151
|
Segel M, Neumann B, Hill MFE, Weber IP, Viscomi C, Zhao C, Young A, Agley CC, Thompson AJ, Gonzalez GA, Sharma A, Holmqvist S, Rowitch DH, Franze K, Franklin RJM, Chalut KJ. Niche stiffness underlies the ageing of central nervous system progenitor cells. Nature 2019; 573:130-134. [PMID: 31413369 PMCID: PMC7025879 DOI: 10.1038/s41586-019-1484-9] [Citation(s) in RCA: 315] [Impact Index Per Article: 52.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Accepted: 07/15/2019] [Indexed: 11/09/2022]
Abstract
Ageing causes a decline in tissue regeneration owing to a loss of function of adult stem cell and progenitor cell populations1. One example is the deterioration of the regenerative capacity of the widespread and abundant population of central nervous system (CNS) multipotent stem cells known as oligodendrocyte progenitor cells (OPCs)2. A relatively overlooked potential source of this loss of function is the stem cell 'niche'-a set of cell-extrinsic cues that include chemical and mechanical signals3,4. Here we show that the OPC microenvironment stiffens with age, and that this mechanical change is sufficient to cause age-related loss of function of OPCs. Using biological and synthetic scaffolds to mimic the stiffness of young brains, we find that isolated aged OPCs cultured on these scaffolds are molecularly and functionally rejuvenated. When we disrupt mechanical signalling, the proliferation and differentiation rates of OPCs are increased. We identify the mechanoresponsive ion channel PIEZO1 as a key mediator of OPC mechanical signalling. Inhibiting PIEZO1 overrides mechanical signals in vivo and allows OPCs to maintain activity in the ageing CNS. We also show that PIEZO1 is important in regulating cell number during CNS development. Thus we show that tissue stiffness is a crucial regulator of ageing in OPCs, and provide insights into how the function of adult stem and progenitor cells changes with age. Our findings could be important not only for the development of regenerative therapies, but also for understanding the ageing process itself.
Collapse
Affiliation(s)
- Michael Segel
- Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Björn Neumann
- Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Myfanwy F E Hill
- Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Isabell P Weber
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Carlo Viscomi
- MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge, UK
| | - Chao Zhao
- Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Adam Young
- Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Chibeza C Agley
- Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - Amelia J Thompson
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Ginez A Gonzalez
- Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Amar Sharma
- Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Staffan Holmqvist
- Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
- Department of Paediatrics, University of Cambridge, Cambridge, UK
| | - David H Rowitch
- Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
- Department of Paediatrics, University of Cambridge, Cambridge, UK
| | - Kristian Franze
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Robin J M Franklin
- Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK.
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK.
| | - Kevin J Chalut
- Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK.
- Cavendish Laboratory, Department of Physics, University of Cambridge, Cambridge, UK.
| |
Collapse
|
152
|
Ma L, Hu J, Cao Y, Xie Y, Wang H, Fan Z, Zhang C, Wang J, Wu CT, Wang S. Maintained Properties of Aged Dental Pulp Stem Cells for Superior Periodontal Tissue Regeneration. Aging Dis 2019; 10:793-806. [PMID: 31440385 PMCID: PMC6675537 DOI: 10.14336/ad.2018.0729] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Accepted: 07/29/2018] [Indexed: 12/23/2022] Open
Abstract
Owing to excellent therapeutic potential, mesenchymal stem cells (MSCs) are gaining increasing popularity with researchers worldwide for applications in tissue engineering, and in treatment of inflammation-related and age-related disorders. However, the senescence of MSCs over passaging has limited their clinical application owing to adverse effect on physiological function maintenance of tissues as well as disease treatment. An inflammatory microenvironment is one of the key contributors to MSC senescence, resulting in low regeneration efficiency. Therefore, MSCs with high resistance to cellular senescence would be a benefit for tissue regeneration. Toward this end, we analyzed the senescence properties of different types of stem cells during culture and under inflammation, including dental pulp stem cells (DPSCs), periodontal ligament stem cells (PDLSCs), bone marrow mesenchymal stem cells (BMMSCs), and adipose-derived stem cells (ADSCs). Overall, the DPSCs had higher proliferation rates, lower cellular senescence, and enhanced osteogenesis maintenance compared to those of non-dental MSCs cultured from passage three to six. The expression profiles of genes related to apoptosis, cell cycle, and cellular protein metabolic process (contributing to the cell self-renewal ability and metabolic processes) significantly differed between DPSCs and BMMSCs at passage three. Moreover, DPSCs were superior to BMMSCs with regards to resistance to lipopolysaccharide-induced apoptosis and senescence, with enhanced osteogenesis in vitro, and showed improved periodontal regeneration after injection in a miniature pig periodontitis model in vivo. Overall, the present study indicates that DPSCs show superior resistance to subculture and inflammation-induced senescence and would be suitable stem cells for tissue engineering with inflammation.
Collapse
Affiliation(s)
- Linsha Ma
- 1Molecular Laboratory for Gene Therapy & Tooth Regeneration, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University, School of Stomatology, Beijing, China
| | - Jingchao Hu
- 1Molecular Laboratory for Gene Therapy & Tooth Regeneration, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University, School of Stomatology, Beijing, China
| | - Yu Cao
- 1Molecular Laboratory for Gene Therapy & Tooth Regeneration, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University, School of Stomatology, Beijing, China
| | - Yilin Xie
- 1Molecular Laboratory for Gene Therapy & Tooth Regeneration, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University, School of Stomatology, Beijing, China
| | - Hua Wang
- 2Department of Experimental Hematology, Beijing Institute of Radiation Medicine, Beijing, China
| | - Zhipeng Fan
- 1Molecular Laboratory for Gene Therapy & Tooth Regeneration, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University, School of Stomatology, Beijing, China
| | - Chunmei Zhang
- 1Molecular Laboratory for Gene Therapy & Tooth Regeneration, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University, School of Stomatology, Beijing, China
| | - Jinsong Wang
- 1Molecular Laboratory for Gene Therapy & Tooth Regeneration, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University, School of Stomatology, Beijing, China.,3Department of Biochemistry and Molecular Biology, Capital Medical University School of Basic Medical Sciences, Beijing, China
| | - Chu-Tse Wu
- 2Department of Experimental Hematology, Beijing Institute of Radiation Medicine, Beijing, China
| | - Songlin Wang
- 1Molecular Laboratory for Gene Therapy & Tooth Regeneration, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University, School of Stomatology, Beijing, China.,3Department of Biochemistry and Molecular Biology, Capital Medical University School of Basic Medical Sciences, Beijing, China
| |
Collapse
|
153
|
Wang MJ, Chen J, Chen F, Liu Q, Sun Y, Yan C, Yang T, Bao Y, Hu YP. Rejuvenating Strategies of Tissue-specific Stem Cells for Healthy Aging. Aging Dis 2019; 10:871-882. [PMID: 31440391 PMCID: PMC6675530 DOI: 10.14336/ad.2018.1119] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Accepted: 11/19/2018] [Indexed: 12/22/2022] Open
Abstract
Although aging is a physiological process, it has raised interest in the science of aging and rejuvenation because of the increasing burden on the rapidly aging global population. With advanced age, there is a decline in homeostatic maintenance and regenerative responsiveness to the injury of various tissues, thereby contributing to the incidence of age-related diseases. The primary cause of the functional declines that occur along with aging is considered to be the exhaustion of stem cell functions in their corresponding tissues. Age-related changes in the systemic environment, the niche, and stem cells contribute to this loss. Thus, the reversal of stem cell aging at the cellular level might lead to the rejuvenation of the animal at an organismic level and the prevention of aging, which would be critical for developing new therapies for age-related dysfunction and diseases. Here, we will explore the effects of aging on stem cells in different tissues. The focus of this discussion is on pro-youth interventions that target intrinsic stem cell properties, environmental niche component, systemic factors, and senescent cellular clearance, which are promising for developing strategies related to the reversal of aged stem cell function and optimizing tissue repair processes.
Collapse
Affiliation(s)
- Min-Jun Wang
- 1Department of Cell Biology, Center for Stem Cell and Medicine, Second Military Medical University, Shanghai 200433, China
| | - Jiajia Chen
- 1Department of Cell Biology, Center for Stem Cell and Medicine, Second Military Medical University, Shanghai 200433, China
| | - Fei Chen
- 1Department of Cell Biology, Center for Stem Cell and Medicine, Second Military Medical University, Shanghai 200433, China
| | - Qinggui Liu
- 1Department of Cell Biology, Center for Stem Cell and Medicine, Second Military Medical University, Shanghai 200433, China
| | - Yu Sun
- 1Department of Cell Biology, Center for Stem Cell and Medicine, Second Military Medical University, Shanghai 200433, China
| | - Chen Yan
- 1Department of Cell Biology, Center for Stem Cell and Medicine, Second Military Medical University, Shanghai 200433, China
| | - Tao Yang
- 1Department of Cell Biology, Center for Stem Cell and Medicine, Second Military Medical University, Shanghai 200433, China
| | - Yiwen Bao
- 1Department of Cell Biology, Center for Stem Cell and Medicine, Second Military Medical University, Shanghai 200433, China.,2Department of Diagnostic radiology, University of Hong Kong, Hong Kong 999077, China
| | - Yi-Ping Hu
- 1Department of Cell Biology, Center for Stem Cell and Medicine, Second Military Medical University, Shanghai 200433, China
| |
Collapse
|
154
|
Pan X, Ruan C, Liu X, Kong L, Ma Y, Wu Q, Li H, Sun Y, Chen A, Zhao Q, Wu F, Wang X, Wang J, Zhu D, Gao P. Perivascular adipose tissue-derived stromal cells contribute to vascular remodeling during aging. Aging Cell 2019; 18:e12969. [PMID: 31087498 PMCID: PMC6612678 DOI: 10.1111/acel.12969] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 03/25/2019] [Accepted: 04/30/2019] [Indexed: 12/12/2022] Open
Abstract
Aging is an independent risk factor for vascular diseases. Perivascular adipose tissue (PVAT), an active component of the vasculature, contributes to vascular dysfunction during aging. Identification of underlying cell types and their changes during aging may provide meaningful insights regarding the clinical relevance of aging‐related vascular diseases. Here, we take advantage of single‐cell RNA sequence to characterize the resident stromal cells in the PVAT (PVASCs) and identified different clusters between young and aged PVASCs. Bioinformatics analysis revealed decreased endothelial and brown adipogenic differentiation capacities of PVASCs during aging, which contributed to neointimal hyperplasia after perivascular delivery to ligated carotid arteries. Mechanistically, in vitro and in vivo studies both suggested that aging‐induced loss of peroxisome proliferator‐activated receptor‐γ coactivator‐1 α (PGC1α) was a key regulator of decreased brown adipogenic differentiation in senescent PVASCs. We further demonstrated the existence of human PVASCs (hPVASCs) and overexpression of PGC1α improved hPVASC delivery‐induced vascular remodeling. Our finding emphasizes that differentiation capacities of PVASCs alter during aging and loss of PGC1α in aged PVASCs contributes to vascular remodeling via decreased brown adipogenic differentiation.
Collapse
Affiliation(s)
- Xiao‐Xi Pan
- Department of Hypertension, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension Shanghai Institute of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine Shanghai China
| | - Cheng‐Chao Ruan
- Department of Hypertension, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension Shanghai Institute of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine Shanghai China
| | - Xiu‐Ying Liu
- Key Laboratory of Genetic Network Biology, Institute of Genetics and Developmental Biology Chinese Academy of Sciences Beijing China
| | - Ling‐Ran Kong
- Department of Hypertension, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension Shanghai Institute of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine Shanghai China
| | - Yu Ma
- Department of Hypertension, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension Shanghai Institute of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine Shanghai China
| | - Qi‐Hong Wu
- Department of Hypertension, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension Shanghai Institute of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine Shanghai China
| | - Hai‐Qing Li
- Department of Cardiac Surgery Ruijin Hospital, Shanghai Jiao Tong University School of Medicine Shanghai China
| | - Yan‐Jun Sun
- Department of Cardiac Surgery Ruijin Hospital, Shanghai Jiao Tong University School of Medicine Shanghai China
| | - An‐Qing Chen
- Department of Cardiac Surgery Ruijin Hospital, Shanghai Jiao Tong University School of Medicine Shanghai China
| | - Qiang Zhao
- Department of Cardiac Surgery Ruijin Hospital, Shanghai Jiao Tong University School of Medicine Shanghai China
| | - Fang Wu
- Department of Geriatrics Ruijin Hospital, Shanghai Jiao Tong University School of Medicine Shanghai China
| | - Xiu‐Jie Wang
- Key Laboratory of Genetic Network Biology, Institute of Genetics and Developmental Biology Chinese Academy of Sciences Beijing China
| | - Ji‐Guang Wang
- Department of Hypertension, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension Shanghai Institute of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine Shanghai China
| | - Ding‐Liang Zhu
- Department of Hypertension, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension Shanghai Institute of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine Shanghai China
| | - Ping‐Jin Gao
- Department of Hypertension, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension Shanghai Institute of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine Shanghai China
| |
Collapse
|
155
|
|
156
|
Jung JS, Volk C, Marga C, Navarrete Santos A, Jung M, Rujescu D, Navarrete Santos A. Adipose-Derived Stem/Stromal Cells Recapitulate Aging Biomarkers and Show Reduced Stem Cell Plasticity Affecting Their Adipogenic Differentiation Capacity. Cell Reprogram 2019; 21:187-199. [PMID: 31298565 DOI: 10.1089/cell.2019.0010] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Stromal mesenchymal stem cells (MSCs) have the capability to self-renew and can differentiate into multiple cell types of the mesoderm germ layer, but their properties are affected by molecular aging mechanisms. MSCs can be obtained from adipose tissue termed as adipose-derived stem/stromal cells (ASCs) representing a promising tool for studying age-related diseases in detail. ASCs from young (16 weeks) and old (>108 weeks) rabbits were successfully isolated and propagated. ASCs showed the typical morphology and stained positive for CD105, Vimentin, Collagenase 1A, and negative for CD14, CD90, and CD73, demonstrating their mesenchymal origin. ASCs expressed MSC markers, including MYC, KLF4, CHD1, REST, and KAT6A, whereas pluripotency-related genes, such as NANOG, OCT4, and SOX2, were not expressed. Aged ASCs showed altered protein and mRNA levels of APOE, ATG7, FGF2, PTEN, and SIRT1. Adipogenic differentiation of old visceral ASCs was significantly decreased compared with young visceral ASCs. We successfully established rabbit ASC cultures representing an in vitro model for the analysis of stem cell aging mechanisms. ASCs, obtained from old female rabbits, showed age- and source-specific alteration due to aging of the donor. Stem cell plasticity was altered with age as shown by reduced adipogenic differentiation capacity.
Collapse
Affiliation(s)
- Juliane-Susanne Jung
- 1Department of Anatomy and Cell Biology, Psychotherapy, Psychosomatic Medicine, Martin Luther University Medical Faculty, Halle, Germany
| | - Christin Volk
- 1Department of Anatomy and Cell Biology, Psychotherapy, Psychosomatic Medicine, Martin Luther University Medical Faculty, Halle, Germany
| | - Christina Marga
- 1Department of Anatomy and Cell Biology, Psychotherapy, Psychosomatic Medicine, Martin Luther University Medical Faculty, Halle, Germany
| | - Alexander Navarrete Santos
- 2Center for Medical Basic Research, Psychotherapy, Psychosomatic Medicine, Martin Luther University Medical Faculty, Halle, Germany
| | - Matthias Jung
- 3Department of Psychiatry, Psychotherapy, Psychosomatic Medicine, Martin Luther University Medical Faculty, Halle, Germany
| | - Dan Rujescu
- 3Department of Psychiatry, Psychotherapy, Psychosomatic Medicine, Martin Luther University Medical Faculty, Halle, Germany
| | - Anne Navarrete Santos
- 1Department of Anatomy and Cell Biology, Psychotherapy, Psychosomatic Medicine, Martin Luther University Medical Faculty, Halle, Germany
| |
Collapse
|
157
|
Fuster JJ, Walsh K. Somatic Mutations and Clonal Hematopoiesis: Unexpected Potential New Drivers of Age-Related Cardiovascular Disease. Circ Res 2019; 122:523-532. [PMID: 29420212 DOI: 10.1161/circresaha.117.312115] [Citation(s) in RCA: 119] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Increasing evidence shows that conventional cardiovascular risk factors are incompletely predictive of cardiovascular disease, particularly in elderly individuals, suggesting that there may still be unidentified causal risk factors. Although the accumulation of somatic DNA mutations is a hallmark of aging, its relevance in cardiovascular disease or other age-related conditions has been, with the exception of cancer, largely unexplored. Here, we review recent clinical and preclinical studies that have identified acquired mutations in hematopoietic stem cells and subsequent clonal hematopoiesis as a new cardiovascular risk factor and a potential major driver of atherosclerosis. Understanding the mechanisms underlying the connection between somatic mutation-driven clonal hematopoiesis and cardiovascular disease will be highly relevant in the context of personalized medicine, as it may provide key information for the design of diagnostic, preventive, or therapeutic strategies tailored to the effects of specific somatic mutations.
Collapse
Affiliation(s)
- José J Fuster
- From the Molecular Cardiology Unit, Whitaker Cardiovascular Institute, Boston University School of Medicine, MA.
| | - Kenneth Walsh
- From the Molecular Cardiology Unit, Whitaker Cardiovascular Institute, Boston University School of Medicine, MA.
| |
Collapse
|
158
|
Denoth-Lippuner A, Jessberger S. Mechanisms of cellular rejuvenation. FEBS Lett 2019; 593:3381-3392. [PMID: 31197818 DOI: 10.1002/1873-3468.13483] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Revised: 04/29/2019] [Accepted: 05/02/2019] [Indexed: 01/15/2023]
Abstract
Aging leads to changes on an organismal but also cellular level. However, the exact mechanisms of cellular aging in mammals remain poorly understood and the identity and functional role of aging factors, some of which have previously been defined in model organisms such as Saccharomyces cerevisiae, remain elusive. Remarkably, during cellular reprogramming most if not all aging hallmarks are erased, offering a novel entry point to study aging and rejuvenation on a cellular level. On the other hand, direct reprogramming of old cells into cells of a different fate preserves many aging signs. Therefore, investigating the process of reprogramming and comparing it to direct reprogramming may yield novel insights about the clearing of aging factors, which is the basis of rejuvenation. Here, we discuss how reprogramming might lead to rejuvenation of a cell, an organ, or even the whole organism.
Collapse
Affiliation(s)
- Annina Denoth-Lippuner
- Laboratory of Neural Plasticity, Faculties of Medicine and Science, Brain Research Institute, University of Zurich, Switzerland
| | - Sebastian Jessberger
- Laboratory of Neural Plasticity, Faculties of Medicine and Science, Brain Research Institute, University of Zurich, Switzerland
| |
Collapse
|
159
|
Rožman P. How Could We Slow or Reverse the Human Aging Process and Extend the Healthy Life Span with Heterochronous Autologous Hematopoietic Stem Cell Transplantation. Rejuvenation Res 2019; 23:159-170. [PMID: 31203790 DOI: 10.1089/rej.2018.2164] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The senescence of the immune system contributes considerably to the age-related diseases that are the main causes of death after the age of 65. In this study, we present an appealing option for the prevention of immune senescence and slowing or reversing the aging process, which can be achieved by heterochronous autologous hematopoietic stem cell transplantation (haHSCT), where healthy autologous bone marrow stem cells are collected from donors while young, cryopreserved and stored for a long period, and reinfused at a later time when indicated. After reinfusion and homing, these young HSCs could participate in normal hemato- and immunopoiesis and improve several immune functions by expanding the immune- as well as hematopoietic cell repertoire. Several animal studies have already confirmed the feasibility of this procedure, which extended the longevity of the treated animals. If translated to human medicine, haHSCT could prevent or mitigate age-related immune defects and extend the healthy life span. In this review, we describe the concept of haHSCT, recent studies that confirm its feasibility, and discuss the further research needed to translate this heterochronous methodology.
Collapse
Affiliation(s)
- Primož Rožman
- Immunohaematology Department, Blood Transfusion Centre of Slovenia, Ljubljana, Slovenia
| |
Collapse
|
160
|
Spencer CA, Leung TH. Research Techniques Made Simple: Parabiosis to Elucidate Humoral Factors in Skin Biology. J Invest Dermatol 2019; 139:1208-1213.e1. [PMID: 31126426 DOI: 10.1016/j.jid.2019.03.1134] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 02/19/2019] [Accepted: 03/07/2019] [Indexed: 10/26/2022]
Abstract
Circulating factors in the blood and lymph support critical functions of living tissues. Parabiosis refers to the condition in which two entire living animals are conjoined and share a single circulatory system. This surgically created animal model was inspired by naturally occurring pairs of conjoined twins. Parabiosis experiments testing whether humoral factors from one animal affect the other have been performed for more than 150 years and have led to advances in endocrinology, neurology, musculoskeletal biology, and dermatology. The development of high-throughput genomics and proteomics approaches permitted the identification of potential circulating factors and rekindled scientific interest in parabiosis studies. For example, this technique may be used to assess how circulating factors may affect skin homeostasis, skin differentiation, skin aging, wound healing, and, potentially, skin cancer.
Collapse
Affiliation(s)
- Casey A Spencer
- Department of Dermatology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | - Thomas H Leung
- Department of Dermatology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA; Corporal Michael Crescenz Veterans Affairs Medical Center, Philadelphia, Pennsylvania, USA; Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.
| |
Collapse
|
161
|
Wild-type and SAMP8 mice show age-dependent changes in distinct stem cell compartments of the interfollicular epidermis. PLoS One 2019; 14:e0215908. [PMID: 31091266 PMCID: PMC6519801 DOI: 10.1371/journal.pone.0215908] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Accepted: 04/10/2019] [Indexed: 11/19/2022] Open
Abstract
Delayed wound healing and reduced barrier function with an increased risk of cancer are characteristics of aged skin and one possible mechanism is misregulation or dysfunction of epidermal stem cells during aging. Recent studies have identified heterogeneous stem cell populations within the mouse interfollicular epidermis that are defined by territorial distribution and cell division frequency; however, it is unknown whether the individual stem cell populations undergo distinct aging processes. Here we provide comprehensive characterization of age-related changes in the mouse epidermis within the specific territories of slow-cycling and fast-dividing stem cells using old wild-type, senescence-accelerated mouse prone 1 (SAMP1) and SAMP8 mice. During aging, the epidermis exhibits structural changes such as irregular micro-undulations and overall thinning of the tissue. We also find that, in the old epidermis, proliferation is preferentially decreased in the region where fast-dividing stem cells reside whereas the lineage differentiation marker appears to be more affected in the slow-cycling stem cell region. Furthermore, SAMP8, but not SAMP1, exhibits precocious aging similar to that of aged wild-type mice, suggesting a potential use of this model for aging study of the epidermis and its stem cells. Taken together, our study reveals distinct aging processes governing the two epidermal stem cell populations and suggests a potential mechanism in differential responses of compartmentalized stem cells and their niches to aging.
Collapse
|
162
|
Shu P, Wu C, Ruan X, Liu W, Hou L, Fu H, Wang M, Liu C, Zeng Y, Chen P, Yin B, Yuan J, Qiang B, Peng X, Zhong W. Opposing Gradients of MicroRNA Expression Temporally Pattern Layer Formation in the Developing Neocortex. Dev Cell 2019; 49:764-785.e4. [PMID: 31080058 DOI: 10.1016/j.devcel.2019.04.017] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Revised: 11/08/2018] [Accepted: 04/07/2019] [Indexed: 12/24/2022]
Abstract
The precisely timed generation of different neuronal types is a hallmark of development from invertebrates to vertebrates. In the developing mammalian neocortex, neural stem cells change competence over time to sequentially produce six layers of functionally distinct neurons. Here, we report that microRNAs (miRNAs) are dispensable for stem-cell self-renewal and neuron production but essential for timing neocortical layer formation and specifying laminar fates. Specifically, as neurogenesis progresses, stem cells reduce miR-128 expression and miR-9 activity but steadily increase let-7 expression, whereas neurons initially maintain the differences in miRNA expression present at birth. Moreover, miR-128, miR-9, and let-7 are functionally distinct; capable of specifying neurons for layer VI and layer V and layers IV, III, and II, respectively; and transiently altering their relative levels of expression can modulate stem-cell competence in a neurogenic-stage-specific manner to shift neuron production between earlier-born and later-born fates, partly by temporally regulating a neurogenesis program involving Hmga2.
Collapse
Affiliation(s)
- Pengcheng Shu
- The State Key Laboratory of Medical Molecular Biology, Neuroscience Center, Medical Primates Research Center and Department of Molecular Biology and Biochemistry, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, China
| | - Chao Wu
- The State Key Laboratory of Medical Molecular Biology, Neuroscience Center, Medical Primates Research Center and Department of Molecular Biology and Biochemistry, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, China
| | - Xiangbin Ruan
- The State Key Laboratory of Medical Molecular Biology, Neuroscience Center, Medical Primates Research Center and Department of Molecular Biology and Biochemistry, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, China
| | - Wei Liu
- Department of Anatomy and Histology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, China
| | - Lin Hou
- The State Key Laboratory of Medical Molecular Biology, Neuroscience Center, Medical Primates Research Center and Department of Molecular Biology and Biochemistry, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, China
| | - Hongye Fu
- The State Key Laboratory of Medical Molecular Biology, Neuroscience Center, Medical Primates Research Center and Department of Molecular Biology and Biochemistry, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, China
| | - Ming Wang
- The State Key Laboratory of Medical Molecular Biology, Neuroscience Center, Medical Primates Research Center and Department of Molecular Biology and Biochemistry, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, China
| | - Chang Liu
- The State Key Laboratory of Medical Molecular Biology, Neuroscience Center, Medical Primates Research Center and Department of Molecular Biology and Biochemistry, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, China
| | - Yi Zeng
- The State Key Laboratory of Medical Molecular Biology, Neuroscience Center, Medical Primates Research Center and Department of Molecular Biology and Biochemistry, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, China
| | - Pan Chen
- The State Key Laboratory of Medical Molecular Biology, Neuroscience Center, Medical Primates Research Center and Department of Molecular Biology and Biochemistry, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, China
| | - Bin Yin
- The State Key Laboratory of Medical Molecular Biology, Neuroscience Center, Medical Primates Research Center and Department of Molecular Biology and Biochemistry, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, China
| | - Jiangang Yuan
- The State Key Laboratory of Medical Molecular Biology, Neuroscience Center, Medical Primates Research Center and Department of Molecular Biology and Biochemistry, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, China
| | - Boqin Qiang
- The State Key Laboratory of Medical Molecular Biology, Neuroscience Center, Medical Primates Research Center and Department of Molecular Biology and Biochemistry, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, China
| | - Xiaozhong Peng
- The State Key Laboratory of Medical Molecular Biology, Neuroscience Center, Medical Primates Research Center and Department of Molecular Biology and Biochemistry, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, China; Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650118, China.
| | - Weimin Zhong
- Department of Molecular, Cellular, and Developmental Biology, Yale University, P.O. Box 208103, New Haven, CT 06520, USA.
| |
Collapse
|
163
|
Mohammad K, Dakik P, Medkour Y, Mitrofanova D, Titorenko VI. Quiescence Entry, Maintenance, and Exit in Adult Stem Cells. Int J Mol Sci 2019; 20:ijms20092158. [PMID: 31052375 PMCID: PMC6539837 DOI: 10.3390/ijms20092158] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 04/24/2019] [Accepted: 04/28/2019] [Indexed: 12/13/2022] Open
Abstract
Cells of unicellular and multicellular eukaryotes can respond to certain environmental cues by arresting the cell cycle and entering a reversible state of quiescence. Quiescent cells do not divide, but can re-enter the cell cycle and resume proliferation if exposed to some signals from the environment. Quiescent cells in mammals and humans include adult stem cells. These cells exhibit improved stress resistance and enhanced survival ability. In response to certain extrinsic signals, adult stem cells can self-renew by dividing asymmetrically. Such asymmetric divisions not only allow the maintenance of a population of quiescent cells, but also yield daughter progenitor cells. A multistep process of the controlled proliferation of these progenitor cells leads to the formation of one or more types of fully differentiated cells. An age-related decline in the ability of adult stem cells to balance quiescence maintenance and regulated proliferation has been implicated in many aging-associated diseases. In this review, we describe many traits shared by different types of quiescent adult stem cells. We discuss how these traits contribute to the quiescence, self-renewal, and proliferation of adult stem cells. We examine the cell-intrinsic mechanisms that allow establishing and sustaining the characteristic traits of adult stem cells, thereby regulating quiescence entry, maintenance, and exit.
Collapse
Affiliation(s)
- Karamat Mohammad
- Department of Biology, Concordia University, 7141 Sherbrooke Street, West, SP Building, Room 501-13, Montreal, QC H4B 1R6, Canada.
| | - Paméla Dakik
- Department of Biology, Concordia University, 7141 Sherbrooke Street, West, SP Building, Room 501-13, Montreal, QC H4B 1R6, Canada.
| | - Younes Medkour
- Department of Biology, Concordia University, 7141 Sherbrooke Street, West, SP Building, Room 501-13, Montreal, QC H4B 1R6, Canada.
| | - Darya Mitrofanova
- Department of Biology, Concordia University, 7141 Sherbrooke Street, West, SP Building, Room 501-13, Montreal, QC H4B 1R6, Canada.
| | - Vladimir I Titorenko
- Department of Biology, Concordia University, 7141 Sherbrooke Street, West, SP Building, Room 501-13, Montreal, QC H4B 1R6, Canada.
| |
Collapse
|
164
|
Lin H, Sohn J, Shen H, Langhans MT, Tuan RS. Bone marrow mesenchymal stem cells: Aging and tissue engineering applications to enhance bone healing. Biomaterials 2019; 203:96-110. [PMID: 29980291 PMCID: PMC6733253 DOI: 10.1016/j.biomaterials.2018.06.026] [Citation(s) in RCA: 258] [Impact Index Per Article: 43.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Revised: 06/18/2018] [Accepted: 06/19/2018] [Indexed: 12/11/2022]
Abstract
Bone has well documented natural healing capacity that normally is sufficient to repair fractures and other common injuries. However, the properties of bone change throughout life, and aging is accompanied by increased incidence of bone diseases and compromised fracture healing capacity, which necessitate effective therapies capable of enhancing bone regeneration. The therapeutic potential of adult mesenchymal stem cells (MSCs) for bone repair has been long proposed and examined. Actions of MSCs may include direct differentiation to become bone cells, attraction and recruitment of other cells, or creation of a regenerative environment via production of trophic growth factors. With systemic aging, MSCs also undergo functional decline, which has been well investigated in a number of recent studies. In this review, we first describe the changes in MSCs during aging and discuss how these alterations can affect bone regeneration. We next review current research findings on bone tissue engineering, which is considered a promising and viable therapeutic solution for structural and functional restoration of bone. In particular, the importance of MSCs and bioscaffolds is highlighted. Finally, potential approaches for the prevention of MSC aging and the rejuvenation of aged MSC are discussed.
Collapse
Affiliation(s)
- Hang Lin
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; McGowan Institute for Regenerative Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Jihee Sohn
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - He Shen
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Division of Nanobiomedicine, Suzhou Institute of Nano-tech and Nano-bionics, Chinese Academy of Sciences, China
| | - Mark T Langhans
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Rocky S Tuan
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; McGowan Institute for Regenerative Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; The Chinese University of Hong Kong, Hong Kong SAR, China.
| |
Collapse
|
165
|
Chen D, Kerr C. The Epigenetics of Stem Cell Aging Comes of Age. Trends Cell Biol 2019; 29:563-568. [PMID: 31030975 DOI: 10.1016/j.tcb.2019.03.006] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 03/21/2019] [Accepted: 03/26/2019] [Indexed: 12/16/2022]
Abstract
Emerging evidence indicates that epigenetic regulators are critically required for the maintenance of tissue-specific stem cells and that the epigenetic marks are altered in stem cells during physiological aging. Intriguingly, aging-associated stem cell functional decline can be reversed by manipulating epigenetic factors that become dysregulated during aging. These observations lend support to the stem cell theory of aging, which postulates that aging is the result of the inability of tissue-specific stem cells to replenish the tissues with functional differentiated cells that maintain the function of a tissue, and open a new era of research on the epigenetics of stem cell aging that may represent therapeutic potential. Recent advances in single cell technologies are revolutionizing our mechanistic understanding of rare populations of cells, such as stem cells, and offer an unprecedented opportunity to address this challenge.
Collapse
Affiliation(s)
- Danica Chen
- Department of Nutritional Sciences and Toxicology, University of California at Berkeley, Berkeley, CA 94720, USA.
| | - Candace Kerr
- Division of Aging Biology, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
166
|
Liu N, Matsumura H, Kato T, Ichinose S, Takada A, Namiki T, Asakawa K, Morinaga H, Mohri Y, De Arcangelis A, Geroges-Labouesse E, Nanba D, Nishimura EK. Stem cell competition orchestrates skin homeostasis and ageing. Nature 2019; 568:344-350. [PMID: 30944469 DOI: 10.1038/s41586-019-1085-7] [Citation(s) in RCA: 269] [Impact Index Per Article: 44.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Accepted: 03/07/2019] [Indexed: 01/09/2023]
Abstract
Stem cells underlie tissue homeostasis, but their dynamics during ageing-and the relevance of these dynamics to organ ageing-remain unknown. Here we report that the expression of the hemidesmosome component collagen XVII (COL17A1) by epidermal stem cells fluctuates physiologically through genomic/oxidative stress-induced proteolysis, and that the resulting differential expression of COL17A1 in individual stem cells generates a driving force for cell competition. In vivo clonal analysis in mice and in vitro 3D modelling show that clones that express high levels of COL17A1, which divide symmetrically, outcompete and eliminate adjacent stressed clones that express low levels of COL17A1, which divide asymmetrically. Stem cells with higher potential or quality are thus selected for homeostasis, but their eventual loss of COL17A1 limits their competition, thereby causing ageing. The resultant hemidesmosome fragility and stem cell delamination deplete adjacent melanocytes and fibroblasts to promote skin ageing. Conversely, the forced maintenance of COL17A1 rescues skin organ ageing, thereby indicating potential angles for anti-ageing therapeutic intervention.
Collapse
Affiliation(s)
- Nan Liu
- Department of Stem Cell Biology, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan
| | - Hiroyuki Matsumura
- Department of Stem Cell Biology, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan.
| | - Tomoki Kato
- Department of Stem Cell Biology, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan
| | - Shizuko Ichinose
- Research Center for Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Aki Takada
- Department of Stem Cell Biology, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan
| | - Takeshi Namiki
- Department of Dermatology, Tokyo Medical and Dental University Graduate School and Faculty of Medicine, Tokyo, Japan
| | - Kyosuke Asakawa
- Department of Stem Cell Biology, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan
| | - Hironobu Morinaga
- Department of Stem Cell Biology, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan
| | - Yasuaki Mohri
- Department of Stem Cell Biology, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan
| | - Adèle De Arcangelis
- CNRS UMR7104, Inserm U1258, Institut de Génétique et de Biologie Moléculaire et Cellulaire, Development and Stem Cells Department, Université de Strasbourg, Strasbourg, France
| | - Elisabeth Geroges-Labouesse
- CNRS UMR7104, Inserm U1258, Institut de Génétique et de Biologie Moléculaire et Cellulaire, Development and Stem Cells Department, Université de Strasbourg, Strasbourg, France
| | - Daisuke Nanba
- Department of Stem Cell Biology, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan
| | - Emi K Nishimura
- Department of Stem Cell Biology, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan.
| |
Collapse
|
167
|
Fu L, Hu Y, Song M, Liu Z, Zhang W, Yu FX, Wu J, Wang S, Izpisua Belmonte JC, Chan P, Qu J, Tang F, Liu GH. Up-regulation of FOXD1 by YAP alleviates senescence and osteoarthritis. PLoS Biol 2019; 17:e3000201. [PMID: 30933975 PMCID: PMC6459557 DOI: 10.1371/journal.pbio.3000201] [Citation(s) in RCA: 119] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Revised: 04/11/2019] [Accepted: 03/13/2019] [Indexed: 12/12/2022] Open
Abstract
Cellular senescence is a driver of various aging-associated disorders, including osteoarthritis. Here, we identified a critical role for Yes-associated protein (YAP), a major effector of Hippo signaling, in maintaining a younger state of human mesenchymal stem cells (hMSCs) and ameliorating osteoarthritis in mice. Clustered regularly interspaced short palindromic repeat (CRISPR)/CRISPR associated protein 9 nuclease (Cas9)-mediated knockout (KO) of YAP in hMSCs resulted in premature cellular senescence. Mechanistically, YAP cooperated with TEA domain transcriptional factor (TEAD) to activate the expression of forkhead box D1 (FOXD1), a geroprotective protein. YAP deficiency led to the down-regulation of FOXD1. In turn, overexpression of YAP or FOXD1 rejuvenated aged hMSCs. Moreover, intra-articular administration of lentiviral vector encoding YAP or FOXD1 attenuated the development of osteoarthritis in mice. Collectively, our findings reveal YAP–FOXD1, a novel aging-associated regulatory axis, as a potential target for gene therapy to alleviate osteoarthritis. The Hippo signalling effector YAP and the transcription factor FOXD1 play a role in alleviating cellular senescence and osteoarthritis, identifying the YAP-FOXD1 axis as a potential therapeutic target for aging-associated disorders. Stem cell aging contributes to aging-associated degenerative diseases. Studies aiming to characterize the mechanisms of stem cell aging are critical for obtaining a comprehensive understanding of the aging process and developing novel strategies to treat aging-related diseases. As a prevalent aging-associated chronic joint disorder, osteoarthritis is a leading cause of disability. Senescent mesenchymal stem cells (MSCs) residing in the joint may be a critical target for the prevention of osteoarthritis; however, the key regulators of MSC senescence are little known, and targeting aging regulatory genes for the treatment of osteoarthritis has not yet been reported. Here, we show that Yes-associated protein (YAP), a major effector of Hippo signaling, represses human mesenchymal stem cell (hMSC) senescence through transcriptional up-regulation of forkhead box D1 (FOXD1). Lentiviral gene transfer of YAP or FOXD1 can rejuvenate aged hMSCs and ameliorate osteoarthritis symptoms in mouse models. We propose that the YAP–FOXD1 axis is a novel target for combating aging-associated diseases.
Collapse
Affiliation(s)
- Lina Fu
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yuqiong Hu
- Beijing Advanced Innovation Center for Genomics, College of Life Sciences, Peking University, Beijing, China
- Biomedical Pioneering Innovation Center, Peking University, Beijing, China
| | - Moshi Song
- University of Chinese Academy of Sciences, Beijing, China
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
| | - Zunpeng Liu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Weiqi Zhang
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- Advanced Innovation Center for Human Brain Protection, National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing, China
| | - Fa-Xing Yu
- Children's Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Jun Wu
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Si Wang
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- Advanced Innovation Center for Human Brain Protection, National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing, China
| | - Juan Carlos Izpisua Belmonte
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, California, United States of America
| | - Piu Chan
- Advanced Innovation Center for Human Brain Protection, National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing, China
| | - Jing Qu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- * E-mail: (JQ); (FT); (GHL)
| | - Fuchou Tang
- Beijing Advanced Innovation Center for Genomics, College of Life Sciences, Peking University, Beijing, China
- Biomedical Pioneering Innovation Center, Peking University, Beijing, China
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
- Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, Beijing, China
- * E-mail: (JQ); (FT); (GHL)
| | - Guang-Hui Liu
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- Advanced Innovation Center for Human Brain Protection, National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing, China
- Beijing Institute for Brain Disorders, Beijing, China
- * E-mail: (JQ); (FT); (GHL)
| |
Collapse
|
168
|
Singh PP, Demmitt BA, Nath RD, Brunet A. The Genetics of Aging: A Vertebrate Perspective. Cell 2019; 177:200-220. [PMID: 30901541 PMCID: PMC7592626 DOI: 10.1016/j.cell.2019.02.038] [Citation(s) in RCA: 165] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Revised: 02/21/2019] [Accepted: 02/22/2019] [Indexed: 02/07/2023]
Abstract
Aging negatively impacts vitality and health. Many genetic pathways that regulate aging were discovered in invertebrates. However, the genetics of aging is more complex in vertebrates because of their specialized systems. This Review discusses advances in the genetic regulation of aging in vertebrates from work in mice, humans, and organisms with exceptional lifespans. We highlight challenges for the future, including sex-dependent differences in lifespan and the interplay between genes and environment. We also discuss how the identification of reliable biomarkers of age and development of new vertebrate models can be leveraged for personalized interventions to counter aging and age-related diseases.
Collapse
Affiliation(s)
- Param Priya Singh
- Department of Genetics, Stanford University, Stanford, CA 94305, USA
| | | | - Ravi D Nath
- Department of Genetics, Stanford University, Stanford, CA 94305, USA
| | - Anne Brunet
- Department of Genetics, Stanford University, Stanford, CA 94305, USA; Glenn Laboratories for the Biology of Aging, Stanford, CA 94305, USA.
| |
Collapse
|
169
|
Argüelles S, Guerrero-Castilla A, Cano M, Muñoz MF, Ayala A. Advantages and disadvantages of apoptosis in the aging process. Ann N Y Acad Sci 2019; 1443:20-33. [PMID: 30839127 DOI: 10.1111/nyas.14020] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2018] [Revised: 12/14/2018] [Accepted: 01/16/2019] [Indexed: 12/12/2022]
Abstract
Researchers cannot predict as yet how long a human being can live. Life expectancy has been steadily increasing in the last century, but perhaps not always the quality of life in parallel with it. Future generations will be faced with the problems of an increased life expectancy along with the emergence of new age-related diseases. A deeper understanding of the aging process is crucial to ameliorate, if not to prevent, these projected new old-age diseases. One of the mechanisms responsible for healthy aging is through the effective maintenance of physiological, biochemical, and immunological functions. To carry this out, the organism needs to create new cells to replace old ones and to induce the disappearance of old and damaged cells. Apoptosis is involved in all these processes. However, if apoptosis is dysregulated, premature senescence-associated diseases are likely to appear. In our review, the focus will be on a better understanding of the role of apoptosis in the aging process. These signaling pathways will most assuredly be pharmacologically targeted in antiaging medicine therapies.
Collapse
Affiliation(s)
- Sandro Argüelles
- Department of Physiology, Faculty of Pharmacy, University of Seville, Seville, Spain
| | | | - Mercedes Cano
- Department of Physiology, Faculty of Pharmacy, University of Seville, Seville, Spain
| | - Mario F Muñoz
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, University of Seville, Seville, Spain
| | - Antonio Ayala
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, University of Seville, Seville, Spain
| |
Collapse
|
170
|
Liang X, Ding Y, Lin F, Zhang Y, Zhou X, Meng Q, Lu X, Jiang G, Zhu H, Chen Y, Lian Q, Fan H, Liu Z. Overexpression of ERBB4 rejuvenates aged mesenchymal stem cells and enhances angiogenesis via PI3K/AKT and MAPK/ERK pathways. FASEB J 2019; 33:4559-4570. [PMID: 30566395 DOI: 10.1096/fj.201801690r] [Citation(s) in RCA: 89] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The age-related functional exhaustion limits potential efficacy of mesenchymal stem cells (MSC) in treating cardiovascular disease. Therefore, rejuvenation of aged MSC in the elderly population is of great interest. We have previously reported that Erb-B2 receptor tyrosine kinase 4 ( ERBB4) plays a critical role in regulating MSC survival under hypoxia. The aim of this study was to investigate whether ERBB4 rejuvenates aged MSC and how ERBB4 enhances therapeutic efficacy of aged MSC in treating myocardial infarction (MI). Compared with vector aged MSC (aged-MSC), ERBB4-engineered aged MSC (ER4-aged-MSC) conferred resistance to oxidative stress-induced cell death and ameliorated the senescent phenotype in vitro. Four weeks after MI, the ER4-aged-MSC group exhibited enhanced blood vessel density, reduced cardiac remodeling and apoptosis with improved heart function compared with the aged-MSC group. Overexpression of ERBB4 caused an increase in phosphorylated v-akt murine thymoma viral oncogene homolog 1 (AKT), and phosphorylated ERK expression under hypoxia. ER4-aged-MSC secreted higher levels of angiopoietin, epithelial neutrophil activating peptide 78, VEGF, and fibroblast growth factor 2, and enhanced tube formation in HUVEC. The impact of ERBB4 on protein expression, proangiogenesis, cell behavior, and cytokine secretion was abolished by inhibiting PI3K/AKT and MAPK/ERK signaling pathway.-Liang, X., Ding, Y., Lin, F., Zhang, Y., Zhou, X., Meng, Q., Lu, X., Jiang, G., Zhu, H., Chen, Y., Lian, Q., Fan, H., Liu, Z. Overexpression of ERBB4 rejuvenates aged mesenchymal stem cells and enhances angiogenesis via PI3K/AKT and MAPK/ERK pathways.
Collapse
Affiliation(s)
- Xiaoting Liang
- Clinical Translational Medical Research Center, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
- Translational Medical Center for Stem Cell Therapy, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yue Ding
- Department of Organ Transplantation, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Fang Lin
- Clinical Translational Medical Research Center, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yuelin Zhang
- Department of Emergency, Guangdong General Hospital, Guangdong Academy of Medical Science, China
- Faculty of Pharmacy, Bengbu Medical College, China
- Department of Medicine, The University of Hong Kong, Hong Kong, China; and
| | - Xiaohui Zhou
- Clinical Translational Medical Research Center, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Qingshu Meng
- Clinical Translational Medical Research Center, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xingyue Lu
- Faculty of Pharmacy, Bengbu Medical College, China
| | - Guojun Jiang
- Faculty of Pharmacy, Bengbu Medical College, China
| | - Hongming Zhu
- Clinical Translational Medical Research Center, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
- Translational Medical Center for Stem Cell Therapy, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yu Chen
- Department of Organ Transplantation, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Qizhou Lian
- Department of Medicine, The University of Hong Kong, Hong Kong, China; and
| | - Huimin Fan
- Department of Cardiovascular and Thoracic Surgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Zhongmin Liu
- Translational Medical Center for Stem Cell Therapy, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
- Department of Cardiovascular and Thoracic Surgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
171
|
Abstract
Modern stem cell research has mainly focused on protein expression and transcriptional networks. However, transmembrane voltage gradients generated by ion channels and transporters have demonstrated to be powerful regulators of cellular processes. These physiological cues exert influence on cell behaviors ranging from differentiation and proliferation to migration and polarity. Bioelectric signaling is a fundamental element of living systems and an untapped reservoir for new discoveries. Dissecting these mechanisms will allow for novel methods of controlling cell fate and open up new opportunities in biomedicine. This review focuses on the role of ion channels and the resting membrane potential in the proliferation and differentiation of skeletal muscle progenitor cells. In addition, findings relevant to this topic are presented and potential implications for tissue engineering and regenerative medicine are discussed.
Collapse
Affiliation(s)
- Colin Fennelly
- Department of Neuroscience, Novartis Institutes for BioMedical Research, Inc., Cambridge, Massachusetts
| | - Shay Soker
- Wake Forest Institute for Regenerative Medicine, Winston-Salem, North Carolina
- Wake Forest School of Medicine, Winston-Salem, North Carolina
| |
Collapse
|
172
|
Zhang X, Liu Z, Liu X, Wang S, Zhang Y, He X, Sun S, Ma S, Shyh-Chang N, Liu F, Wang Q, Wang X, Liu L, Zhang W, Song M, Liu GH, Qu J. Telomere-dependent and telomere-independent roles of RAP1 in regulating human stem cell homeostasis. Protein Cell 2019; 10:649-667. [PMID: 30796637 PMCID: PMC6711945 DOI: 10.1007/s13238-019-0610-7] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2018] [Accepted: 01/03/2019] [Indexed: 01/19/2023] Open
Abstract
RAP1 is a well-known telomere-binding protein, but its functions in human stem cells have remained unclear. Here we generated RAP1-deficient human embryonic stem cells (hESCs) by using CRISPR/Cas9 technique and obtained RAP1-deficient human mesenchymal stem cells (hMSCs) and neural stem cells (hNSCs) via directed differentiation. In both hMSCs and hNSCs, RAP1 not only negatively regulated telomere length but also acted as a transcriptional regulator of RELN by tuning the methylation status of its gene promoter. RAP1 deficiency enhanced self-renewal and delayed senescence in hMSCs, but not in hNSCs, suggesting complicated lineage-specific effects of RAP1 in adult stem cells. Altogether, these results demonstrate for the first time that RAP1 plays both telomeric and nontelomeric roles in regulating human stem cell homeostasis.
Collapse
Affiliation(s)
- Xing Zhang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.,Advanced Innovation Center for Human Brain Protection, National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing, 100053, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Zunpeng Liu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xiaoqian Liu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Si Wang
- Advanced Innovation Center for Human Brain Protection, National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing, 100053, China.,National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China.,Institute for Stem cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China
| | - Yiyuan Zhang
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xiaojuan He
- Advanced Innovation Center for Human Brain Protection, National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing, 100053, China
| | - Shuhui Sun
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Shuai Ma
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Ng Shyh-Chang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.,University of Chinese Academy of Sciences, Beijing, 100049, China.,Institute for Stem cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China
| | - Feng Liu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.,University of Chinese Academy of Sciences, Beijing, 100049, China.,Institute for Stem cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China
| | - Qiang Wang
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.,University of Chinese Academy of Sciences, Beijing, 100049, China.,Institute for Stem cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China
| | - Xiaoqun Wang
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China.,University of Chinese Academy of Sciences, Beijing, 100049, China.,Institute for Stem cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China
| | - Lin Liu
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071, China
| | - Weiqi Zhang
- Advanced Innovation Center for Human Brain Protection, National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing, 100053, China. .,National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China. .,University of Chinese Academy of Sciences, Beijing, 100049, China. .,Institute for Stem cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.
| | - Moshi Song
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China. .,University of Chinese Academy of Sciences, Beijing, 100049, China. .,Institute for Stem cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.
| | - Guang-Hui Liu
- Advanced Innovation Center for Human Brain Protection, National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing, 100053, China. .,National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China. .,University of Chinese Academy of Sciences, Beijing, 100049, China. .,Institute for Stem cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China. .,Key Laboratory of Regenerative Medicine of Ministry of Education, Institute of Aging and Regenerative Medicine, Jinan University, Guangzhou, 510632, China. .,Beijing Institute for Brain Disorders, Capital Medical University, Beijing, 100069, China.
| | - Jing Qu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China. .,University of Chinese Academy of Sciences, Beijing, 100049, China. .,Institute for Stem cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.
| |
Collapse
|
173
|
The global clonal complexity of the murine blood system declines throughout life and after serial transplantation. Blood 2019; 133:1927-1942. [PMID: 30782612 DOI: 10.1182/blood-2018-09-873059] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2018] [Accepted: 02/18/2019] [Indexed: 01/02/2023] Open
Abstract
Although many recent studies describe the emergence and prevalence of "clonal hematopoiesis of indeterminate potential" in aged human populations, a systematic analysis of the numbers of clones supporting steady-state hematopoiesis throughout mammalian life is lacking. Previous efforts relied on transplantation of "barcoded" hematopoietic stem cells (HSCs) to track the contribution of HSC clones to reconstituted blood. However, ex vivo manipulation and transplantation alter HSC function and thus may not reflect the biology of steady-state hematopoiesis. Using a noninvasive in vivo color-labeling system, we report the first comprehensive analysis of the changing global clonal complexity of steady-state hematopoiesis during the natural murine lifespan. We observed that the number of clones (ie, clonal complexity) supporting the major blood and bone marrow hematopoietic compartments decline with age by ∼30% and ∼60%, respectively. Aging dramatically reduced HSC in vivo-repopulating activity and lymphoid potential while increasing functional heterogeneity. Continuous challenge of the hematopoietic system by serial transplantation provoked the clonal collapse of both young and aged hematopoietic systems. Whole-exome sequencing of serially transplanted aged and young hematopoietic clones confirmed oligoclonal hematopoiesis and revealed mutations in at least 27 genes, including nonsense, missense, and deletion mutations in Bcl11b, Hist1h2ac, Npy2r, Notch3, Ptprr, and Top2b.
Collapse
|
174
|
Sousa‐Franco A, Rebelo K, da Rocha ST, Bernardes de Jesus B. LncRNAs regulating stemness in aging. Aging Cell 2019; 18:e12870. [PMID: 30456884 PMCID: PMC6351848 DOI: 10.1111/acel.12870] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 09/18/2018] [Accepted: 09/28/2018] [Indexed: 12/21/2022] Open
Abstract
One of the most outstanding observations from next-generation sequencing approaches was that only 1.5% of our genes code for proteins. The biggest part is transcribed but give rise to different families of RNAs without coding potential. The functional relevance of these abundant transcripts remains far from elucidated. Among them are the long non-coding RNAs (lncRNAs), a relatively large and heterogeneous group of RNAs shown to be highly tissue-specific, indicating a prominent role in processes controlling cellular identity. In particular, lncRNAs have been linked to both stemness properties and detrimental pathways regulating the aging process, being novel players in the intricate network guiding tissue homeostasis. Here, we summarize the up-to-date information on the role of lncRNAs that affect stemness and hence impact upon aging, highlighting the likelihood that lncRNAs may represent an unexploited reservoir of potential therapeutic targets for reprogramming applications and aging-related diseases.
Collapse
Affiliation(s)
- António Sousa‐Franco
- Instituto de Medicina MolecularFaculdade de Medicina da Universidade de LisboaLisboaPortugal
| | - Kenny Rebelo
- Instituto de Medicina MolecularFaculdade de Medicina da Universidade de LisboaLisboaPortugal
| | - Simão Teixeira da Rocha
- Instituto de Medicina MolecularFaculdade de Medicina da Universidade de LisboaLisboaPortugal
| | - Bruno Bernardes de Jesus
- Instituto de Medicina MolecularFaculdade de Medicina da Universidade de LisboaLisboaPortugal
- Department of Medical Sciences and Institute of Biomedicine—iBiMEDUniversity of AveiroAveiroPortugal
| |
Collapse
|
175
|
Heterogeneity of Stem Cells in the Hippocampus. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1169:31-53. [DOI: 10.1007/978-3-030-24108-7_2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
176
|
Abstract
A number of recent epidemiological studies have associated the clonal expansion of hematopoietic cells, a process referred to as clonal hematopoiesis, with increased mortality. Clonal hematopoiesis increases the risk of hematological cancer, but this overall risk cannot account for the increase in mortality in the general population. Surprisingly, these mutations have also been associated with higher rates of cardiovascular disease, suggesting a previously unrecognized link between somatic mutations in hematopoietic cells and chronic disease. Here, we review recent epidemiological and experimental studies on clonal hematopoiesis that relate to cardiovascular disease.
Collapse
Affiliation(s)
- Soichi Sano
- Hematovascular Biology Center, Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine
| | - Ying Wang
- Hematovascular Biology Center, Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine
| | - Kenneth Walsh
- Hematovascular Biology Center, Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine
| |
Collapse
|
177
|
Arnhold S, Elashry MI, Klymiuk MC, Wenisch S. Biological macromolecules and mesenchymal stem cells: Basic research for regenerative therapies in veterinary medicine. Int J Biol Macromol 2018; 123:889-899. [PMID: 30452985 DOI: 10.1016/j.ijbiomac.2018.11.158] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 11/05/2018] [Accepted: 11/15/2018] [Indexed: 02/07/2023]
Affiliation(s)
- Stefan Arnhold
- Institute of Veterinary Anatomy-, Histology and -Embryology, University of Giessen, 35392 Giessen, Germany
| | - Mohamed I Elashry
- Institute of Veterinary Anatomy-, Histology and -Embryology, University of Giessen, 35392 Giessen, Germany; Anatomy and Embryology Department, Faculty of Veterinary Medicine, University of Mansoura 35516, Egypt.
| | - Michele C Klymiuk
- Institute of Veterinary Anatomy-, Histology and -Embryology, University of Giessen, 35392 Giessen, Germany
| | - Sabine Wenisch
- Clinic of Small Animals, c/o Institute of Veterinary Anatomy, Histology and Embryology, Justus Liebig University of Giessen 35392, Giessen, Germany
| |
Collapse
|
178
|
Identity Noise and Adipogenic Traits Characterize Dermal Fibroblast Aging. Cell 2018; 175:1575-1590.e22. [DOI: 10.1016/j.cell.2018.10.012] [Citation(s) in RCA: 110] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Revised: 08/01/2018] [Accepted: 10/02/2018] [Indexed: 01/01/2023]
|
179
|
Vilas JM, Carneiro C, Da Silva-Álvarez S, Ferreirós A, González P, Gómez M, Ortega S, Serrano M, García-Caballero T, González-Barcia M, Vidal A, Collado M. Adult Sox2+ stem cell exhaustion in mice results in cellular senescence and premature aging. Aging Cell 2018; 17:e12834. [PMID: 30129215 PMCID: PMC6156495 DOI: 10.1111/acel.12834] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Revised: 06/25/2018] [Accepted: 07/21/2018] [Indexed: 12/14/2022] Open
Abstract
Aging is characterized by a gradual functional decline of tissues with age. Adult stem and progenitor cells are responsible for tissue maintenance, repair, and regeneration, but during aging, this population of cells is decreased or its activity is reduced, compromising tissue integrity and causing pathologies that increase vulnerability, and ultimately lead to death. The causes of stem cell exhaustion during aging are not clear, and whether a reduction in stem cell function is a cause or a consequence of aging remains unresolved. Here, we took advantage of a mouse model of induced adult Sox2+ stem cell depletion to address whether accelerated stem cell depletion can promote premature aging. After a short period of partial repetitive depletion of this adult stem cell population in mice, we observed increased kyphosis and hair graying, and reduced fat mass, all of them signs of premature aging. It is interesting that cellular senescence was identified in kidney after this partial repetitive Sox2+ cell depletion. To confirm these observations, we performed a prolonged protocol of partial repetitive depletion of Sox2+ cells, forcing regeneration from the remaining Sox2+ cells, thereby causing their exhaustion. Senescence specific staining and the analysis of the expression of genetic markers clearly corroborated that adult stem cell exhaustion can lead to cellular senescence induction and premature aging.
Collapse
Affiliation(s)
- Jéssica M. Vilas
- Laboratorio de Células Madre en Cáncer y Envejecimiento, Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS); Xerencia de Xestión Integrada de Santiago (XXIS/SERGAS); Santiago de Compostela Spain
| | - Carmen Carneiro
- Departamento de Fisioloxía and Centro de Investigación en Medicina Molecular (CIMUS), Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS); Universidade de Santiago de Compostela; Santiago de Compostela Spain
| | - Sabela Da Silva-Álvarez
- Laboratorio de Células Madre en Cáncer y Envejecimiento, Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS); Xerencia de Xestión Integrada de Santiago (XXIS/SERGAS); Santiago de Compostela Spain
| | - Alba Ferreirós
- Laboratorio de Células Madre en Cáncer y Envejecimiento, Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS); Xerencia de Xestión Integrada de Santiago (XXIS/SERGAS); Santiago de Compostela Spain
| | - Patricia González
- Histopathology Core Unit; Spanish National Cancer Research Centre (CNIO); Madrid Spain
| | - María Gómez
- Histopathology Core Unit; Spanish National Cancer Research Centre (CNIO); Madrid Spain
| | - Sagrario Ortega
- Trasgenic Mice Unit; Spanish National Cancer Research Centre (CNIO); Madrid Spain
| | - Manuel Serrano
- Tumor Suppression Group; Spanish National Cancer Research Centre (CNIO); Madrid Spain
- Institute for Research in Biomedicine (IRB Barcelona); The Barcelona Institute of Science and Technology (BIST); Barcelona Spain
- Catalan Institution for Research and Advanced Studies (ICREA); Barcelona Spain
| | - Tomás García-Caballero
- Departamento de Ciencias Morfológicas, Facultad de Medicina; USC, Xerencia de Xestión Integrada de Santiago (XXIS/SERGAS); Santiago de Compostela Spain
| | - Miguel González-Barcia
- Servicio de Farmacia; Xerencia de Xestión Integrada de Santiago (XXIS/SERGAS); Santiago de Compostela Spain
| | - Anxo Vidal
- Departamento de Fisioloxía and Centro de Investigación en Medicina Molecular (CIMUS), Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS); Universidade de Santiago de Compostela; Santiago de Compostela Spain
| | - Manuel Collado
- Laboratorio de Células Madre en Cáncer y Envejecimiento, Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS); Xerencia de Xestión Integrada de Santiago (XXIS/SERGAS); Santiago de Compostela Spain
| |
Collapse
|
180
|
Abstract
Longer human lives have led to a global burden of late-life disease. However, some older people experience little ill health, a trait that should be extended to the general population. Interventions into lifestyle, including increased exercise and reduction in food intake and obesity, can help to maintain healthspan. Altered gut microbiota, removal of senescent cells, blood factors obtained from young individuals and drugs can all improve late-life health in animals. Application to humans will require better biomarkers of disease risk and responses to interventions, closer alignment of work in animals and humans, and increased use of electronic health records, biobank resources and cohort studies.
Collapse
|
181
|
Effect of heterochromatin stability on intestinal stem cell aging in Drosophila. Mech Ageing Dev 2018; 173:50-60. [DOI: 10.1016/j.mad.2018.04.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Revised: 03/21/2018] [Accepted: 04/02/2018] [Indexed: 12/12/2022]
|
182
|
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
|
183
|
Rattan SIS. Biogerontology: research status, challenges and opportunities. ACTA BIO-MEDICA : ATENEI PARMENSIS 2018; 89:291-301. [PMID: 29957767 PMCID: PMC6179011 DOI: 10.23750/abm.v89i2.7403] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Accepted: 03/29/2018] [Indexed: 01/09/2023]
Abstract
Biogerontology is the study of the biological basis of ageing and age-related diseases. The phenomenon and the process of ageing are well understood in evolutionary and biological terms; and a conceptual framework has been established within which general principles of ageing and longevity can be formulated. The phenotype of ageing in terms of progressive loss of physical function and fitness is best seen during the period of survival after the evolution-determined essential lifespan (ELS) of a species. However, the ageing phenotype is highly heterogenous and individualistic at all levels from the whole body to the molecular one. Most significantly, the process and the progression of ageing are not determined by any specific gerontogenes. Ageing is the result of imperfect maintenance and repair systems that allow a progressive shrinkage of the homeodynamic space of an individual. The challenge is to develop and apply wholistic approaches to the complex trait of ageing for maintaining and/or improving health. One such approach is that of mild stress-induced physiological hormesis by physical, mental and nutritional hormetins. Biogerontological research offers numerous opportunities for developing evidence-based novel biomedical technologies for maintaining and improving health, for preventing the onset of age-related diseases, and for extending the health-span.
Collapse
Affiliation(s)
- Suresh I S Rattan
- Laboratory of Cellular Ageing, Department of Molecular Biology and Genetics, Aarhus University, Denmark.
| |
Collapse
|
184
|
Rožman P. The potential of non-myeloablative heterochronous autologous hematopoietic stem cell transplantation for extending a healthy life span. GeroScience 2018; 40:221-242. [PMID: 29948868 PMCID: PMC6060192 DOI: 10.1007/s11357-018-0027-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Accepted: 06/03/2018] [Indexed: 12/11/2022] Open
Abstract
Aging is a complex multifactorial process, a prominent component being the senescence of the immune system. Consequently, immune-related diseases develop, including atherosclerosis, cancer, and life-threatening infections, which impact on health and longevity. Rejuvenating the aged immune system could mitigate these diseases, thereby contributing to longevity and health. Currently, an appealing option for rejuvenating the immune system is heterochronous autologous hematopoietic stem cell transplantation (haHSCT), where healthy autologous bone marrow/peripheral blood stem cells are collected during the youth of an individual, cryopreserved, and re-infused when he or she has reached an older age. After infusion, young hematopoietic stem cells can reconstitute the compromised immune system and improve immune function. Several studies using animal models have achieved substantial extension of the life span of animals treated with haHSCT. Therefore, haHSCT could be regarded as a potential procedure for preventing age-related immune defects and extending healthy longevity. In this review, the pros, cons, and future feasibility of this approach are discussed.
Collapse
Affiliation(s)
- Primož Rožman
- Blood Transfusion Centre of Slovenia, Šlajmerjeva 6, 1000, Ljubljana, Slovenia.
| |
Collapse
|
185
|
Accelerated aging in schizophrenia and related disorders: Future research. Schizophr Res 2018; 196:4-8. [PMID: 28689755 DOI: 10.1016/j.schres.2017.06.034] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2017] [Revised: 06/15/2017] [Accepted: 06/19/2017] [Indexed: 01/17/2023]
Abstract
Several lines of evidence suggest schizophrenia is a segmental progeria, that is, some but not all aspects of accelerated aging may be present. However, the evidence has not been consistent. Problems with matching and confounding may account for some of these discrepancies. Given the etiopathophysiological heterogeneity of schizophrenia, it is possible that only a specific pathophysiological group within schizophrenia is associated with progeroid features, while others are not, or that one group is associated with a particular segment of aging features, while other progeroid features are found in another pathophysiological subgroup. In the aging research field, significant progress has been made in identifying the molecular pathways that confer aging: epigenetic changes, inflammation, proteostasis, adult stem cell function, metabolic changes, and adaptation to stress, and macromolecular damage. In addition to replication and clarification of existing kinds of evidence, examining these aging pathways would improve our understanding of progeria in schizophrenia.
Collapse
|
186
|
Xie L, Yin A, Nichenko AS, Beedle AM, Call JA, Yin H. Transient HIF2A inhibition promotes satellite cell proliferation and muscle regeneration. J Clin Invest 2018. [PMID: 29533927 PMCID: PMC5983316 DOI: 10.1172/jci96208] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
The remarkable regeneration capability of skeletal muscle depends on the coordinated proliferation and differentiation of satellite cells (SCs). The self-renewal of SCs is critical for long-term maintenance of muscle regeneration potential. Hypoxia profoundly affects the proliferation, differentiation, and self-renewal of cultured myoblasts. However, the physiological relevance of hypoxia and hypoxia signaling in SCs in vivo remains largely unknown. Here, we demonstrate that SCs are in an intrinsic hypoxic state in vivo and express hypoxia-inducible factor 2A (HIF2A). HIF2A promotes the stemness and long-term homeostatic maintenance of SCs by maintaining their quiescence, increasing their self-renewal, and blocking their myogenic differentiation. HIF2A stabilization in SCs cultured under normoxia augments their engraftment potential in regenerative muscle. Conversely, HIF2A ablation leads to the depletion of SCs and their consequent regenerative failure in the long-term. In contrast, transient pharmacological inhibition of HIF2A accelerates muscle regeneration by increasing SC proliferation and differentiation. Mechanistically, HIF2A induces the quiescence and self-renewal of SCs by binding the promoter of the Spry1 gene and activating Spry1 expression. These findings suggest that HIF2A is a pivotal mediator of hypoxia signaling in SCs and may be therapeutically targeted to improve muscle regeneration.
Collapse
Affiliation(s)
- Liwei Xie
- Department of Biochemistry and Molecular Biology.,Center for Molecular Medicine, and
| | - Amelia Yin
- Department of Biochemistry and Molecular Biology.,Center for Molecular Medicine, and
| | - Anna S Nichenko
- Department of Kinesiology, The University of Georgia, Athens, Georgia, USA
| | - Aaron M Beedle
- Department of Pharmaceutical Sciences, Binghamton University-SUNY, Binghamton, New York, USA
| | - Jarrod A Call
- Department of Kinesiology, The University of Georgia, Athens, Georgia, USA.,Regenerative Bioscience Center, The University of Georgia, Athens, Georgia, USA
| | - Hang Yin
- Department of Biochemistry and Molecular Biology.,Center for Molecular Medicine, and
| |
Collapse
|
187
|
Muñoz MF, Argüelles S, Guzman-Chozas M, Guillén-Sanz R, Franco JM, Pintor-Toro JA, Cano M, Ayala A. Cell tracking, survival, and differentiation capacity of adipose-derived stem cells after engraftment in rat tissue. J Cell Physiol 2018; 233:6317-6328. [PMID: 29319169 DOI: 10.1002/jcp.26439] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Accepted: 01/05/2018] [Indexed: 12/30/2022]
Abstract
Adipose tissue is an important source of adipose derived stem cells (ADSCs). These cells have the potential of being used for certain therapies, in which the main objective is to recover the function of a tissue/organ affected by a disease. In order to contribute to repair of the tissue, these cells should be able to survive and carry out their functions in unfavorable conditions after being transplanted. This process requires a better understanding of the biology involved: such as the time cells remain in the implant site, how long they stay there, and whether or not they differentiate into host tissue cells. This report focuses on these questions. ADSC were injected into three different tissues (substantia nigra, ventricle, liver) and they were tracked in vivo with a dual GFP-Luc reporter system. The results show that ADSCs were able to survive up to 4 months after the engraftment and some of them started showing resident cell tissue phenotype. These results demonstrate their long-term capacity of survival and differentiation when injected in vivo.
Collapse
Affiliation(s)
- Mario F Muñoz
- Departamento de Bioquímica y Biología Molecular, Universidad de Sevilla, Sevilla, Spain
| | - Sandro Argüelles
- Departamento de Fisiología, Universidad de Sevilla, Sevilla, Spain
| | - Matias Guzman-Chozas
- Departamento de Nutrición, Bromatología, Toxicología y Medicina Legal, . Facultad de Farmacia, Universidad de Sevilla, Sevilla, Spain
| | - Remedios Guillén-Sanz
- Departamento de Nutrición, Bromatología, Toxicología y Medicina Legal, . Facultad de Farmacia, Universidad de Sevilla, Sevilla, Spain
| | - Jaime M Franco
- Centro Andaluz de Biología Molecular y Medicina Regenerativa (CABIMER), Departamento de Señalización Celular, Universidad de Sevilla, Sevilla, Spain
| | - José A Pintor-Toro
- Centro Andaluz de Biología Molecular y Medicina Regenerativa (CABIMER), Departamento de Señalización Celular, Universidad de Sevilla, Sevilla, Spain
| | - Mercedes Cano
- Departamento de Fisiología, Universidad de Sevilla, Sevilla, Spain
| | - Antonio Ayala
- Departamento de Bioquímica y Biología Molecular, Universidad de Sevilla, Sevilla, Spain
| |
Collapse
|
188
|
Watanabe R, Kanno SI, Mohammadi Roushandeh A, Ui A, Yasui A. Nucleosome remodelling, DNA repair and transcriptional regulation build negative feedback loops in cancer and cellular ageing. Philos Trans R Soc Lond B Biol Sci 2018; 372:rstb.2016.0473. [PMID: 28847829 DOI: 10.1098/rstb.2016.0473] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/17/2017] [Indexed: 12/12/2022] Open
Abstract
Nucleosome remodelling (NR) regulates transcription in an ATP-dependent manner, and influences gene expression required for development and cellular functions, including those involved in anti-cancer and anti-ageing processes. ATP-utilizing chromatin assembly and remodelling factor (ACF) and Brahma-associated factor (BAF) complexes, belonging to the ISWI and SWI/SNF families, respectively, are involved in various types of DNA repair. Suppression of several BAF factors makes U2OS cells significantly sensitive to X-rays, UV and especially to cisplatin, and these BAF factors contribute to the accumulation of repair proteins at various types of DNA damage and to DNA repair. Recent cancer genome sequencing and expression analysis has shown that BAF factors are frequently mutated or, more frequently, silenced in various types of cancer cells. Thus, those cancer cells are potentially X-ray- and especially cisplatin-sensitive, suggesting a way of optimizing current cancer therapy. Recent single-stem cell analysis suggests that mutations and epigenetic changes influence stem cell functionality leading to cellular ageing. Genetic and epigenetic changes in the BAF factors diminish DNA repair as well as transcriptional regulation activities, and DNA repair defects in turn negatively influence NR and transcriptional regulation. Thus, they build negative feedback loops, which accelerate both cellular senescence and transformation as common and rare cellular events, respectively, causing cellular ageing.This article is part of the themed issue 'Chromatin modifiers and remodellers in DNA repair and signalling'.
Collapse
Affiliation(s)
- Reiko Watanabe
- Division of Dynamic Proteome and IDAC Fellow Research Group for DNA Repair and Dynamic Proteome Institute of Development, Aging and Cancer (IDAC), Tohoku University, Sendai 980-8575, Japan
| | - Shin-Ichiro Kanno
- Division of Dynamic Proteome and IDAC Fellow Research Group for DNA Repair and Dynamic Proteome Institute of Development, Aging and Cancer (IDAC), Tohoku University, Sendai 980-8575, Japan
| | - Amaneh Mohammadi Roushandeh
- Division of Dynamic Proteome and IDAC Fellow Research Group for DNA Repair and Dynamic Proteome Institute of Development, Aging and Cancer (IDAC), Tohoku University, Sendai 980-8575, Japan
| | - Ayako Ui
- Division of Dynamic Proteome and IDAC Fellow Research Group for DNA Repair and Dynamic Proteome Institute of Development, Aging and Cancer (IDAC), Tohoku University, Sendai 980-8575, Japan
| | - Akira Yasui
- Division of Dynamic Proteome and IDAC Fellow Research Group for DNA Repair and Dynamic Proteome Institute of Development, Aging and Cancer (IDAC), Tohoku University, Sendai 980-8575, Japan
| |
Collapse
|
189
|
Lv YJ, Yang Y, Sui BD, Hu CH, Zhao P, Liao L, Chen J, Zhang LQ, Yang TT, Zhang SF, Jin Y. Resveratrol counteracts bone loss via mitofilin-mediated osteogenic improvement of mesenchymal stem cells in senescence-accelerated mice. Theranostics 2018; 8:2387-2406. [PMID: 29721087 PMCID: PMC5928897 DOI: 10.7150/thno.23620] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Accepted: 02/18/2018] [Indexed: 01/08/2023] Open
Abstract
Rational: Senescence of mesenchymal stem cells (MSCs) and the related functional decline of osteogenesis have emerged as the critical pathogenesis of osteoporosis in aging. Resveratrol (RESV), a small molecular compound that safely mimics the effects of dietary restriction, has been well documented to extend lifespan in lower organisms and improve health in aging rodents. However, whether RESV promotes function of senescent stem cells in alleviating age-related phenotypes remains largely unknown. Here, we intend to investigate whether RESV counteracts senescence-associated bone loss via osteogenic improvement of MSCs and the underlying mechanism. Methods: MSCs derived from bone marrow (BMMSCs) and the bone-specific, senescence-accelerated, osteoblastogenesis/osteogenesis-defective mice (the SAMP6 strain) were used as experimental models. In vivo application of RESV was performed at 100 mg/kg intraperitoneally once every other day for 2 months, and in vitro application of RESV was performed at 10 μM. Bone mass, bone formation rates and osteogenic differentiation of BMMSCs were primarily evaluated. Metabolic statuses of BMMSCs and the mitochondrial activity, transcription and morphology were also examined. Mitofilin expression was assessed at both mRNA and protein levels, and short hairpin RNA (shRNA)-based gene knockdown was applied for mechanistic experiments. Results: Chronic intermittent application of RESV enhances bone formation and counteracts accelerated bone loss, with RESV improving osteogenic differentiation of senescent BMMSCs. Furthermore, in rescuing osteogenic decline under BMMSC senescence, RESV restores cellular metabolism through mitochondrial functional recovery via facilitating mitochondrial autonomous gene transcription. Molecularly, in alleviating senescence-associated mitochondrial disorders of BMMSCs, particularly the mitochondrial morphological alterations, RESV upregulates Mitofilin, also known as inner membrane protein of mitochondria (Immt) or Mic60, which is the core component of the mitochondrial contact site and cristae organizing system (MICOS). Moreover, Mitofilin is revealed to be indispensable for mitochondrial homeostasis and osteogenesis of BMMSCs, and that insufficiency of Mitofilin leads to BMMSC senescence and bone loss. More importantly, Mitofilin mediates resveratrol-induced mitochondrial and osteogenic improvements of BMMSCs in senescence. Conclusion: Our findings uncover osteogenic functional improvements of senescent MSCs as critical impacts in anti-osteoporotic practice of RESV, and unravel Mitofilin as a novel mechanism mediating RESV promotion on mitochondrial function in stem cell senescence.
Collapse
|
190
|
López-Lázaro M. The stem cell division theory of cancer. Crit Rev Oncol Hematol 2018; 123:95-113. [DOI: 10.1016/j.critrevonc.2018.01.010] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2017] [Revised: 11/13/2017] [Accepted: 01/17/2018] [Indexed: 02/07/2023] Open
|
191
|
Howard BH, Hirai TH, Russanova VR. Epigenome comparisons reveal linkage between gene expression and postnatal remodeling of chromatin domain topology. PLoS One 2018; 13:e0191033. [PMID: 29466355 PMCID: PMC5821309 DOI: 10.1371/journal.pone.0191033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Accepted: 12/27/2017] [Indexed: 11/19/2022] Open
Abstract
Substantial evidence has accumulated linking epigenome change to alterations in stem cell function during postnatal development and aging. Yet much remains to be learned about causal relationships, and large gaps remain in our understanding of epigenome-transcriptome interactions. Here we investigate structural features of large histone H3K27me3-enriched regions in human stem cell-like monocytes and their dendritic cell derivatives, where the H3K27me3 modification is considered to demarcate Polycomb (PcG) domains. Both differentiation- and postnatal development-related change are explored, initially by confirming expected reciprocal relationships between transcript abundance and span of PcG domains overlapping transcribed regions. PcG-associated postnatal transcriptome change specific to the stem cell-like monocytes is found to be incompletely explained by conventional measures of PcG region structure. To address this, we introduce algorithms that quantify local nucleosome-scale conservation of PcG-region topology. It is shown that topology-based comparisons can reveal broad statistical linkage between postnatal gene down-regulation and epigenome remodeling; further, such comparisons provide access to a previously unexplored dimension of epigenome architecture.
Collapse
Affiliation(s)
- Bruce H. Howard
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, United States of America
- * E-mail:
| | - Tazuko H. Hirai
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Valya R. Russanova
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, United States of America
| |
Collapse
|
192
|
Ruan L, Zhang X, Li R. Recent insights into the cellular and molecular determinants of aging. J Cell Sci 2018; 131:131/3/jcs210831. [PMID: 29420249 DOI: 10.1242/jcs.210831] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Aging is the gradual decline of physiological functions and organismal fitness, which leads to age-dependent fitness loss, diseases and eventually mortality. Understanding the cause of aging constitutes one of most intriguing areas of research in biology. On both the cellular and molecular levels, it has been hypothesized that there are aging determinants to control the onset and progression of aging, including the loss of beneficial components and accumulation of detrimental factors. This Review highlights the recent advance in identifying various factors that affect the aging process, focusing on how these determinants affect the lifespan and fitness of a cell or organism. With more and more aging determinants revealed, further understanding about their functions and interconnections could enable the development of specific intervention to extend healthy lifespan and reduce the risk of age-related diseases.
Collapse
Affiliation(s)
- Linhao Ruan
- Center for Cell Dynamics, Department of Cell Biology, Johns Hopkins University School of Medicine, 855 North Wolfe Street, Baltimore, MD 21205, USA.,Biochemistry, Cellular and Molecular Biology (BCMB) Graduate Program, Johns Hopkins University School of Medicine, 1830 E. Monument Street, Baltimore, MD 21287, USA
| | - Xi Zhang
- Center for Cell Dynamics, Department of Cell Biology, Johns Hopkins University School of Medicine, 855 North Wolfe Street, Baltimore, MD 21205, USA
| | - Rong Li
- Center for Cell Dynamics, Department of Cell Biology, Johns Hopkins University School of Medicine, 855 North Wolfe Street, Baltimore, MD 21205, USA .,Department of Chemical and Biomolecular Engineering, Whiting School of Engineering, Johns Hopkins University, 3400 North Charles Street, Baltimore, MD 21218, USA
| |
Collapse
|
193
|
Abstract
Abstract
Hematopoietic stem cells (HSCs) ensure a balanced production of all blood cells throughout life. As they age, HSCs gradually lose their self-renewal and regenerative potential, whereas the occurrence of cellular derailment strongly increases. Here we review our current understanding of the molecular mechanisms that contribute to HSC aging. We argue that most of the causes that underlie HSC aging result from cell-intrinsic pathways, and reflect on which aspects of the aging process may be reversible. Because many hematological pathologies are strongly age-associated, strategies to intervene in aspects of the stem cell aging process may have significant clinical relevance.
Collapse
|
194
|
Affiliation(s)
- Michael A Bellio
- From the Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, FL
| | - Aisha Khan
- From the Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, FL.
| |
Collapse
|
195
|
Costantino S, Camici GG, Mohammed SA, Volpe M, Lüscher TF, Paneni F. Epigenetics and cardiovascular regenerative medicine in the elderly. Int J Cardiol 2018; 250:207-214. [PMID: 28988828 DOI: 10.1016/j.ijcard.2017.09.188] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Accepted: 09/22/2017] [Indexed: 12/21/2022]
|
196
|
Cesselli D, Aleksova A, Mazzega E, Caragnano A, Beltrami AP. Cardiac stem cell aging and heart failure. Pharmacol Res 2018; 127:26-32. [DOI: 10.1016/j.phrs.2017.01.013] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Revised: 01/07/2017] [Accepted: 01/11/2017] [Indexed: 12/11/2022]
|
197
|
Migliaccio AR. A vicious interplay between genetic and environmental insults in the etiology of blood cancers. Exp Hematol 2017; 59:9-13. [PMID: 29248611 DOI: 10.1016/j.exphem.2017.12.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 12/06/2017] [Accepted: 12/07/2017] [Indexed: 02/06/2023]
Abstract
Over the years, the etiology of cancer has been attributed alternatively to genetic and environmental insults. According to the genetic hypothesis, cancer cells arise from the acquisition of mutations that dysregulate the intrinsic mechanisms controlling normal cell growth and survival. In contrast, the environmental hypothesis holds that cancer can be caused by multiple extrinsic challenges that alter normal tissue homeostasis, but may not necessarily involve mutations. It is, however, quite possible that these two mechanisms are not mutually exclusive. According to this third hypothesis, environmental challenges, by mechanisms still poorly understood, may act by imposing a selection pressure that confers a proliferative advantage on cells carrying specific driver mutations, leading to disease initiation. The authors of a brief report published in this journal (Exp Hematol. 2017;56:1-6) tested this third hypothesis experimentally and provide new evidence that chronic inflammation, by increasing tumor necrosis factor (TNF)-α, may positively select malignant hematopoietic stem cells (HSCs) carrying loss-of-function TET2 mutations that switch the TNF-α signaling responses to activate proliferation rather than inducing quiescence. Furthermore, these mutations skew hematopoietic differentiation toward the myelomonocytic lineage and the output of macrophages producing TNF-α constitutively, promoting further environment-independent expansion of the malignant HSCs. These findings support a model in which the formation of a malignant population is triggered by a vicious interplay between genetic (TET2 mutations) and environmental (inflammation) insults, suggesting the need for strategies that target both the malignant cells and their environment.
Collapse
Affiliation(s)
- Anna Rita Migliaccio
- Department of Biomedical and Neuromotorial Sciences, Alma Mater University, Bologna, Italy; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai (ISMMS), New York, New York.
| |
Collapse
|
198
|
Liu P, Zhao H, Luo Y. Anti-Aging Implications of Astragalus Membranaceus (Huangqi): A Well-Known Chinese Tonic. Aging Dis 2017; 8:868-886. [PMID: 29344421 PMCID: PMC5758356 DOI: 10.14336/ad.2017.0816] [Citation(s) in RCA: 205] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Accepted: 08/16/2017] [Indexed: 12/20/2022] Open
Abstract
Owing to a dramatic increase in average life expectancy and the Family Planning program of the 1970s - 1990s, China is rapidly becoming an aging society. Therefore, the investigation of healthspan-extending drugs becomes more urgent. Astragalus membranaceus (Huangqi) is a major medicinal herb that has been commonly used in many herbal formulations in the practice of traditional Chinese medicine (TCM) to treat a wide variety of diseases and body disorders, or marketed as life-prolonging extracts for human use in China, for more than 2000 years. The major components of Astragalus membranaceus are polysaccharides, flavonoids, and saponins. Pharmacological research indicates that the extract component of Astragalus membranaceus can increase telomerase activity, and has antioxidant, anti-inflammatory, immunoregulatory, anticancer, hypolipidemic, antihyperglycemic, hepatoprotective, expectorant, and diuretic effects. A proprietary extract of the dried root of Astragalus membranaceus, called TA-65, was associated with a significant age-reversal effect in the immune system. Our review focuses on the function and the underlying mechanisms of Astragalus membranaceus in lifespan extension, anti-vascular aging, anti-brain aging, and anti-cancer effects, based on experimental and clinical studies.
Collapse
Affiliation(s)
- Ping Liu
- 1Cerebrovascular Diseases Research Institute, and Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Haiping Zhao
- 1Cerebrovascular Diseases Research Institute, and Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Yumin Luo
- 1Cerebrovascular Diseases Research Institute, and Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China.,2Center of Stroke, Beijing Institute for Brain Disorders, Beijing, China.,3Beijing Key Laboratory of Translational Medicine for Cerebrovascular Diseases, Beijing, China
| |
Collapse
|
199
|
Abstract
Although the core concept of remyelination - based on the activation, migration, proliferation and differentiation of CNS progenitors - has not changed over the past 20 years, our understanding of the detailed mechanisms that underlie this process has developed considerably. We can now decorate the central events of remyelination with a host of pathways, molecules, mediators and cells, revealing a complex and precisely orchestrated process. These advances have led to recent drug-based and cell-based clinical trials for myelin diseases and have opened up hitherto unrecognized opportunities for drug-based approaches to therapeutically enhance remyelination.
Collapse
|
200
|
Mashinchian O, Pisconti A, Le Moal E, Bentzinger CF. The Muscle Stem Cell Niche in Health and Disease. Curr Top Dev Biol 2017; 126:23-65. [PMID: 29305000 DOI: 10.1016/bs.ctdb.2017.08.003] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
The regulation of stem cells that maintain and regenerate postnatal tissues depends on extrinsic signals originating from their microenvironment, commonly referred to as the stem cell niche. Complex higher-order regulatory interrelationships with the tissue and factors in the systemic circulation are integrated and propagated to the stem cells through the niche. The stem cell niche in skeletal muscle tissue is both a paradigm for a structurally and functionally relatively static niche that maintains stem cell quiescence during tissue homeostasis, and a highly dynamic regenerative niche that is subject to extensive structural remodeling and a flux of different support cell populations. Conditions ranging from aging to chronically degenerative skeletal muscle diseases affect the composition of the niche and thereby impair the regenerative potential of muscle stem cells. A holistic and integrative understanding of the extrinsic mechanisms regulating muscle stem cells in health and disease in a broad systemic context will be imperative for the identification of regulatory hubs in the niche interactome that can be targeted to maintain, restore, or enhance the regenerative capacity of muscle tissue. Here, we review the microenvironmental regulation of muscle stem cells, summarize how niche dysfunction can contribute to disease, and discuss emerging therapeutic implications.
Collapse
Affiliation(s)
- Omid Mashinchian
- Nestlé Institute of Health Sciences, Lausanne, Switzerland; École Polytechnique Fédérale de Lausanne, Doctoral Program in Biotechnology and Bioengineering, Lausanne, Switzerland
| | - Addolorata Pisconti
- Institute of Integrative Biology, University of Liverpool, Liverpool, United Kingdom
| | - Emmeran Le Moal
- Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - C Florian Bentzinger
- Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, QC, Canada.
| |
Collapse
|