immunology, School of Medicine, Jiangsu University, Zhenjiang, 212013, Jiangsu, People's Republic of China.,Department of Immunology, Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, No. 301 Xuefu Road, Zhenjiang, 212013, Jiangsu, People's Republic of China
| | - Hui Wang
- Department of Immunology, School of Medicine, Jiangsu University, Zhenjiang, 212013, Jiangsu, People's Republic of China. .,Department of Immunology, Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, No. 301 Xuefu Road, Zhenjiang, 212013, Jiangsu, People's Republic of China.
| | - Qixiang Shao
- Department of Immunology, School of Medicine, Jiangsu University, Zhenjiang, 212013, Jiangsu, People's Republic of China. .,Department of Immunology, Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, No. 301 Xuefu Road, Zhenjiang, 212013, Jiangsu, People's Republic of China. .,Jiangsu College of Nursing, School of Medical Science and Laboratory Medicine, Huai'an, 223002, Jiangsu, People's Republic of China.
| |
Collapse
|
13
|
Li B, Li W, Liu W, Xing J, Wu Y, Ma Y, Xu D, Li Y. Comprehensive analysis of lncRNAs, miRNAs and mRNAs related to thymic development and involution in goose. Genomics 2020; 113:1176-1188. [PMID: 33276006 DOI: 10.1016/j.ygeno.2020.11.031] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 11/11/2020] [Accepted: 11/30/2020] [Indexed: 11/16/2022]
Abstract
Thymic involution is a sign of immunosenescence, but little is known about it in goose. miRNAs and lncRNAs are critical factors regulating organ growth and development. In this study, we comprehensively analyzed the profiles of lncRNAs, miRNAs and mRNAs during the development and involution of the thymus in Magang goose. The results showed that 2436 genes, 16 miRNAs and 417 lncRNAs were differentially co-expressed between the developmental (20-embryo age, 3-day post-hatch and 3-month age) and degenerative (6-month age) stages. The functional analysis showed that these differentially expressed genes were significantly enriched in cell proliferation, cell adhesion, apoptotic signaling pathway, and Notch signaling pathway. In addition, we established a gene-gene network through the STRING database and identified 50 key genes. Finally, we constructed a miRNA-mRNA network followed by a lncRNA-miRNA-mRNA network. These results suggest that lncRNAs and miRNAs may be involved in the regulation of thymic development and involution in goose.
Collapse
Affiliation(s)
- Bingxin Li
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Wanyan Li
- Guangdong Province Key Laboratory of Waterfowl Healthy Breeding, Zhongkai University of Agriculture and Engineering, Guangzhou 510225, China
| | - Wenjun Liu
- Guangdong Province Key Laboratory of Waterfowl Healthy Breeding, Zhongkai University of Agriculture and Engineering, Guangzhou 510225, China
| | - Jingjing Xing
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Yingying Wu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Yongjiang Ma
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Danning Xu
- Guangdong Province Key Laboratory of Waterfowl Healthy Breeding, Zhongkai University of Agriculture and Engineering, Guangzhou 510225, China.
| | - Yugu Li
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China.
| |
Collapse
|
14
|
Abstract
Following periods of haematopoietic cell stress, such as after chemotherapy, radiotherapy, infection and transplantation, patient outcomes are linked to the degree of immune reconstitution, specifically of T cells. Delayed or defective recovery of the T cell pool has significant clinical consequences, including prolonged immunosuppression, poor vaccine responses and increased risks of infections and malignancies. Thus, strategies that restore thymic function and enhance T cell reconstitution can provide considerable benefit to individuals whose immune system has been decimated in various settings. In this Review, we focus on the causes and consequences of impaired adaptive immunity and discuss therapeutic strategies that can recover immune function, with a particular emphasis on approaches that can promote a diverse repertoire of T cells through de novo T cell formation.
Collapse
|
15
|
Fan P, Han B, Hu H, Wei Q, Zhang X, Meng L, Nie J, Tang X, Tian X, Zhang L, Wang L, Li J. Proteome of thymus and spleen reveals that 10-hydroxydec-2-enoic acid could enhance immunity in mice. Expert Opin Ther Targets 2020; 24:267-279. [PMID: 32077781 DOI: 10.1080/14728222.2020.1733529] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Objectives: 10-hydroxydec-2-enoic acid (10-HDA), a unique component of royal jelly existing only in nature, has the potential to promote human health. Knowledge of 10-HDA in regulating immuno-activity, however, is lacking. The aim of our work is to gain a novel understanding of 10-HDA in promoting immunity.Methods: Immuno-suppressed mice were generated by cyclophosphamide injection, After 10-HDA supplementation to the mice to rescue their immunity, the proteomes of the thymus and spleen were analyzed.Results: The weight of the body, thymus, and spleen in cyclophosphamide-induced mice recovered by 10-HDA indicate its potential role in immuno-organ protection. In the thymus, the enhanced activity of pathways associated with DNA/RNA/protein activities may be critical for T-lymphocyte proliferation/differentiation, and cytotoxicity. In the spleen, the induced pathways involved in DNA/RNA/protein activities, and cell proliferative stimulation suggest their vital role in B-lymphocyte affinity maturation, antigen presentation, and macrophage activity. The up-regulated proteins highly connected in networks modulated by 10-HDA indicate that the mice may evolve tactics to respond to immuno-organ impairment by activating critical physiological processes.Conclusion: Our data constitute a proof-of-concept that 10-HDA is a potential agent to improve immunity in the thymus and spleen and offer a new venue for applying natural products to the therapy for hypoimmunity.
Collapse
Affiliation(s)
- Pei Fan
- Institute of Apicultural Research, Chinese Academy of Agricultural Sciences, Beijing, PR China.,College of Biological Engineering, Henan University of Technology, Zhengzhou, PR China
| | - Bin Han
- Institute of Apicultural Research, Chinese Academy of Agricultural Sciences, Beijing, PR China
| | - Han Hu
- Institute of Apicultural Research, Chinese Academy of Agricultural Sciences, Beijing, PR China
| | - Qiaohong Wei
- Institute of Apicultural Research, Chinese Academy of Agricultural Sciences, Beijing, PR China
| | - Xufeng Zhang
- Institute of Apicultural Research, Chinese Academy of Agricultural Sciences, Beijing, PR China
| | - Lifeng Meng
- Institute of Apicultural Research, Chinese Academy of Agricultural Sciences, Beijing, PR China
| | - Jing Nie
- Department of Technology, Hunan SJA Laboratory Animal Co., Ltd, Changsha, PR China
| | - Xiaofeng Tang
- Department of Technology, Hunan SJA Laboratory Animal Co., Ltd, Changsha, PR China
| | - Xinyue Tian
- College of Biological Engineering, Henan University of Technology, Zhengzhou, PR China
| | - Lu Zhang
- College of Biological Engineering, Henan University of Technology, Zhengzhou, PR China
| | - Liping Wang
- Department of Research & Development, Henan Jianda Bio Sci. & Tech. Co., Ltd, Zhengzhou, PR China
| | - Jianke Li
- Institute of Apicultural Research, Chinese Academy of Agricultural Sciences, Beijing, PR China
| |
Collapse
|
16
|
Cruz-Chamorro I, Álvarez-Sánchez N, Escalante-Andicoechea C, Carrillo-Vico A, Rubio A, Guerrero JM, Molinero P, Lardone PJ. Temporal expression patterns of the melatoninergic system in the human thymus of children. Mol Metab 2019; 28:83-90. [PMID: 31378599 PMCID: PMC6822200 DOI: 10.1016/j.molmet.2019.07.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 07/12/2019] [Accepted: 07/21/2019] [Indexed: 02/08/2023] Open
Abstract
OBJECTIVES To obtain greater knowledge of the extra-pineal sources of melatonin during development, the amount of indolamine and the expression levels of the last two enzymes involved in its biosynthesis, Arylalkylamine N-acetyltransferase (AANAT) and acetylserotonin O-methyltransferase (ASMT), were analyzed in the human thymus from children from three different age groups (from days to years). The melatonin membrane and nuclear receptor expression levels also were studied. METHODS Quantitative reverse transcriptase PCR and western blot were performed to investigate the receptor and enzyme expression levels. The results were examined and correlated with the ages of the thymuses. RESULTS We found high levels of indolamine in the thymuses of newborns (younger than 1 month), which decreased during development; thymuses from the months (from 2 to 11 months) and years (from 1 to 12 years) groups showed lower levels. A similar decline was also observed in the mRNA of the AANAT enzyme and the expression levels of melatonin receptors. However, ASMT expression was exactly the opposite, with low levels in the newborn group and higher levels in the years group. Our results show that the thymic synthesis of melatonin occurs very early in childhood. Additionally, this is the first report that is focused on melatonin receptors expression in the human thymus. CONCLUSION Considering the limited melatonin synthesis performed by the newborn pineal gland, we suggest that the high levels of melatonin found in human thymus in this experimental group arise from synthesis in the tissue itself, which could be contributing to the immune efficiency at the thymic level.
Collapse
Affiliation(s)
- Ivan Cruz-Chamorro
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Avda. Manuel Siurot S/N, 41013, Seville, Spain; Departamento de Bioquímica Médica y Biología Molecular e Inmunología, Universidad de Sevilla, Avda. Sanchez Pizjuan S/N, 41009, Seville, Spain
| | - Nuria Álvarez-Sánchez
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Avda. Manuel Siurot S/N, 41013, Seville, Spain
| | - Cristina Escalante-Andicoechea
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Avda. Manuel Siurot S/N, 41013, Seville, Spain
| | - Antonio Carrillo-Vico
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Avda. Manuel Siurot S/N, 41013, Seville, Spain; Departamento de Bioquímica Médica y Biología Molecular e Inmunología, Universidad de Sevilla, Avda. Sanchez Pizjuan S/N, 41009, Seville, Spain
| | - Amalia Rubio
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Avda. Manuel Siurot S/N, 41013, Seville, Spain; Departamento de Bioquímica Médica y Biología Molecular e Inmunología, Universidad de Sevilla, Avda. Sanchez Pizjuan S/N, 41009, Seville, Spain
| | - Juan Miguel Guerrero
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Avda. Manuel Siurot S/N, 41013, Seville, Spain; Departamento de Bioquímica Médica y Biología Molecular e Inmunología, Universidad de Sevilla, Avda. Sanchez Pizjuan S/N, 41009, Seville, Spain; Departamento de Bioquímica Clínica, Hospital Universitario Virgen del Rocío, Avda. Manuel Siurot S/N, 41013, Seville, Spain
| | - Patrocinio Molinero
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Avda. Manuel Siurot S/N, 41013, Seville, Spain; Departamento de Bioquímica Médica y Biología Molecular e Inmunología, Universidad de Sevilla, Avda. Sanchez Pizjuan S/N, 41009, Seville, Spain
| | - Patricia J Lardone
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Avda. Manuel Siurot S/N, 41013, Seville, Spain; Departamento de Bioquímica Médica y Biología Molecular e Inmunología, Universidad de Sevilla, Avda. Sanchez Pizjuan S/N, 41009, Seville, Spain.
| |
Collapse
|
17
|
Xu M, Gan T, Ning H, Wang L. MicroRNA Functions in Thymic Biology: Thymic Development and Involution. Front Immunol 2018; 9:2063. [PMID: 30254640 PMCID: PMC6141719 DOI: 10.3389/fimmu.2018.02063] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Accepted: 08/21/2018] [Indexed: 01/02/2023] Open
Abstract
During the entire processes of thymus organogenesis, maturation, and involution, gene regulation occurs post-transcriptionally via recently discovered microRNA (miRNA) transcripts. Numerous reports indicate that miRNAs may be involved in the construction of a normal thymic microenvironment, which constitutes a critical component to support T lymphocyte development. MiRNAs are also expressed in thymic stromal cells including thymic epithelial cells (TECs) during maturation and senescence. This review focuses on the function of miRNAs in thymic development and involution. A better understanding of these processes will provide new insights into the regulatory network of TECs and further comprehension of how genes control TECs to maintain the thymic microenvironment during thymus development and aging, thus supporting a normal cellular immune system.
Collapse
Affiliation(s)
- Minwen Xu
- First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Tao Gan
- Department of Biotechnology, Gannan Medical University, Ganzhou, China
| | - Huiting Ning
- Department of Biotechnology, Gannan Medical University, Ganzhou, China
| | - Liefeng Wang
- Department of Biotechnology, Gannan Medical University, Ganzhou, China
| |
Collapse
|
18
|
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: 4.3] [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
|
19
|
Ferrando-Martinez S, De Pablo-Bernal RS, De Luna-Romero M, De Ory SJ, Genebat M, Pacheco YM, Parras FJ, Montero M, Blanco JR, Gutierrez F, Santos J, Vidal F, Koup RA, Muñoz-Fernández MÁ, Leal M, Ruiz-Mateos E. Thymic Function Failure Is Associated With Human Immunodeficiency Virus Disease Progression. Clin Infect Dis 2018; 64:1191-1197. [PMID: 28158588 DOI: 10.1093/cid/cix095] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Accepted: 01/28/2017] [Indexed: 12/14/2022] Open
Abstract
Background Thymic function has been mainly analyzed with surrogate peripheral markers affected by peripheral T-cell expansion, making it difficult to assess the role of thymic failure in human immunodeficiency virus (HIV) disease progression. The assay of signal-joint/DβJβ T-cell rearrangement excision circles (sj/β-TREC ratio) overcomes this limitation but has only been assayed in small cohorts. Thus, the aim of this study was to determine the role of thymic function, measured by the sj/β-TREC ratio, on CD4 T-cell maintenance in prospective HIV cohorts that include patients with a wide age range and different immunological phenotypes. Methods Seven hundred seventy-four patients including typical progressors, long-term nonprogressors (LTNPs), and vertically HIV-infected subjects were analyzed. Thymic function was quantified in peripheral blood samples using the sj/β-TREC ratio. Associations between thymic function and CD4 T-cell dynamics and combination antiretroviral therapy (cART) onset were analyzed using linear, logistic, and Cox proportional hazard models. Results Thymic function failure (sj/β-TREC ratio <10) was independently associated with HIV progression. In agreement, patients with distinctive high CD4 T-cell levels and low progression rates (vertically HIV-infected patients and LTNPs, including HIV controllers) had significantly higher thymic function levels whereas patients with thymic function failure had lower CD4 T-cell levels, lower nadir, and faster CD4 T-cell decay. Conclusions This work establishes the relevance of thymic function, measured by sj/β-TREC ratio, in HIV disease progression by analyzing a large number of patients in 3 cohorts with different HIV disease progression phenotypes. These results support and help to understand the mechanisms underlying the rationale of early cART onset.
Collapse
Affiliation(s)
- Sara Ferrando-Martinez
- Immunology Laboratory, Vaccine Research Center, National Institute for Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Rebeca S De Pablo-Bernal
- Laboratory of Immunovirology, Institute of Biomedicine of Seville, Virgen del Rocío University Hospital/CSIC/University of Seville, Spain
| | - Marta De Luna-Romero
- Laboratory of Immunovirology, Institute of Biomedicine of Seville, Virgen del Rocío University Hospital/CSIC/University of Seville, Spain
| | - Santiago J De Ory
- Molecular Immunobiology Laboratory, General University Hospital Gregorio Marañon, Health Research Institute Gregorio Marañon, Spanish HIV HGM BioBank, Networking Research Center on Bioengineering, Biomaterials and Nanomedicine, Madrid, Spain
| | - Miguel Genebat
- Laboratory of Immunovirology, Institute of Biomedicine of Seville, Virgen del Rocío University Hospital/CSIC/University of Seville, Spain
| | - Yolanda M Pacheco
- Laboratory of Immunovirology, Institute of Biomedicine of Seville, Virgen del Rocío University Hospital/CSIC/University of Seville, Spain
| | - Francisco J Parras
- Infectious Disease Unit, General Universitary Hospital Gregorio Marañon, Madrid, Spain
| | - Marta Montero
- Infectious Disease Unit, Polytechnic and University Hospital La Fe, Valencia, Spain
| | - Jose Ramón Blanco
- Infectious Diseases Department, Hospital San Pedro, Center for Biomedical Research of La Rioja, Logrono, Spain
| | - Felix Gutierrez
- Hospital General de Elche and Universidad Miguel Hernández, Alicante, Spain
| | - Jesus Santos
- Infectious Diseases Unit, Virgen de la Victoria University Hospital, Instituto de Investigación Biomédica de Málaga, Spain
| | - Francisco Vidal
- Hospital Universitari de Tarragona Joan XXIII, IISPV, Universitat Rovira i Virgili, Spain
| | - Richard A Koup
- Immunology Laboratory, Vaccine Research Center, National Institute for Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - María Ángeles Muñoz-Fernández
- Molecular Immunobiology Laboratory, General University Hospital Gregorio Marañon, Health Research Institute Gregorio Marañon, Spanish HIV HGM BioBank, Networking Research Center on Bioengineering, Biomaterials and Nanomedicine, Madrid, Spain
| | - Manuel Leal
- Laboratory of Immunovirology, Institute of Biomedicine of Seville, Virgen del Rocío University Hospital/CSIC/University of Seville, Spain
| | - Ezequiel Ruiz-Mateos
- Laboratory of Immunovirology, Institute of Biomedicine of Seville, Virgen del Rocío University Hospital/CSIC/University of Seville, Spain
| |
Collapse
|
20
|
Wallace J, Lutgen V, Avasarala S, St Croix B, Winn RA, Al-Harthi L. Wnt7a induces a unique phenotype of monocyte-derived macrophages with lower phagocytic capacity and differential expression of pro- and anti-inflammatory cytokines. Immunology 2017; 153:203-213. [PMID: 28872671 DOI: 10.1111/imm.12830] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Revised: 08/24/2017] [Accepted: 08/26/2017] [Indexed: 12/25/2022] Open
Abstract
The variation of macrophage functions suggests the involvement of multiple signalling pathways in fine tuning their differentiation. Macrophages that originate from monocytes in the blood migrate to tissue in response to homeostatic or 'danger' signals and undergo substantial morphological and functional modifications to meet the needs of the dominant signals in the microenvironment. Wnts are secreted glycoproteins that play a significant role in organ and cell differentiation, yet their impact on monocyte differentiation is not clear. In this study, we assessed the role of Wnt1 and Wnt7a on the differentiation of monocytes and the subsequent phenotype and function of monocyte-derived macrophages (MDMs). We show that Wnt7a decreased the expression of CD14, CD11b, CD163 and CD206, whereas Wnt1 had no effect. The Wnt7a effect on CD11b was also observed in the brain and spleen of Wnt7a-/- adult brain mouse tissue and in embryonic Wnt7a-/- tissue. Wnt7a reduced the phagocytic capacity of M-MDMs, decreased interleukin-10 (IL-10) and IL-12 secretion and increased IL-6 secretion. Collectively, these findings demonstrate that Wnt7a generates an MDM phenotype with both pro-inflammatory and alternative MDM cytokine profiles and reduced phagocytic capacity. As such, Wnt7a can have a significant impact on macrophage responses in health and disease.
Collapse
Affiliation(s)
- Jennillee Wallace
- Department of Immunology and Microbiology, Rush University Medical Center, Chicago, IL, USA
| | - Victoria Lutgen
- Department of Immunology and Microbiology, Rush University Medical Center, Chicago, IL, USA
| | - Sreedevi Avasarala
- University of Illinois Cancer Center, University of Illinois at Chicago, Chicago, IL, USA
| | - Brad St Croix
- Center for Cancer Research (CCR), National Cancer Institute (NCI), Frederick, MD, USA
| | - Robert A Winn
- University of Illinois Cancer Center, University of Illinois at Chicago, Chicago, IL, USA
| | - Lena Al-Harthi
- Department of Immunology and Microbiology, Rush University Medical Center, Chicago, IL, USA
| |
Collapse
|
21
|
MicroRNAs Regulate Thymic Epithelium in Age-Related Thymic Involution via Down- or Upregulation of Transcription Factors. J Immunol Res 2017; 2017:2528957. [PMID: 29226156 PMCID: PMC5684555 DOI: 10.1155/2017/2528957] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Revised: 08/09/2017] [Accepted: 08/20/2017] [Indexed: 12/14/2022] Open
Abstract
Age-related thymic involution is primarily induced by defects in nonhematopoietic thymic epithelial cells (TECs). It is characterized by dysfunction of multiple transcription factors (TFs), such as p63 and FoxN1, and also involves other TEC-associated regulators, such as Aire. These TFs and regulators are controlled by complicated regulatory networks, in which microRNAs (miRNAs) act as a key player. miRNAs can either directly target the 3'-UTRs (untranslated regions) of the TFs to suppress TF expression or target TF inhibitors to reduce or increase TF inhibitor expression and thereby indirectly enhance or inhibit TF expression. Here, we review the current understanding and recent studies about how miRNAs are involved in age-related thymic involution via regulation of TEC-autonomous TFs. We also discuss potential strategies for targeting miRNAs to rejuvenate age-related declined thymic function.
Collapse
|
22
|
Marthandan S, Menzel U, Priebe S, Groth M, Guthke R, Platzer M, Hemmerich P, Kaether C, Diekmann S. Conserved genes and pathways in primary human fibroblast strains undergoing replicative and radiation induced senescence. Biol Res 2016; 49:34. [PMID: 27464526 PMCID: PMC4963952 DOI: 10.1186/s40659-016-0095-2] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Accepted: 07/19/2016] [Indexed: 01/01/2023] Open
Abstract
Background Cellular senescence is induced either internally, for example by replication exhaustion and cell division, or externally, for example by irradiation. In both cases, cellular damages accumulate which, if not successfully repaired, can result in senescence induction. Recently, we determined the transcriptional changes combined with the transition into replicative senescence in primary human fibroblast strains. Here, by γ-irradiation we induced premature cellular senescence in the fibroblast cell strains (HFF and MRC-5) and determined the corresponding transcriptional changes by high-throughput RNA sequencing. Results Comparing the transcriptomes, we found a high degree of similarity in differential gene expression in replicative as well as in irradiation induced senescence for both cell strains suggesting, in each cell strain, a common cellular response to error accumulation. On the functional pathway level, “Cell cycle” was the only pathway commonly down-regulated in replicative and irradiation-induced senescence in both fibroblast strains, confirming the tight link between DNA repair and cell cycle regulation. However, “DNA repair” and “replication” pathways were down-regulated more strongly in fibroblasts undergoing replicative exhaustion. We also retrieved genes and pathways in each of the cell strains specific for irradiation induced senescence. Conclusion We found the pathways associated with “DNA repair” and “replication” less stringently regulated in irradiation induced compared to replicative senescence. The strong regulation of these pathways in replicative senescence highlights the importance of replication errors for its induction. Electronic supplementary material The online version of this article (doi:10.1186/s40659-016-0095-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Shiva Marthandan
- Leibniz Institute for Age Research-Fritz Lipmann Institute e.V. (FLI), Beutenbergstrasse 11, 07745, Jena, Germany.
| | - Uwe Menzel
- Leibniz Institute for Natural Product Research and Infection Biology-Hans-Knöll-Institute e.V. (HKI), Jena, Germany
| | - Steffen Priebe
- Leibniz Institute for Natural Product Research and Infection Biology-Hans-Knöll-Institute e.V. (HKI), Jena, Germany
| | - Marco Groth
- Leibniz Institute for Age Research-Fritz Lipmann Institute e.V. (FLI), Beutenbergstrasse 11, 07745, Jena, Germany
| | - Reinhard Guthke
- Leibniz Institute for Natural Product Research and Infection Biology-Hans-Knöll-Institute e.V. (HKI), Jena, Germany
| | - Matthias Platzer
- Leibniz Institute for Age Research-Fritz Lipmann Institute e.V. (FLI), Beutenbergstrasse 11, 07745, Jena, Germany
| | - Peter Hemmerich
- Leibniz Institute for Age Research-Fritz Lipmann Institute e.V. (FLI), Beutenbergstrasse 11, 07745, Jena, Germany
| | - Christoph Kaether
- Leibniz Institute for Age Research-Fritz Lipmann Institute e.V. (FLI), Beutenbergstrasse 11, 07745, Jena, Germany
| | - Stephan Diekmann
- Leibniz Institute for Age Research-Fritz Lipmann Institute e.V. (FLI), Beutenbergstrasse 11, 07745, Jena, Germany
| |
Collapse
|
23
|
Li J, Cai D, Yao X, Zhang Y, Chen L, Jing P, Wang L, Wang Y. Protective Effect of Ginsenoside Rg1 on Hematopoietic Stem/Progenitor Cells through Attenuating Oxidative Stress and the Wnt/β-Catenin Signaling Pathway in a Mouse Model of d-Galactose-induced Aging. Int J Mol Sci 2016; 17:ijms17060849. [PMID: 27294914 PMCID: PMC4926383 DOI: 10.3390/ijms17060849] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Revised: 05/19/2016] [Accepted: 05/25/2016] [Indexed: 12/30/2022] Open
Abstract
Stem cell senescence is an important and current hypothesis accounting for organismal aging, especially the hematopoietic stem cell (HSC). Ginsenoside Rg1 is the main active pharmaceutical ingredient of ginseng, which is a traditional Chinese medicine. This study explored the protective effect of ginsenoside Rg1 on Sca-1⁺ hematopoietic stem/progenitor cells (HSC/HPCs) in a mouse model of d-galactose-induced aging. The mimetic aging mouse model was induced by continuous injection of d-gal for 42 days, and the C57BL/6 mice were respectively treated with ginsenoside Rg1, Vitamin E or normal saline after 7 days of d-gal injection. Compared with those in the d-gal administration alone group, ginsenoside Rg1 protected Sca-1⁺ HSC/HPCs by decreasing SA-β-Gal and enhancing the colony forming unit-mixture (CFU-Mix), and adjusting oxidative stress indices like reactive oxygen species (ROS), total anti-oxidant (T-AOC), superoxide dismutase (SOD), glutathione peroxidase (GSH-px) and malondialdehyde (MDA). In addition, ginsenoside Rg1 decreased β-catenin and c-Myc mRNA expression and enhanced the phosphorylation of GSK-3β. Moreover, ginsenoside Rg1 down-regulated advanced glycation end products (AGEs), 4-hydroxynonenal (4-HNE), phospho-histone H2A.X (r-H2A.X), 8-OHdG, p16(Ink4a), Rb, p21(Cip1/Waf1) and p53 in senescent Sca-1⁺ HSC/HPCs. Our findings indicated that ginsenoside Rg1 can improve the resistance of Sca-1⁺ HSC/HPCs in a mouse model of d-galactose-induced aging through the suppression of oxidative stress and excessive activation of the Wnt/β-catenin signaling pathway, and reduction of DNA damage response, p16(Ink4a)-Rb and p53-p21(Cip1/Waf1) signaling.
Collapse
Affiliation(s)
- Jing Li
- Laboratory of Stem Cells and Tissue Engineering, Chongqing Medical University, Chongqing 400016, China.
- Department of Pathophysiology, Chongqing Medical University, Chongqing 400016, China.
| | - Dachuan Cai
- Department of Infectious Diseases, the Second Affiliated Hospital, Chongqing Medical University, Chongqing 400016, China.
| | - Xin Yao
- Department of Pathophysiology, Chongqing Medical University, Chongqing 400016, China.
| | - Yanyan Zhang
- Laboratory of Stem Cells and Tissue Engineering, Chongqing Medical University, Chongqing 400016, China.
| | - Linbo Chen
- Laboratory of Stem Cells and Tissue Engineering, Chongqing Medical University, Chongqing 400016, China.
| | - Pengwei Jing
- Laboratory of Stem Cells and Tissue Engineering, Chongqing Medical University, Chongqing 400016, China.
| | - Lu Wang
- Laboratory of Stem Cells and Tissue Engineering, Chongqing Medical University, Chongqing 400016, China.
- Department of Histology and Embryology, Chongqing Medical University, Chongqing 400016, China.
| | - Yaping Wang
- Laboratory of Stem Cells and Tissue Engineering, Chongqing Medical University, Chongqing 400016, China.
- Department of Histology and Embryology, Chongqing Medical University, Chongqing 400016, China.
| |
Collapse
|