1
|
Nong C, Wu Z, Yang C, Xu W, Luo L, Zhou J, Shen L, Chen Y, Yuan Y, Hu G. Cdc42 improve SARS-CoV-2 spike protein-induced cellular senescence through activating of Wnt/β-Catenin signaling pathway. Front Cell Infect Microbiol 2024; 14:1449423. [PMID: 39559701 PMCID: PMC11570593 DOI: 10.3389/fcimb.2024.1449423] [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: 06/15/2024] [Accepted: 10/11/2024] [Indexed: 11/20/2024] Open
Abstract
Introduction SARS-CoV-2 infection drove senescent cells and the senescence-associated phenotypes were reported playing roles in disease progression, which contributes to severe COVID-19 and related sequelae. Cdc42 is involved in the regulation of cellular senescence. This study, aimed to investigate the mechanism of the SARS-CoV-2 spike protein regulating cellular senescence through Cdc42. Methods K18-hACE2 mice were infected with SARS-CoV-2 Omicron BA.4 or stimulated with spike protein through the airway, the senescent cells and Cdc42 expression in lung tissue were detected. Overexpression of spike protein or exogenous incubation of spike protein was used to simulate the induction of cellular senescence by spike protein. Mechanistic insights into the role of Cdc42 were mainly explored using Western Blot and qRT-PCR. Results Spike protein, SARS-CoV-2 infection related, accelerates cell aging by upregulating Cdc42 expression, which furtherly activated the Wnt/β-catenin signaling pathway. Conversely, treatment with ML141 in animal models, a Cdc42 inhibitor, reduced cellular senescence and ameliorated lung injury and inflammation. These results suggest that the upregulation of Cdc42 by the SARS-CoV-2 spike protein induces cellular senescence and enhances β-catenin nuclear translocation. Discussion This study provides insights into the mechanisms underlying cellular senescence induced by the SARS-CoV-2 spike protein, offering potential strategies to mitigate the inflammatory response and complications associated with COVID-19 in both the acute and long-term phases.
Collapse
Affiliation(s)
- Chunmei Nong
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, China
- Department of Respiratory and Critical Care Medicine, The Tenth Affiliated Hospital of Southern Medical University, Dongguan, Guangdong, China
- Department of Respiratory and Critical Care Medicine, Institute of Respiratory and Critical Care Medicine, Dongguan, Guangdong, China
| | - Zhenzhen Wu
- Department of Respiratory and Critical Care Medicine, The Tenth Affiliated Hospital of Southern Medical University, Dongguan, Guangdong, China
| | - Chan Yang
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Wei Xu
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Linyi Luo
- Intensive Care Unit, The Tenth Affiliated Hospital of Southern Medical University, Dongguan, Guangdong, China
| | - Jianping Zhou
- Department of Thoracic Surgery, The Tenth Affiliated Hospital of Southern Medical University, Dongguan, Guangdong, China
| | - Lihan Shen
- Intensive Care Unit, The Tenth Affiliated Hospital of Southern Medical University, Dongguan, Guangdong, China
| | - Yinghua Chen
- Dongguan People’s Hospital Biobank, Clinical Research Center, The Tenth Affiliated Hospital of Southern Medical University, Dongguan, Guangdong, China
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Yaoqin Yuan
- Intensive Care Unit, The Tenth Affiliated Hospital of Southern Medical University, Dongguan, Guangdong, China
| | - Guodong Hu
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, China
- Department of Respiratory and Critical Care Medicine, The Tenth Affiliated Hospital of Southern Medical University, Dongguan, Guangdong, China
- Department of Respiratory and Critical Care Medicine, Institute of Respiratory and Critical Care Medicine, Dongguan, Guangdong, China
| |
Collapse
|
2
|
Matteini F, Montserrat‐Vazquez S, Florian MC. Rejuvenating aged stem cells: therapeutic strategies to extend health and lifespan. FEBS Lett 2024; 598:2776-2787. [PMID: 38604982 PMCID: PMC11586596 DOI: 10.1002/1873-3468.14865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 02/03/2024] [Accepted: 03/07/2024] [Indexed: 04/13/2024]
Abstract
Aging is associated with a global decline in stem cell function. To date, several strategies have been proposed to rejuvenate aged stem cells: most of these result in functional improvement of the tissue where the stem cells reside, but the impact on the lifespan of the whole organism has been less clearly established. Here, we review some of the most recent work dealing with interventions that improve the regenerative capacity of aged somatic stem cells in mammals and that might have important translational possibilities. Overall, we underscore that somatic stem cell rejuvenation represents a strategy to improve tissue homeostasis upon aging and present some recent approaches with the potential to affect health span and lifespan of the whole organism.
Collapse
Affiliation(s)
- Francesca Matteini
- Stem Cell Aging Group, Regenerative Medicine ProgramThe Bellvitge Institute for Biomedical Research (IDIBELL)BarcelonaSpain
- Program for Advancing the Clinical Translation of Regenerative Medicine of Catalonia (P‐CMR[C])BarcelonaSpain
| | - Sara Montserrat‐Vazquez
- Stem Cell Aging Group, Regenerative Medicine ProgramThe Bellvitge Institute for Biomedical Research (IDIBELL)BarcelonaSpain
- Program for Advancing the Clinical Translation of Regenerative Medicine of Catalonia (P‐CMR[C])BarcelonaSpain
| | - M. Carolina Florian
- Stem Cell Aging Group, Regenerative Medicine ProgramThe Bellvitge Institute for Biomedical Research (IDIBELL)BarcelonaSpain
- Program for Advancing the Clinical Translation of Regenerative Medicine of Catalonia (P‐CMR[C])BarcelonaSpain
- Center for Networked Biomedical Research on Bioengineering, Biomaterials and Nanomedicine (CIBER‐BBN)MadridSpain
- The Catalan Institution for Research and Advanced Studies (ICREA)BarcelonaSpain
| |
Collapse
|
3
|
Zheng Z, Li J, Liu T, Fan Y, Zhai QC, Xiong M, Wang QR, Sun X, Zheng QW, Che S, Jiang B, Zheng Q, Wang C, Liu L, Ping J, Wang S, Gao DD, Ye J, Yang K, Zuo Y, Ma S, Yang YG, Qu J, Zhang F, Jia P, Liu GH, Zhang W. DNA methylation clocks for estimating biological age in Chinese cohorts. Protein Cell 2024; 15:575-593. [PMID: 38482631 PMCID: PMC11259550 DOI: 10.1093/procel/pwae011] [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: 11/11/2023] [Accepted: 01/10/2024] [Indexed: 07/21/2024] Open
Abstract
Epigenetic clocks are accurate predictors of human chronological age based on the analysis of DNA methylation (DNAm) at specific CpG sites. However, a systematic comparison between DNA methylation data and other omics datasets has not yet been performed. Moreover, available DNAm age predictors are based on datasets with limited ethnic representation. To address these knowledge gaps, we generated and analyzed DNA methylation datasets from two independent Chinese cohorts, revealing age-related DNAm changes. Additionally, a DNA methylation aging clock (iCAS-DNAmAge) and a group of DNAm-based multi-modal clocks for Chinese individuals were developed, with most of them demonstrating strong predictive capabilities for chronological age. The clocks were further employed to predict factors influencing aging rates. The DNAm aging clock, derived from multi-modal aging features (compositeAge-DNAmAge), exhibited a close association with multi-omics changes, lifestyles, and disease status, underscoring its robust potential for precise biological age assessment. Our findings offer novel insights into the regulatory mechanism of age-related DNAm changes and extend the application of the DNAm clock for measuring biological age and aging pace, providing the basis for evaluating aging intervention strategies.
Collapse
Affiliation(s)
- Zikai Zheng
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jiaming Li
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Tianzi Liu
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing 100101, China
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yanling Fan
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing 100101, China
| | - Qiao-Cheng Zhai
- Division of Orthopaedics, Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou 324000, China
- The Joint Innovation Center for Engineering in Medicine, Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou 324000, China
| | - Muzhao Xiong
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qiao-Ran Wang
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiaoyan Sun
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qi-Wen Zheng
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing 100101, China
| | - Shanshan Che
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Beier Jiang
- The Joint Innovation Center for Engineering in Medicine, Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou 324000, China
| | - Quan Zheng
- The Joint Innovation Center for Engineering in Medicine, Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou 324000, China
| | - Cui Wang
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Lixiao Liu
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jiale Ping
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Si Wang
- Advanced Innovation Center for Human Brain Protection, and National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing 100053, China
- Aging Translational Medicine Center, International Center for Aging and Cancer, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
- Aging Biomarker Consortium, Beijing 100101, China
| | - Dan-Dan Gao
- The Joint Innovation Center for Engineering in Medicine, Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou 324000, China
| | - Jinlin Ye
- The Joint Innovation Center for Engineering in Medicine, Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou 324000, China
| | - Kuan Yang
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yuesheng Zuo
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Shuai Ma
- Aging Biomarker Consortium, Beijing 100101, China
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Yun-Gui Yang
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jing Qu
- University of Chinese Academy of Sciences, Beijing 100049, China
- Aging Biomarker Consortium, Beijing 100101, China
- Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Feng Zhang
- Division of Orthopaedics, Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou 324000, China
| | - Peilin Jia
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- National Genomics Data Center, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing 100101, China
| | - Guang-Hui Liu
- University of Chinese Academy of Sciences, Beijing 100049, China
- Advanced Innovation Center for Human Brain Protection, and National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing 100053, China
- Aging Translational Medicine Center, International Center for Aging and Cancer, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
- Aging Biomarker Consortium, Beijing 100101, China
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Weiqi Zhang
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Aging Biomarker Consortium, Beijing 100101, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China
| |
Collapse
|
4
|
Luan S, Zhang L, Cheng X, Wang Y, Feng Q, Wei L, Jiang F, Liu J. The ability and optimal cutoff value of serum cell division cycle 42 in estimating major adverse cardiac event in STEMI patients treated with percutaneous coronary intervention. Heart Vessels 2024; 39:277-287. [PMID: 38153423 DOI: 10.1007/s00380-023-02350-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 10/12/2023] [Indexed: 12/29/2023]
Abstract
Cell division cycle 42 (CDC42) regulates cholesterol efflux, chronic inflammation, and reendothelialization in various atherosclerotic diseases. This study aimed to investigate the correlation of serum CDC42 with myocardial injury indicators and major adverse cardiac event (MACE) in ST-elevation myocardial infarction (STEMI) patients who were treated with percutaneous coronary intervention (PCI). In 250 STEMI patients about to receive PCI, serum samples were collected at enrollment before PCI treatment, and the serum samples were also obtained from 100 healthy controls (HCs) at enrollment. Serum CDC42 was detected by enzyme-linked immunosorbent assay. Serum CDC42 was decreased (versus HCs, P < 0.001) and negatively correlated with diabetes mellitus (P = 0.017), multivessel disease (P = 0.016), cardiac troponin I (P < 0.001), creatine kinase MB (P = 0.012), stent diameter ≥ 3.5 mm (P = 0.039), white blood cell (P < 0.001), low-density lipoprotein cholesterol (P = 0.049), and C-reactive protein (P < 0.001) in STEMI patients. Besides, 29 (11.6%) STEMI patients experienced MACE. The 1-year, 2-year, and 3-year accumulating MACE rates were 7.5%, 17.3%, and 19.3%, accordingly. Serum CDC42 was reduced in STEMI patients who experienced MACE compared to those who did not (P = 0.001). Serum CDC42 ≥ 250 pg/mL, ≥ 400 pg/mL, ≥ 700 pg/mL (cut by near integer value of 1/4th quartile, median, and 3/4th quartile) were associated with decreased accumulating MACE rates in STEMI patients (all P < 0.050). Notably, serum CDC42 ≥ 250 pg/mL (hazard ratio = 0.435, P = 0.031) was independently related to reduced accumulating MACE risk in STEMI patients. A serum CDC42 level of ≥ 250 pg/mL well predicts decreased MACE risk in STEMI patients who are treated with PCI.
Collapse
Affiliation(s)
- Shaohua Luan
- Department of Cardiology, HanDan Central Hospital, No.15 Zhonghua Road, Handan, 056001, China
| | - Lei Zhang
- Department of Cardiology, HanDan Central Hospital, No.15 Zhonghua Road, Handan, 056001, China.
| | - Xiaodan Cheng
- Department of Cardiology, HanDan Central Hospital, No.15 Zhonghua Road, Handan, 056001, China
| | - Yuanyuan Wang
- Department of Cardiology, HanDan Central Hospital, No.15 Zhonghua Road, Handan, 056001, China
| | - Qiang Feng
- Department of Cardiology, HanDan Central Hospital, No.15 Zhonghua Road, Handan, 056001, China
| | - Lei Wei
- Department of Cardiovascular Surgery, Shanxi Provincial People's Hospital, Taiyuan, 030032, China
| | - Fan Jiang
- School of Environment and Health, Yanching Institute of Technology, Langfang, 065201, China
| | - Jinjun Liu
- Department of Cardiology, HanDan Central Hospital, No.15 Zhonghua Road, Handan, 056001, China
| |
Collapse
|
5
|
Allegra A, Caserta S, Mirabile G, Gangemi S. Aging and Age-Related Epigenetic Drift in the Pathogenesis of Leukemia and Lymphomas: New Therapeutic Targets. Cells 2023; 12:2392. [PMID: 37830606 PMCID: PMC10572300 DOI: 10.3390/cells12192392] [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: 09/04/2023] [Revised: 09/24/2023] [Accepted: 09/28/2023] [Indexed: 10/14/2023] Open
Abstract
One of the traits of cancer cells is abnormal DNA methylation patterns. The idea that age-related epigenetic changes may partially explain the increased risk of cancer in the elderly is based on the observation that aging is also accompanied by comparable changes in epigenetic patterns. Lineage bias and decreased stem cell function are signs of hematopoietic stem cell compartment aging. Additionally, aging in the hematopoietic system and the stem cell niche have a role in hematopoietic stem cell phenotypes linked with age, such as leukemia and lymphoma. Understanding these changes will open up promising pathways for therapies against age-related disorders because epigenetic mechanisms are reversible. Additionally, the development of high-throughput epigenome mapping technologies will make it possible to identify the "epigenomic identity card" of every hematological disease as well as every patient, opening up the possibility of finding novel molecular biomarkers that can be used for diagnosis, prediction, and prognosis.
Collapse
Affiliation(s)
- Alessandro Allegra
- Division of Hematology, Department of Human Pathology in Adulthood and Childhood “Gaetano Barresi”, University of Messina, Via Consolare Valeria, 98125 Messina, Italy; (S.C.); (G.M.)
| | - Santino Caserta
- Division of Hematology, Department of Human Pathology in Adulthood and Childhood “Gaetano Barresi”, University of Messina, Via Consolare Valeria, 98125 Messina, Italy; (S.C.); (G.M.)
| | - Giuseppe Mirabile
- Division of Hematology, Department of Human Pathology in Adulthood and Childhood “Gaetano Barresi”, University of Messina, Via Consolare Valeria, 98125 Messina, Italy; (S.C.); (G.M.)
| | - Sebastiano Gangemi
- Allergy and Clinical Immunology Unit, Department of Clinical and Experimental Medicine, University of Messina, Via Consolare Valeria, 98125 Messina, Italy;
| |
Collapse
|
6
|
Role of a small GTPase Cdc42 in aging and age-related diseases. Biogerontology 2023; 24:27-46. [PMID: 36598630 DOI: 10.1007/s10522-022-10008-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 12/13/2022] [Indexed: 01/05/2023]
Abstract
A small GTPase, Cdc42 is evolutionarily one of the most ancient members of the Rho family, which is ubiquitously expressed and involved in a wide range of fundamental cellular functions. The crucial role of Cdc42 includes regulation of the actin cytoskeleton, cell polarity, morphology and migration, endocytosis and exocytosis, cell cycle, and proliferation in many different cell types. Many studies have provided compelling yet contradicting evidence that Cdc42 dysregulation plays an important role in cellular and tissue aging. Furthermore, Cdc42 is a critical factor in the development and progression of aging-related pathologies, such as neurodegenerative and cardiovascular disorders, diabetes type 2, and aging-related disorders of the joints and bones, and the inhibition of the Cdc42 demonstrates potentially significant therapeutic and anti-aging effects in animal models of aging and disease. However, regulation of Cdc42 expression and activity is very complex and depends on many factors, such as the origin and complexity of the tissues, hormonal status, etc. Therefore, this review is focused on current advances in understanding the underlying cellular and molecular mechanisms associated with Cdc42 activity and regulation of senescence in different cell types since they may provide a foundation for novel therapeutic strategies and targeted drugs to reverse the aging process and treat aging-associated disorders.
Collapse
|
7
|
Montserrat-Vazquez S, Ali NJ, Matteini F, Lozano J, Zhaowei T, Mejia-Ramirez E, Marka G, Vollmer A, Soller K, Sacma M, Sakk V, Mularoni L, Mallm JP, Plass M, Zheng Y, Geiger H, Florian MC. Transplanting rejuvenated blood stem cells extends lifespan of aged immunocompromised mice. NPJ Regen Med 2022; 7:78. [PMID: 36581635 PMCID: PMC9800381 DOI: 10.1038/s41536-022-00275-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 12/16/2022] [Indexed: 12/30/2022] Open
Abstract
One goal of regenerative medicine is to rejuvenate tissues and extend lifespan by restoring the function of endogenous aged stem cells. However, evidence that somatic stem cells can be targeted in vivo to extend lifespan is still lacking. Here, we demonstrate that after a short systemic treatment with a specific inhibitor of the small RhoGTPase Cdc42 (CASIN), transplanting aged hematopoietic stem cells (HSCs) from treated mice is sufficient to extend the healthspan and lifespan of aged immunocompromised mice without additional treatment. In detail, we show that systemic CASIN treatment improves strength and endurance of aged mice by increasing the myogenic regenerative potential of aged skeletal muscle stem cells. Further, we show that CASIN modifies niche localization and H4K16ac polarity of HSCs in vivo. Single-cell profiling reveals changes in HSC transcriptome, which underlie enhanced lymphoid and regenerative capacity in serial transplantation assays. Overall, we provide proof-of-concept evidence that a short systemic treatment to decrease Cdc42 activity improves the regenerative capacity of different endogenous aged stem cells in vivo, and that rejuvenated HSCs exert a broad systemic effect sufficient to extend murine health- and lifespan.
Collapse
Affiliation(s)
- Sara Montserrat-Vazquez
- grid.417656.7Stem Cell Aging Group, Regenerative Medicine Program, The Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet de Llobregat, Barcelona, Spain ,grid.417656.7Program for advancing the Clinical Translation of Regenerative Medicine of Catalonia, P-CMR[C], L’Hospitalet de Llobregat, Barcelona, Spain
| | - Noelle J. Ali
- grid.6582.90000 0004 1936 9748Institute of Molecular Medicine, University of Ulm, Ulm, Germany
| | - Francesca Matteini
- grid.417656.7Stem Cell Aging Group, Regenerative Medicine Program, The Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet de Llobregat, Barcelona, Spain ,grid.417656.7Program for advancing the Clinical Translation of Regenerative Medicine of Catalonia, P-CMR[C], L’Hospitalet de Llobregat, Barcelona, Spain
| | - Javier Lozano
- grid.417656.7Stem Cell Aging Group, Regenerative Medicine Program, The Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet de Llobregat, Barcelona, Spain ,grid.417656.7Program for advancing the Clinical Translation of Regenerative Medicine of Catalonia, P-CMR[C], L’Hospitalet de Llobregat, Barcelona, Spain
| | - Tu Zhaowei
- grid.239573.90000 0000 9025 8099Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
| | - Eva Mejia-Ramirez
- grid.417656.7Stem Cell Aging Group, Regenerative Medicine Program, The Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet de Llobregat, Barcelona, Spain ,grid.417656.7Program for advancing the Clinical Translation of Regenerative Medicine of Catalonia, P-CMR[C], L’Hospitalet de Llobregat, Barcelona, Spain ,grid.512890.7Center for Networked Biomedical Research on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid, Spain
| | - Gina Marka
- grid.6582.90000 0004 1936 9748Institute of Molecular Medicine, University of Ulm, Ulm, Germany
| | - Angelika Vollmer
- grid.6582.90000 0004 1936 9748Institute of Molecular Medicine, University of Ulm, Ulm, Germany
| | - Karin Soller
- grid.6582.90000 0004 1936 9748Institute of Molecular Medicine, University of Ulm, Ulm, Germany
| | - Mehmet Sacma
- grid.6582.90000 0004 1936 9748Institute of Molecular Medicine, University of Ulm, Ulm, Germany
| | - Vadim Sakk
- grid.6582.90000 0004 1936 9748Institute of Molecular Medicine, University of Ulm, Ulm, Germany
| | - Loris Mularoni
- grid.417656.7Program for advancing the Clinical Translation of Regenerative Medicine of Catalonia, P-CMR[C], L’Hospitalet de Llobregat, Barcelona, Spain
| | | | - Mireya Plass
- grid.417656.7Program for advancing the Clinical Translation of Regenerative Medicine of Catalonia, P-CMR[C], L’Hospitalet de Llobregat, Barcelona, Spain ,grid.512890.7Center for Networked Biomedical Research on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid, Spain ,grid.417656.7Gene Regulation of Cell Identity Group, Regenerative Medicine Program, The Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet de Llobregat, Barcelona, Spain
| | - Yi Zheng
- grid.239573.90000 0000 9025 8099Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
| | - Hartmut Geiger
- grid.6582.90000 0004 1936 9748Institute of Molecular Medicine, University of Ulm, Ulm, Germany
| | - M. Carolina Florian
- grid.417656.7Stem Cell Aging Group, Regenerative Medicine Program, The Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet de Llobregat, Barcelona, Spain ,grid.417656.7Program for advancing the Clinical Translation of Regenerative Medicine of Catalonia, P-CMR[C], L’Hospitalet de Llobregat, Barcelona, Spain ,grid.512890.7Center for Networked Biomedical Research on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid, Spain
| |
Collapse
|
8
|
Emmrich S, Trapp A, Tolibzoda Zakusilo F, Straight ME, Ying AK, Tyshkovskiy A, Mariotti M, Gray S, Zhang Z, Drage MG, Takasugi M, Klusmann J, Gladyshev VN, Seluanov A, Gorbunova V. Characterization of naked mole-rat hematopoiesis reveals unique stem and progenitor cell patterns and neotenic traits. EMBO J 2022; 41:e109694. [PMID: 35694726 PMCID: PMC9340489 DOI: 10.15252/embj.2021109694] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 05/03/2022] [Accepted: 05/06/2022] [Indexed: 12/13/2022] Open
Abstract
Naked mole rats (NMRs) are the longest-lived rodents yet their stem cell characteristics remain enigmatic. Here, we comprehensively mapped the NMR hematopoietic landscape and identified unique features likely contributing to longevity. Adult NMRs form red blood cells in spleen and marrow, which comprise a myeloid bias toward granulopoiesis together with decreased B-lymphopoiesis. Remarkably, youthful blood and marrow single-cell transcriptomes and cell compositions are largely maintained until at least middle age. Similar to primates, the primitive stem and progenitor cell (HSPC) compartment is marked by CD34 and THY1. Stem cell polarity is seen for Tubulin but not CDC42, and is not lost until 12 years of age. HSPC respiration rates are as low as in purified human stem cells, in concert with a strong expression signature for fatty acid metabolism. The pool of quiescent stem cells is higher than in mice, and the cell cycle of hematopoietic cells is prolonged. By characterizing the NMR hematopoietic landscape, we identified resilience phenotypes such as an increased quiescent HSPC compartment, absence of age-related decline, and neotenic traits likely geared toward longevity.
Collapse
Affiliation(s)
| | | | | | | | - Albert K Ying
- Division of GeneticsDepartment of MedicineBrigham and Women’s HospitalHarvard Medical SchoolBostonMAUSA
| | - Alexander Tyshkovskiy
- Division of GeneticsDepartment of MedicineBrigham and Women’s HospitalHarvard Medical SchoolBostonMAUSA
| | - Marco Mariotti
- Division of GeneticsDepartment of MedicineBrigham and Women’s HospitalHarvard Medical SchoolBostonMAUSA
| | - Spencer Gray
- Department of BiologyUniversity of RochesterRochesterNYUSA
| | - Zhihui Zhang
- Department of BiologyUniversity of RochesterRochesterNYUSA
| | - Michael G Drage
- Pathology and Laboratory MedicineUniversity of Rochester Medical CenterRochesterNYUSA
| | | | - Jan‐Henning Klusmann
- Pediatric Hematology and OncologyMartin‐Luther‐University Halle‐WittenbergHalleGermany
| | - Vadim N Gladyshev
- Division of GeneticsDepartment of MedicineBrigham and Women’s HospitalHarvard Medical SchoolBostonMAUSA
| | | | - Vera Gorbunova
- Department of BiologyUniversity of RochesterRochesterNYUSA
| |
Collapse
|
9
|
Amoah A, Keller A, Emini R, Hoenicka M, Liebold A, Vollmer A, Eiwen K, Soller K, Sakk V, Zheng Y, Florian MC, Geiger H. Aging of human hematopoietic stem cells is linked to changes in Cdc42 activity. Haematologica 2021; 107:393-402. [PMID: 33440922 PMCID: PMC8804569 DOI: 10.3324/haematol.2020.269670] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Indexed: 11/23/2022] Open
Abstract
In this study, we characterize age-related phenotypes of human hematopoietic stem cells (HSC). We report increased frequencies of HSC, hematopoietic progenitor cells and lineage negative cells in the elderly but a decreased frequency of multi-lymphoid progenitors. Aged human HSC further exhibited a delay in initiating division ex vivo though without changes in their division kinetics. The activity of the small RhoGTPase Cdc42 was elevated in aged human hematopoietic cells and we identified a positive correlation between Cdc42 activity and the frequency of HSC upon aging. The frequency of human HSC polar for polarity proteins was, similar to the mouse, decreased upon aging, while inhibition of Cdc42 activity via the specific pharmacological inhibitor of Cdc42 activity, CASIN, resulted in re-polarization of aged human HSC with respect to Cdc42. Elevated activity of Cdc42 in aged HSC thus contributed to age-related changes in HSC. Xenotransplant, using NBSGW mice as recipients, showed elevated chimerism in recipients of aged compared to young HSC. Aged HSC treated with CASIN ex vivo displayed an engraftment profile similar to recipients of young HSC. Taken together, our work reveals strong evidence for a role of elevated Cdc42 activity in driving aging of human HSC, and similar to mice, this presents a likely possibility for attenuation of aging in human HSC.
Collapse
Affiliation(s)
- Amanda Amoah
- Institute of Molecular Medicine, Ulm University, Meyerhofstrasse, 89081 Ulm
| | - Anja Keller
- Institute of Molecular Medicine, Ulm University, Meyerhofstrasse, 89081 Ulm
| | - Ramiz Emini
- Department of Cardiothoracic and Vascular Surgery, Ulm University Hospital, Ulm
| | - Markus Hoenicka
- Department of Cardiothoracic and Vascular Surgery, Ulm University Hospital, Ulm
| | - Andreas Liebold
- Department of Cardiothoracic and Vascular Surgery, Ulm University Hospital, Ulm
| | - Angelika Vollmer
- Institute of Molecular Medicine, Ulm University, Meyerhofstrasse, 89081 Ulm
| | - Karina Eiwen
- Institute of Molecular Medicine, Ulm University, Meyerhofstrasse, 89081 Ulm
| | - Karin Soller
- Institute of Molecular Medicine, Ulm University, Meyerhofstrasse, 89081 Ulm
| | - Vadim Sakk
- Institute of Molecular Medicine, Ulm University, Meyerhofstrasse, 89081 Ulm
| | - Yi Zheng
- Cincinnati Children's Hospital Medical Center and University of Cincinnati
| | | | - Hartmut Geiger
- Institute of Molecular Medicine, Ulm University, Meyerhofstrasse, 89081 Ulm.
| |
Collapse
|
10
|
Florian MC, Leins H, Gobs M, Han Y, Marka G, Soller K, Vollmer A, Sakk V, Nattamai KJ, Rayes A, Zhao X, Setchell K, Mulaw M, Wagner W, Zheng Y, Geiger H. Inhibition of Cdc42 activity extends lifespan and decreases circulating inflammatory cytokines in aged female C57BL/6 mice. Aging Cell 2020; 19:e13208. [PMID: 32755011 PMCID: PMC7511875 DOI: 10.1111/acel.13208] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 04/27/2020] [Accepted: 07/07/2020] [Indexed: 12/12/2022] Open
Abstract
Cdc42 is a small RhoGTPase regulating multiple functions in eukaryotic cells. The activity of Cdc42 is significantly elevated in several tissues of aged mice, while the Cdc42 gain‐of‐activity mouse model presents with a premature aging‐like phenotype and with decreased lifespan. These data suggest a causal connection between elevated activity of Cdc42, aging, and reduced lifespan. Here, we demonstrate that systemic treatment of aged (75‐week‐old) female C57BL/6 mice with a Cdc42 activity‐specific inhibitor (CASIN) for 4 consecutive days significantly extends average and maximum lifespan. Moreover, aged CASIN‐treated animals displayed a youthful level of the aging‐associated cytokines IL‐1β, IL‐1α, and INFγ in serum and a significantly younger epigenetic clock as based on DNA methylation levels in blood cells. Overall, our data show that systemic administration of CASIN to reduce Cdc42 activity in aged mice extends murine lifespan.
Collapse
Affiliation(s)
- Maria Carolina Florian
- Program of Regenerative Medicine, IDIBELL, Barcelona, Spain.,Institute of Molecular Medicine and Stem Cell Aging, Ulm University, Ulm, Germany
| | - Hanna Leins
- Institute of Molecular Medicine and Stem Cell Aging, Ulm University, Ulm, Germany
| | - Michael Gobs
- Helmholtz-Institute for Biomedical Engineering, Stem Cell Biology and Cellular Engineering, RWTH Aachen University Medical School, Aachen, Germany
| | - Yang Han
- Helmholtz-Institute for Biomedical Engineering, Stem Cell Biology and Cellular Engineering, RWTH Aachen University Medical School, Aachen, Germany
| | - Gina Marka
- Institute of Molecular Medicine and Stem Cell Aging, Ulm University, Ulm, Germany
| | - Karin Soller
- Institute of Molecular Medicine and Stem Cell Aging, Ulm University, Ulm, Germany
| | - Angelika Vollmer
- Institute of Molecular Medicine and Stem Cell Aging, Ulm University, Ulm, Germany
| | - Vadim Sakk
- Institute of Molecular Medicine and Stem Cell Aging, Ulm University, Ulm, Germany
| | - Kalpana J Nattamai
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio, USA
| | - Ahmad Rayes
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio, USA
| | - Xueheng Zhao
- Division of Pathology, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio, USA
| | - Kenneth Setchell
- Division of Pathology, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio, USA
| | - Medhanie Mulaw
- Institute of Experimental Cancer Research, Medical Faculty, University of Ulm, Ulm, Germany
| | - Wolfgang Wagner
- Helmholtz-Institute for Biomedical Engineering, Stem Cell Biology and Cellular Engineering, RWTH Aachen University Medical School, Aachen, Germany
| | - Yi Zheng
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio, USA
| | - Hartmut Geiger
- Institute of Molecular Medicine and Stem Cell Aging, Ulm University, Ulm, Germany
| |
Collapse
|
11
|
Ermogenous C, Green C, Jackson T, Ferguson M, Lord JM. Treating age-related multimorbidity: the drug discovery challenge. Drug Discov Today 2020; 25:1403-1415. [DOI: 10.1016/j.drudis.2020.06.016] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 05/19/2020] [Accepted: 06/16/2020] [Indexed: 12/12/2022]
|
12
|
Acute Myeloid Leukemia: Aging and Epigenetics. Cancers (Basel) 2019; 12:cancers12010103. [PMID: 31906064 PMCID: PMC7017261 DOI: 10.3390/cancers12010103] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 12/23/2019] [Accepted: 12/27/2019] [Indexed: 12/15/2022] Open
Abstract
Acute myeloid leukemia (AML) is an aggressive hematological disorder mainly affecting people of older age. AML initiation is primarily attributed to mutations in crucial cellular regulators such as epigenetic factors, transcription factors, and signaling genes. AML’s aggressiveness and responsiveness to treatment depends on the specific cell type where leukemia first arose. Aged hematopoietic cells are often genetically and/or epigenetically altered and, therefore, present with a completely different cellular context for AML development compared to young cells. In this review, we summarize key aspects of AML development, and we focus, in particular, on the contribution of cellular aging to leukemogenesis and on current treatment options for elderly AML patients. Hematological disorders and leukemia grow exponentially with age. So far, with conventional induction therapy, many elderly patients experience a very poor overall survival rate requiring substantial social and medical costs during the relatively few remaining months of life. The global population’s age is increasing rapidly without an acceptable equal growth in therapeutic management of AML in the elderly; this is in sharp contrast to the increase in successful therapies for leukemia in younger patients. Therefore, a focus on the understanding of the biology of aging in the hematopoietic system, the development of appropriate research models, and new therapeutic approaches are urged.
Collapse
|
13
|
Mammoto T, Torisawa YS, Muyleart M, Hendee K, Anugwom C, Gutterman D, Mammoto A. Effects of age-dependent changes in cell size on endothelial cell proliferation and senescence through YAP1. Aging (Albany NY) 2019; 11:7051-7069. [PMID: 31487690 PMCID: PMC6756888 DOI: 10.18632/aging.102236] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2019] [Accepted: 08/21/2019] [Indexed: 04/14/2023]
Abstract
Angiogenesis - the growth of new blood capillaries- is impaired in aging animals. Biophysical factors such as changes in cell size control endothelial cell (EC) proliferation and differentiation. However, the effects of aging on EC size and the mechanism by which changes in cell size control age-dependent decline in EC proliferation are largely unknown. Here, we have demonstrated that aged ECs are larger than young ECs and that age-dependent increases in EC size control EC proliferation and senescence through CDC42-Yes-associated protein (YAP1) signaling. Reduction of aged EC size by culturing on single-cell sized fibronectin-coated smaller islands decreases CDC42 activity, stimulates YAP1 nuclear translocation and attenuates EC senescence. Stimulation of YAP1 or inhibition of CDC42 activity in aged ECs also restores blood vessel formation. Age-dependent changes in EC size and/or CDC42 and YAP1 activity may be the key control point of age-related decline in angiogenesis.
Collapse
Affiliation(s)
- Tadanori Mammoto
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Yu-Suke Torisawa
- Hakubi Center for Advanced Research, Kyoto University, Kyoto 615-8540, Japan
- Department of Micro Engineering, Kyoto University, Kyoto 615-8540, Japan
| | - Megan Muyleart
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Kathryn Hendee
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Charles Anugwom
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - David Gutterman
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Akiko Mammoto
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| |
Collapse
|
14
|
Bonaventura A, Montecucco F, Dallegri F, Carbone F, Lüscher TF, Camici GG, Liberale L. Novel findings in neutrophil biology and their impact on cardiovascular disease. Cardiovasc Res 2019; 115:1266-1285. [PMID: 30918936 DOI: 10.1093/cvr/cvz084] [Citation(s) in RCA: 117] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/30/2023] Open
Affiliation(s)
- Aldo Bonaventura
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, Genoa, Italy
- Division of Cardiology, Department of Internal Medicine, Pauley Heart Center, Virginia Commonwealth University, Richmond, VA, USA
| | - Fabrizio Montecucco
- First Clinic of Internal Medicine, Department of Internal Medicine and Centre of Excellence for Biomedical Research (CEBR), University of Genoa, 6 viale Benedetto XV, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino Genoa – Italian Cardiovascular Network, 10 Largo Benzi, Genoa, Italy
| | - Franco Dallegri
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino Genoa – Italian Cardiovascular Network, 10 Largo Benzi, Genoa, Italy
| | - Federico Carbone
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, Genoa, Italy
| | - Thomas F Lüscher
- Center for Molecular Cardiology, University of Zürich, Wagistrasse 12, Schlieren, Switzerland
- Heart Division, Royal Brompton and Harefield Hospitals and Imperial College, London, UK
| | - Giovanni G Camici
- Center for Molecular Cardiology, University of Zürich, Wagistrasse 12, Schlieren, Switzerland
- University Heart Center, University Hospital Zürich, Rämistrasse 100, Zürich, Switzerland
- Department of Research and Education, University Hospital Zürich, Rämistrasse 100, Zürich, Switzerland
| | - Luca Liberale
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, Genoa, Italy
- Center for Molecular Cardiology, University of Zürich, Wagistrasse 12, Schlieren, Switzerland
| |
Collapse
|
15
|
Fransquet PD, Wrigglesworth J, Woods RL, Ernst ME, Ryan J. The epigenetic clock as a predictor of disease and mortality risk: a systematic review and meta-analysis. Clin Epigenetics 2019; 11:62. [PMID: 30975202 PMCID: PMC6458841 DOI: 10.1186/s13148-019-0656-7] [Citation(s) in RCA: 193] [Impact Index Per Article: 32.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2018] [Accepted: 03/25/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Ageing is one of the principal risk factors for many chronic diseases. However, there is considerable between-person variation in the rate of ageing and individual differences in their susceptibility to disease and death. Epigenetic mechanisms may play a role in human ageing, and DNA methylation age biomarkers may be good predictors of age-related diseases and mortality risk. The aims of this systematic review were to identify and synthesise the evidence for an association between peripherally measured DNA methylation age and longevity, age-related disease, and mortality risk. METHODS A systematic search was conducted in line with the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines. Using relevant search terms, MEDLINE, Embase, Cochrane Central Register of Controlled Trials, and PsychINFO databases were searched to identify articles meeting the inclusion criteria. Studies were assessed for bias using Joanna Briggs Institute critical appraisal checklists. Data was extracted from studies measuring age acceleration as a predictor of age-related diseases, mortality or longevity, and the findings for similar outcomes compared. Using Review Manager 5.3 software, two meta-analyses (one per epigenetic clock) were conducted on studies measuring all-cause mortality. RESULTS Twenty-three relevant articles were identified, including a total of 41,607 participants. Four studies focused on ageing and longevity, 11 on age-related disease (cancer, cardiovascular disease, and dementia), and 11 on mortality. There was some, although inconsistent, evidence for an association between increased DNA methylation age and risk of disease. Meta-analyses indicated that each 5-year increase in DNA methylation age was associated an 8 to 15% increased risk of mortality. CONCLUSION Due to the small number of studies and heterogeneity in study design and outcomes, the association between DNA methylation age and age-related disease and longevity is inconclusive. Increased epigenetic age was associated with mortality risk, but positive publication bias needs to be considered. Further research is needed to determine the extent to which DNA methylation age can be used as a clinical biomarker.
Collapse
Affiliation(s)
- Peter D Fransquet
- Department of Epidemiology and Preventive Medicine, Monash University, ASPREE, Level 5, The Alfred Centre, 99 Commercial Road, Melbourne, Victoria, 3004, Australia.,Disease Epigenetics, Murdoch Childrens Research Institute, The University of Melbourne, Parkville, Victoria, 3052, Australia
| | - Jo Wrigglesworth
- Department of Epidemiology and Preventive Medicine, Monash University, ASPREE, Level 5, The Alfred Centre, 99 Commercial Road, Melbourne, Victoria, 3004, Australia
| | - Robyn L Woods
- Department of Epidemiology and Preventive Medicine, Monash University, ASPREE, Level 5, The Alfred Centre, 99 Commercial Road, Melbourne, Victoria, 3004, Australia
| | - Michael E Ernst
- Department of Pharmacy Practice and Science, College of Pharmacy, The University of Iowa, Iowa City, IA, USA.,Department of Family Medicine, Carver College of Medicine, The University of Iowa, Iowa City, IA, USA
| | - Joanne Ryan
- Department of Epidemiology and Preventive Medicine, Monash University, ASPREE, Level 5, The Alfred Centre, 99 Commercial Road, Melbourne, Victoria, 3004, Australia. .,Disease Epigenetics, Murdoch Childrens Research Institute, The University of Melbourne, Parkville, Victoria, 3052, Australia. .,INSERM, U1061, Neuropsychiatrie, Recherche Clinique et Epidémiologique, Neuropsychiatry: Research Epidemiological and Clinic, Université Montpellier, 34000, Montpellier, France.
| |
Collapse
|
16
|
|
17
|
Leins H, Mulaw M, Eiwen K, Sakk V, Liang Y, Denkinger M, Geiger H, Schirmbeck R. Aged murine hematopoietic stem cells drive aging-associated immune remodeling. Blood 2018; 132:565-576. [PMID: 29891535 PMCID: PMC6137572 DOI: 10.1182/blood-2018-02-831065] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 05/30/2018] [Indexed: 02/07/2023] Open
Abstract
Aging-associated remodeling of the immune system impairs its functional integrity and contributes to increased morbidity and mortality in the elderly. Aging of hematopoietic stem cells (HSCs), from which all cells of the adaptive immune system ultimately originate, might play a crucial role in the remodeling of the aged immune system. We recently reported that aging of HSCs is, in part, driven by elevated activity of the small RhoGTPase Cdc42 and that aged HSCs can be rejuvenated in vitro by inhibition of the elevated Cdc42 activity in aged HSCs with the pharmacological compound CASIN. To study the quality of immune systems stemming selectively from young or aged HSCs, we established a HSC transplantation model in T- and B-cell-deficient young RAG1-/- hosts. We report that both phenotypic and functional changes in the immune system on aging are primarily a consequence of changes in the function of HSCs on aging and, to a large extent, independent of the thymus, as young and aged HSCs reconstituted distinct T- and B-cell subsets in RAG1-/- hosts that mirrored young and aged immune systems. Importantly, aged HSCs treated with CASIN reestablished an immune system similar to that of young animals, and thus capable of mounting a strong immune response to vaccination. Our studies further imply that epigenetic signatures already imprinted in aged HSCs determine the transcriptional profile and function of HSC-derived T and B cells.
Collapse
Affiliation(s)
- Hanna Leins
- Department of Internal Medicine I, University Hospital of Ulm, Ulm, Germany
- Institute of Molecular Medicine, Stem Cell and Aging, Ulm University, Ulm, Germany
| | - Medhanie Mulaw
- Institute of Experimental Cancer Research, University Hospital of Ulm, Ulm, Germany
| | - Karina Eiwen
- Institute of Molecular Medicine, Stem Cell and Aging, Ulm University, Ulm, Germany
| | - Vadim Sakk
- Institute of Molecular Medicine, Stem Cell and Aging, Ulm University, Ulm, Germany
| | - Ying Liang
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY
| | - Michael Denkinger
- AGAPLESION Bethesda Hospital, Geriatric Research Unit and
- Geriatric Center Ulm/Alb-Donau, Ulm University, Ulm, Germany; and
| | - Hartmut Geiger
- Institute of Molecular Medicine, Stem Cell and Aging, Ulm University, Ulm, Germany
- Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | | |
Collapse
|
18
|
Pawelec G. Does patient age influence anti-cancer immunity? Semin Immunopathol 2018; 41:125-131. [DOI: 10.1007/s00281-018-0697-6] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 07/06/2018] [Indexed: 12/30/2022]
|
19
|
Long-term moderate exercise enhances specific proteins that constitute neurotrophin signaling pathway: A TMT-based quantitative proteomic analysis of rat plasma. J Proteomics 2018; 185:39-50. [PMID: 29953961 DOI: 10.1016/j.jprot.2018.06.017] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 06/15/2018] [Accepted: 06/17/2018] [Indexed: 12/15/2022]
Abstract
Physical exercise has been reported to increase neurotrophin in brain tissues as hippocampus as well as increased neurotrophic level peripherally in blood plasma and might have an effect on/or affect molecular processes of energy metabolism (and homeostasis). In this study, using quantitative proteomic analysis, we obtained a plasma protein profile from the rat with long-term moderate exercise. A total of 752 proteins were identified in the plasma. Among them, 54 proteins were significant up-regulated and 47 proteins were down-regulated in the plasma of exercise group compared with the control group. Bioinformatic analyses showed that these altered proteins are widely involved in multiple biological processes, molecular functions and cellular components, which connect with 11 signaling pathways. Interestingly, 5 up-regulated proteins Rap1b, PTPN11, ARHGDIA, Cdc42 and YWHAE, confirmed by Western blots, are involved in the neurotrophin signaling pathway which shows the lowest P value among the identified pathways. Further analyses showed that the 5 neurotrophin-signaling-pathway-related proteins participate in two important protein-protein interaction networks associated to cell survival and apoptosis, axonal development, synapse formation and plasticity. This study provides an exercise-induced plasma protein profile, suggesting that long-term exercise enhances the proteins involved in neurotrophin signaling pathway which may contribute to health benefit. SIGNIFICANCE Physical activity contributes to myriad benefits on body health across the lifespan. The changes in plasma proteins after chronic moderate exercise may be used as biomarkers for health and may also play important roles in increase of cardiovascular fitness, enhancement of immune competence, prevention of obesity, decrease of risk for neurological disorders, cancer, stroke, diabetes and other metabolic disorders. Using a TMT-based proteomic method, this study identified 101 altered proteins in the plasma of rats after long-term moderate treadmill running, which may provide novel biomarkers for further investigation of the underlying mechanism of physical exercise. We confirmed that exercise enhances 5 proteins of the neurotrophin signaling pathway that may contribute to health benefits.
Collapse
|
20
|
Chaker D, Mouawad C, Azar A, Quilliot D, Achkar I, Fajloun Z, Makdissy N. Inhibition of the RhoGTPase Cdc42 by ML141 enhances hepatocyte differentiation from human adipose-derived mesenchymal stem cells via the Wnt5a/PI3K/miR-122 pathway: impact of the age of the donor. Stem Cell Res Ther 2018; 9:167. [PMID: 29921325 PMCID: PMC6009972 DOI: 10.1186/s13287-018-0910-5] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 05/08/2018] [Accepted: 05/20/2018] [Indexed: 12/11/2022] Open
Abstract
Background Human adipose-derived mesenchymal stem cells (hADSCs) are promising cells that may promote hepatocyte differentiation (Hep-Dif) and improve liver function, but the involvement of Cdc42, a key small RhoGTPase which plays a crucial role in aging, is still not well established. We hypothesized that the inhibition of Cdc42 may rescue the hepatogenic potential of hADSCs derived from aged donors. Methods hADSCs isolated from 61 women of different ages were cultured for evaluation of the proliferation of cells, adherence, apoptosis, immunomodulation, immunophenotyping, multipotency, gene expression, and cell function during Hep-Dif. Inhibition of Cdc42 by ML141 was realized during two phases: initiation (days –2 to 14 (D–2/14)) from undifferentiated to hepatoblast-like cells, or maturation (days 14 to 28 (D14/28)) from undifferentiated to hepatocyte-like cells. Mechanistic insights of the Wnt(s)/MAPK/PI3K/miR-122 pathways were studied. Results Cdc42 activity in undifferentiated hADSCs showed an age-dependent significant increase in Cdc42-GTP correlated to a decrease in Cdc42GAP; the low potentials of cell proliferation, doubling, adherence, and immunomodulatory ability (proinflammatory over anti-inflammatory) contrary to the apoptotic index of the aged group were significantly reversed by ML141. Aged donor cells showed a decreased potential for Hep-Dif which was rescued by ML141 treatment, giving rise to mature and functional hepatocyte-like cells as assessed by hepatic gene expression, cytochrome activity, urea and albumin production, low-density lipoprotein (LDL) uptake, and glycogen storage. ML141-induced Hep-Dif showed an improvement in mesenchymal-epithelial transition, a switch from Wtn-3a/β-catenin to Wnt5a signaling, involvement of PI3K/PKB but not the MAPK (ERK/JNK/p38) pathway, induction of miR-122 expression, reinforcing the exosomes release and the production of albumin, and epigenetic changes. Inhibition of PI3K and miR-122 abolished completely the effects of ML141 indicating that inhibition of Cdc42 promotes the Hep-Dif through a Wnt5a/PI3K/miR-122/HNF4α/albumin/E-cadherin-positive action. The ML141(D–2/14) protocol had more pronounced effects when compared with ML141(D14/28); inhibition of DNA methylation in combination with ML141(D–2/14) showed more efficacy in rescuing the Hep-Dif of aged hADSCs. In addition to Hep-Dif, the multipotency of aged hADSC-treated ML141 was observed by rescuing the adipocyte and neural differentiation by inducing PPARγ/FABP4 and NeuN/O4 but inhibiting Pref-1 and GFAP, respectively. Conclusion ML141 has the potential to reverse the age-related aberrations in aged stem cells and promotes their hepatogenic differentiation. Selective inhibition of Cdc42 could be a potential target of drug therapy for aging and may give new insights on the improvement of Hep-Dif. Electronic supplementary material The online version of this article (10.1186/s13287-018-0910-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Diana Chaker
- Lebanese University, Doctoral School for Sciences and Technology, Laboratory of Applied Biotechnology, Azm Center for Research in Biotechnology and its Applications, Tripoli, Lebanon.,Reviva Regenerative Medicine Center, Human Genetic Center, Middle East Institute of Health Hospital, Bsalim, Lebanon.,Paris Saclay University, Doctoral School, Therapeutical Innovation, Inserm UMR935, Villejuif, France
| | | | - Albert Azar
- Reviva Regenerative Medicine Center, Human Genetic Center, Middle East Institute of Health Hospital, Bsalim, Lebanon
| | - Didier Quilliot
- Diabetologia-Endocrinology & Nutrition, CHRU Nancy, INSERM 954, University Henri Poincaré de Lorraine, Faculty of Medicine, Nancy, France
| | | | - Ziad Fajloun
- Lebanese University, Doctoral School for Sciences and Technology, Laboratory of Applied Biotechnology, Azm Center for Research in Biotechnology and its Applications, Tripoli, Lebanon.,Lebanese University, Faculty of Sciences III, Department of Biology, Kobbe, Lebanon
| | - Nehman Makdissy
- Lebanese University, Doctoral School for Sciences and Technology, Laboratory of Applied Biotechnology, Azm Center for Research in Biotechnology and its Applications, Tripoli, Lebanon. .,Lebanese University, Faculty of Sciences III, Department of Biology, Kobbe, Lebanon.
| |
Collapse
|
21
|
Inflammation and neutrophil immunosenescence in health and disease: Targeted treatments to improve clinical outcomes in the elderly. Exp Gerontol 2018; 105:70-77. [DOI: 10.1016/j.exger.2017.12.020] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Revised: 12/21/2017] [Accepted: 12/22/2017] [Indexed: 12/20/2022]
|
22
|
Sapey E, Patel JM, Greenwood HL, Walton GM, Hazeldine J, Sadhra C, Parekh D, Dancer RCA, Nightingale P, Lord JM, Thickett DR. Pulmonary Infections in the Elderly Lead to Impaired Neutrophil Targeting, Which Is Improved by Simvastatin. Am J Respir Crit Care Med 2017; 196:1325-1336. [PMID: 28657793 PMCID: PMC5694832 DOI: 10.1164/rccm.201704-0814oc] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Accepted: 06/27/2017] [Indexed: 01/08/2023] Open
Abstract
RATIONALE Dysregulated neutrophil functions with age and sepsis are described. Statins are associated with improved infection survival in some observational studies, but trials in critically ill patients have not shown benefit. Statins also alter neutrophil responses in vitro. OBJECTIVES To assess neutrophil migratory accuracy with age during respiratory infections and determine if and how a statin intervention could alter these blunted responses. METHODS The migratory accuracy of blood neutrophils from young (aged <35 yr) and old (aged >60 yr) patients in health and during a lower respiratory tract infection, community-acquired pneumonia, and pneumonia associated with sepsis was assessed with and without simvastatin. In vitro results were confirmed in a double-blind randomized clinical trial in healthy elders. Cell adhesion markers were assessed. MEASUREMENTS AND MAIN RESULTS In vitro neutrophil migratory accuracy in the elderly deteriorated as the severity of the infectious pulmonary insult increased, without recovery at 6 weeks. Simvastatin rescued neutrophil migration with age and during mild to moderate infection, at high dose in older adults, but not during more severe sepsis. Confirming in vitro results, high-dose (80-mg) simvastatin improved neutrophil migratory accuracy without impeding other neutrophil functions in a double-blind randomized clinical trial in healthy elders. Simvastatin modified surface adhesion molecule expression and activity, facilitating accurate migration in the elderly. CONCLUSIONS Infections in older adults are associated with prolonged, impaired neutrophil migration, potentially contributing to poor outcomes. Statins improve neutrophil migration in vivo in health and in vitro in milder infective events, but not in severe sepsis, supporting their potential utility as an early intervention during pulmonary infections. Clinical trial registered with www.clinicaltrialsregister.eu (2011-002082-38).
Collapse
Affiliation(s)
- Elizabeth Sapey
- Institute of Inflammation and Ageing, University of Birmingham, Edgbaston, United Kingdom; and
| | - Jaimin M. Patel
- Institute of Inflammation and Ageing, University of Birmingham, Edgbaston, United Kingdom; and
| | - Hannah L. Greenwood
- Institute of Inflammation and Ageing, University of Birmingham, Edgbaston, United Kingdom; and
| | - Georgia M. Walton
- Institute of Inflammation and Ageing, University of Birmingham, Edgbaston, United Kingdom; and
| | - Jon Hazeldine
- Institute of Inflammation and Ageing, University of Birmingham, Edgbaston, United Kingdom; and
| | - Charendeep Sadhra
- Institute of Inflammation and Ageing, University of Birmingham, Edgbaston, United Kingdom; and
| | - Dhruv Parekh
- Institute of Inflammation and Ageing, University of Birmingham, Edgbaston, United Kingdom; and
| | - Rachel C. A. Dancer
- Institute of Inflammation and Ageing, University of Birmingham, Edgbaston, United Kingdom; and
| | - Peter Nightingale
- Medical Statistics, University Hospital Birmingham NHS Foundation Trust, Birmingham, United Kingdom
| | - Janet M. Lord
- Institute of Inflammation and Ageing, University of Birmingham, Edgbaston, United Kingdom; and
| | - David R. Thickett
- Institute of Inflammation and Ageing, University of Birmingham, Edgbaston, United Kingdom; and
| |
Collapse
|