1
|
Frost OG, Ramkilawan P, Rebbaa A, Stolzing A. A systematic review of lifespan studies in rodents using stem cell transplantations. Ageing Res Rev 2024; 97:102295. [PMID: 38588866 DOI: 10.1016/j.arr.2024.102295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Revised: 03/27/2024] [Accepted: 04/03/2024] [Indexed: 04/10/2024]
Abstract
Organismal aging involves the progressive decline in organ function and increased susceptibility to age-associated diseases. Regardless of its origin, cellular aging is consequently reflected at the level of organ and associated systems dysfunction. Aging of stem cell populations within the body and their decreased ability to self-renew, differentiate, and regenerate damaged tissues, is a key contributor to organismal decline. Based on this, supplementing young stem cells may delay tissue aging, improve frailty and extend health and lifespan. This review investigates studies in rodents using stem cell transplantation from either mice or human donors. The aim is to consolidate available information on the efficacy of stem cell therapies in rodent models and provide insights to guide further research efforts. Out of the 21 studies included in this review, the methodology varied significantly including the lifespan measurement. To enable comparison the median lifespan was calculated using WebPlotDigitizer 4.6 if not provided by the literature. A total of 18 out of 21 studies evidenced significant lifespan extension post stem cell transplant, with 7 studies demonstrating benefits in reduced frailty and other aging complications.
Collapse
Affiliation(s)
- Oliver G Frost
- Centre for Biological Engineering, School of Mechanical, Electrical and Manufacturing Engineering, Loughborough University, Loughborough LE11 3TU, UK; SENS Research Foundation, Mountain View, CA 94041, USA
| | | | | | - Alexandra Stolzing
- Centre for Biological Engineering, School of Mechanical, Electrical and Manufacturing Engineering, Loughborough University, Loughborough LE11 3TU, UK.
| |
Collapse
|
2
|
Kakridonis F, Pneumatikos SG, Vakonaki E, Berdiaki A, Tzatzarakis MN, Fragkiadaki P, Spandidos DA, Baliou S, Ioannou P, Hatzidaki E, Nikitovic D, Tsatsakis A, Vasiliadis E. Telomere length as a predictive biomarker in osteoporosis (Review). Biomed Rep 2023; 19:87. [PMID: 37881605 PMCID: PMC10594068 DOI: 10.3892/br.2023.1669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Accepted: 09/22/2023] [Indexed: 10/27/2023] Open
Abstract
Telomeres are the ends of chromosomes that protect them from DNA damage. There is evidence to suggest that telomere shortening appears with advanced age. Since aging is a significant risk factor for developing age-related complications, it is plausible that telomere shortening may be involved in the development of osteoporosis. The present review summarizes the potential of telomere shortening as a biomarker for detecting the onset of osteoporosis. For the purposes of the present review, the following scientific databases were searched for relevant articles: PubMed/NCBI, Cochrane Library of Systematic Reviews, Scopus, Embase and Google Scholar. The present review includes randomized and non-randomized controlled studies and case series involving humans, irrespective of the time of their publication. In six out of the 11 included studies providing data on humans, there was at least a weak association between telomere length and osteoporosis, with the remaining studies exhibiting no such association. As a result, telomere shortening may be used as a biomarker or as part of a panel of biomarkers for tracking the onset and progression of osteoporosis.
Collapse
Affiliation(s)
- Fotios Kakridonis
- 5th Department of Orthopaedics, KAT Attica General Hospital, 14561 Athens, Greece
| | - Spyros G. Pneumatikos
- 3rd Department of Orthopaedics, KAT Attica General Hospital, 14561 Athens, Greece
- Department of Orthopaedics, Medical School, Kapodistrian University of Athens, 11527 Athens, Greece
| | - Elena Vakonaki
- Laboratory of Toxicology, Medical School, University of Crete, 71003 Heraklion, Greece
| | - Aikaterini Berdiaki
- Laboratory of Anatomy-Histology-Embryology, Medical School, University of Crete, 71003 Heraklion, Greece
| | | | - Persefoni Fragkiadaki
- Laboratory of Toxicology, Medical School, University of Crete, 71003 Heraklion, Greece
| | - Demetrios A. Spandidos
- Laboratory of Clinical Virology, Medical School, University of Crete, 71003 Heraklion, Greece
| | - Stella Baliou
- Laboratory of Toxicology, Medical School, University of Crete, 71003 Heraklion, Greece
| | - Petros Ioannou
- Laboratory of Internal Medicine, Medical School, University of Crete, 71003 Heraklion, Greece
| | - Eleftheria Hatzidaki
- Department of Neonatology and NICU, University Hospital of Heraklion, 71500 Heraklion, Greece
| | - Dragana Nikitovic
- Laboratory of Anatomy-Histology-Embryology, Medical School, University of Crete, 71003 Heraklion, Greece
| | - Aristidis Tsatsakis
- Laboratory of Toxicology, Medical School, University of Crete, 71003 Heraklion, Greece
| | - Elias Vasiliadis
- 3rd Department of Orthopaedics, KAT Attica General Hospital, 14561 Athens, Greece
| |
Collapse
|
3
|
Wang Y, Gao T, Wang B. Application of mesenchymal stem cells for anti-senescence and clinical challenges. Stem Cell Res Ther 2023; 14:260. [PMID: 37726805 PMCID: PMC10510299 DOI: 10.1186/s13287-023-03497-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 09/13/2023] [Indexed: 09/21/2023] Open
Abstract
Senescence is a hot topic nowadays, which shows the accumulation of senescent cells and inflammatory factors, leading to the occurrence of various senescence-related diseases. Although some methods have been identified to partly delay senescence, such as strengthening exercise, restricting diet, and some drugs, these only slow down the process of senescence and cannot fundamentally delay or even reverse senescence. Stem cell-based therapy is expected to be a potential effective way to alleviate or cure senescence-related disorders in the coming future. Mesenchymal stromal cells (MSCs) are the most widely used cell type in treating various diseases due to their potentials of self-replication and multidirectional differentiation, paracrine action, and immunoregulatory effects. Some biological characteristics of MSCs can be well targeted at the pathological features of aging. Therefore, MSC-based therapy is also a promising strategy to combat senescence-related diseases. Here we review the recent progresses of MSC-based therapies in the research of age-related diseases and the challenges in clinical application, proving further insight and reference for broad application prospects of MSCs in effectively combating senesce in the future.
Collapse
Affiliation(s)
- Yaping Wang
- Clinical Stem Cell Center, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, 210008, People's Republic of China
- Clinical Stem Cell Center, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, People's Republic of China
| | - Tianyun Gao
- Clinical Stem Cell Center, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, 210008, People's Republic of China
| | - Bin Wang
- Clinical Stem Cell Center, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, 210008, People's Republic of China.
| |
Collapse
|
4
|
Jazbec K, Jež M, Švajger U, Smrekar B, Miceska S, Rajčevič U, Justin M, Završnik J, Malovrh T, Švara T, Gombač M, Ramšak Ž, Rožman P. The Influence of Heterochronic Non-Myeloablative Bone Marrow Transplantation on the Immune System, Frailty, General Health, and Longevity of Aged Murine Recipients. Biomolecules 2022; 12:biom12040595. [PMID: 35454183 PMCID: PMC9028083 DOI: 10.3390/biom12040595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 04/06/2022] [Accepted: 04/14/2022] [Indexed: 12/10/2022] Open
Abstract
The stem cell theory of aging postulates that stem cells become inefficient at maintaining the original functions of the tissues. We, therefore, hypothesized that transplanting young bone marrow (BM) to old recipients would lead to rejuvenating effects on immunity, followed by improved general health, decreased frailty, and possibly life span extension. We developed a murine model of non-myeloablative heterochronic BM transplantation in which old female BALB/c mice at 14, 16, and 18(19) months of age received altogether 125.1 ± 15.6 million nucleated BM cells from young male donors aged 7–13 weeks. At 21 months, donor chimerism was determined, and the immune system’s innate and adaptive arms were analyzed. Mice were then observed for general health and frailty until spontaneous death, when their lifespan, post-mortem examinations, and histopathological changes were recorded. The results showed that the old mice developed on average 18.7 ± 9.6% donor chimerism in the BM and showed certain improvements in their innate and adaptive arms of the immune system, such as favorable counts of neutrophils in the spleen and BM, central memory Th cells, effector/effector memory Th and Tc cells in the spleen, and B1a and B1b cells in the peritoneal cavity. Borderline enhanced lymphocyte proliferation capacity was also seen. The frailty parameters, pathomorphological results, and life spans did not differ significantly in the transplanted vs. control group of mice. In conclusion, although several favorable effects are obtained in our heterochronic non-myeloablative transplantation model, additional optimization is needed for better rejuvenation effects.
Collapse
Affiliation(s)
- Katerina Jazbec
- Diagnostic Services, Blood Transfusion Centre of Slovenia, 1000 Ljubljana, Slovenia; (M.J.); (U.Š.); (B.S.); (S.M.); (U.R.); (M.J.); (P.R.)
- Correspondence:
| | - Mojca Jež
- Diagnostic Services, Blood Transfusion Centre of Slovenia, 1000 Ljubljana, Slovenia; (M.J.); (U.Š.); (B.S.); (S.M.); (U.R.); (M.J.); (P.R.)
| | - Urban Švajger
- Diagnostic Services, Blood Transfusion Centre of Slovenia, 1000 Ljubljana, Slovenia; (M.J.); (U.Š.); (B.S.); (S.M.); (U.R.); (M.J.); (P.R.)
- Chair of Clinical Chemistry, Faculty of Pharmacy, University of Ljubljana, Aškerčeva 7, 1000 Ljubljana, Slovenia
| | - Boštjan Smrekar
- Diagnostic Services, Blood Transfusion Centre of Slovenia, 1000 Ljubljana, Slovenia; (M.J.); (U.Š.); (B.S.); (S.M.); (U.R.); (M.J.); (P.R.)
| | - Simona Miceska
- Diagnostic Services, Blood Transfusion Centre of Slovenia, 1000 Ljubljana, Slovenia; (M.J.); (U.Š.); (B.S.); (S.M.); (U.R.); (M.J.); (P.R.)
| | - Uroš Rajčevič
- Diagnostic Services, Blood Transfusion Centre of Slovenia, 1000 Ljubljana, Slovenia; (M.J.); (U.Š.); (B.S.); (S.M.); (U.R.); (M.J.); (P.R.)
| | - Mojca Justin
- Diagnostic Services, Blood Transfusion Centre of Slovenia, 1000 Ljubljana, Slovenia; (M.J.); (U.Š.); (B.S.); (S.M.); (U.R.); (M.J.); (P.R.)
| | - Janja Završnik
- Department of Biochemistry and Molecular and Structural Biology, Jožef Stefan Institute, 1000 Ljubljana, Slovenia;
| | - Tadej Malovrh
- Institute of Microbiology and Parasitology, Veterinary Faculty, University of Ljubljana, 1000 Ljubljana, Slovenia;
| | - Tanja Švara
- Institute of Pathology, Wild Animals, Fish and Bees, Veterinary Faculty, University of Ljubljana, 1000 Ljubljana, Slovenia; (T.Š.); (M.G.)
| | - Mitja Gombač
- Institute of Pathology, Wild Animals, Fish and Bees, Veterinary Faculty, University of Ljubljana, 1000 Ljubljana, Slovenia; (T.Š.); (M.G.)
| | - Živa Ramšak
- National Institute of Biology, 1000 Ljubljana, Slovenia;
| | - Primož Rožman
- Diagnostic Services, Blood Transfusion Centre of Slovenia, 1000 Ljubljana, Slovenia; (M.J.); (U.Š.); (B.S.); (S.M.); (U.R.); (M.J.); (P.R.)
| |
Collapse
|
5
|
Ganoderic Acid D Protects Human Amniotic Mesenchymal Stem Cells against Oxidative Stress-Induced Senescence through the PERK/NRF2 Signaling Pathway. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:8291413. [PMID: 32774686 PMCID: PMC7407022 DOI: 10.1155/2020/8291413] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Revised: 06/19/2020] [Accepted: 06/26/2020] [Indexed: 12/19/2022]
Abstract
Aging is an important risk factor in the occurrence of many chronic diseases. Senescence and exhaustion of adult stem cells are considered as a hallmark of aging in organisms. In this study, a senescent human amniotic mesenchymal stem cell (hAMSC) model subjected to oxidative stress was established in vitro using hydrogen peroxide. We investigated the effects of ganoderic acid D (GA-D), a natural triterpenoid compound produced from Ganoderma lucidum, on hAMSC senescence. GA-D significantly inhibited β-galactosidase (a senescence-associated marker) formation, in a dose-dependent manner, with doses ranging from 0.1 μM to 10 μM, without inducing cytotoxic side-effects. Furthermore, GA-D markedly inhibited the generation of reactive oxygen species (ROS) and the expression of p21 and p16 proteins, relieved the cell cycle arrest, and enhanced telomerase activity in senescent hAMSCs. Furthermore, GA-D upregulated the expression of phosphorylated protein kinase R- (PKR-) like endoplasmic reticulum kinase (PERK), peroxidase III (PRDX3), and nuclear factor-erythroid 2-related factor (NRF2) and promoted intranuclear transfer of NRF2 in senescent cells. The PERK inhibitor GSK2656157 and/or the NRF2 inhibitor ML385 suppressed the PERK/NRF2 signaling, which was activated by GA-D. They induced a rebound for the generation of ROS and β-galactosidase-positive cells and attenuated the differentiation capacity. These findings suggest that GA-D retards hAMSC senescence through activation of the PERK/NRF2 signaling pathway and may be a promising candidate for the discovery of antiaging agents.
Collapse
|
6
|
Ren X, Hu B, Song M, Ding Z, Dang Y, Liu Z, Zhang W, Ji Q, Ren R, Ding J, Chan P, Jiang C, Ye K, Qu J, Tang F, Liu GH. Maintenance of Nucleolar Homeostasis by CBX4 Alleviates Senescence and Osteoarthritis. Cell Rep 2020; 26:3643-3656.e7. [PMID: 30917318 DOI: 10.1016/j.celrep.2019.02.088] [Citation(s) in RCA: 78] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 01/27/2019] [Accepted: 02/21/2019] [Indexed: 01/12/2023] Open
Abstract
CBX4, a component of polycomb repressive complex 1 (PRC1), plays important roles in the maintenance of cell identity and organ development through gene silencing. However, whether CBX4 regulates human stem cell homeostasis remains unclear. Here, we demonstrate that CBX4 counteracts human mesenchymal stem cell (hMSC) aging via the maintenance of nucleolar homeostasis. CBX4 protein is downregulated in aged hMSCs, whereas CBX4 knockout in hMSCs results in destabilized nucleolar heterochromatin, enhanced ribosome biogenesis, increased protein translation, and accelerated cellular senescence. CBX4 maintains nucleolar homeostasis by recruiting nucleolar protein fibrillarin (FBL) and heterochromatin protein KRAB-associated protein 1 (KAP1) at nucleolar rDNA, limiting the excessive expression of rRNAs. Overexpression of CBX4 alleviates physiological hMSC aging and attenuates the development of osteoarthritis in mice. Altogether, our findings reveal a critical role of CBX4 in counteracting cellular senescence by maintaining nucleolar homeostasis, providing a potential therapeutic target for aging-associated disorders.
Collapse
Affiliation(s)
- Xiaoqing Ren
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; 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
| | - Boqiang Hu
- Biodynamic Optical Imaging Center, College of Life Sciences, Peking University, Beijing 100871, China
| | - Moshi Song
- University of Chinese Academy of Sciences, Beijing 100049, China; State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China
| | - Zhichao Ding
- 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
| | - Yujiao Dang
- Biodynamic Optical Imaging Center, College of Life Sciences, Peking University, Beijing 100871, 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
| | - Weiqi 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; Institute for Stem Cell and Regeneration, 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
| | - Qianzhao Ji
- 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
| | - Ruotong Ren
- 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
| | - Jianjian Ding
- Army Diagnosis and Treatment Center for Oral Disease, 306th Hospital of the PLA, Beijing 100101, China
| | - Piu Chan
- Advanced Innovation Center for Human Brain Protection, National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing 100053, China
| | - Changtao Jiang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education and Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing 100191, China
| | - Keqiong Ye
- University of Chinese Academy of Sciences, Beijing 100049, China; Key Laboratory of RNA Biology, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, 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.
| | - Fuchou Tang
- Biodynamic Optical Imaging Center, College of Life Sciences, Peking University, Beijing 100871, China; Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China; Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, Beijing 100871, China; Biomedical Institute for Pioneering Investigation via Convergence, Peking University, Beijing 100871, China.
| | - Guang-Hui Liu
- 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; Advanced Innovation Center for Human Brain Protection, National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing 100053, China; Beijing Institute for Brain Disorders, Capital Medical University, Beijing 100069, China.
| |
Collapse
|
7
|
Oliva AA, McClain-Moss L, Pena A, Drouillard A, Hare JM. Allogeneic mesenchymal stem cell therapy: A regenerative medicine approach to geroscience. Aging Med (Milton) 2019; 2:142-146. [PMID: 31667462 PMCID: PMC6820701 DOI: 10.1002/agm2.12079] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Extraordinary advances in medicine and public health have contributed to increasing life expectancy worldwide. However, health span-"healthy aging"-has paradoxically lagged to parallel this increase. Consequently, aging-associated illnesses, such as Alzheimer's disease and aging frailty, are having a growing impact on patients, their families, and entire health-care systems. Typically, such disorders have been treated as isolated disease entities. However, the inextricable links between aging-associated disorders and the aging process itself have become increasingly recognized, leading to formation of the field of geroscience. The geroscience concept is that treating the aging process itself should lead to treatment and prevention of aging-related disorders. However, the aging process is complex, dictated by highly interrelated pleiotropic processes. As such, therapeutics with pleiotropic mechanisms of action (either alone, or as part of combinatorial strategies) will be required for preventing and treating both aging and related disorders. Mesenchymal stem cells (MSCs) have multiple mechanisms of action that make these highly promising geroscience therapeutic candidates. These cells have a high safety profile for clinical use, are amenable to allogeneic use since tissue-type matching is not required, and can have sustained activity after transplantation. Herein, we review preclinical and clinical data supporting the utility of allogeneic MSCs as a geroscience therapeutic candidate.
Collapse
Affiliation(s)
| | | | | | | | - Joshua M Hare
- Longeveron LLC, Miami, FL, USA.,Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| |
Collapse
|
8
|
Fernandez RJ, Johnson FB. A regulatory loop connecting WNT signaling and telomere capping: possible therapeutic implications for dyskeratosis congenita. Ann N Y Acad Sci 2019; 1418:56-68. [PMID: 29722029 DOI: 10.1111/nyas.13692] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Revised: 03/02/2018] [Accepted: 03/04/2018] [Indexed: 12/15/2022]
Abstract
The consequences of telomere dysfunction are most apparent in rare inherited syndromes caused by genetic deficiencies in factors that normally maintain telomeres. The principal disease is known as dyskeratosis congenita (DC), but other syndromes with similar underlying genetic defects share some clinical aspects with this disease. Currently, there are no curative therapies for these diseases of telomere dysfunction. Here, we review recent findings demonstrating that dysfunctional (i.e., uncapped) telomeres can downregulate the WNT pathway, and that restoration of WNT signaling helps to recap telomeres by increasing expression of shelterins, proteins that naturally bind and protect telomeres. We discuss how these findings are different from previous observations connecting WNT and telomere biology, and discuss potential links between WNT and clinical manifestations of the DC spectrum of diseases. Finally, we argue for exploring the use of WNT agonists, specifically lithium, as a possible therapeutic approach for patients with DC.
Collapse
Affiliation(s)
- Rafael Jesus Fernandez
- Cell and Molecular Biology Program, Biomedical Graduate Studies, Medical Scientist Training Program, Philadelphia, Pennsylvania.,Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - F Brad Johnson
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
9
|
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: 107] [Impact Index Per Article: 21.4] [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
|
10
|
Carrero D, Soria-Valles C, López-Otín C. Hallmarks of progeroid syndromes: lessons from mice and reprogrammed cells. Dis Model Mech 2017; 9:719-35. [PMID: 27482812 PMCID: PMC4958309 DOI: 10.1242/dmm.024711] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Ageing is a process that inevitably affects most living organisms and involves the accumulation of macromolecular damage, genomic instability and loss of heterochromatin. Together, these alterations lead to a decline in stem cell function and to a reduced capability to regenerate tissue. In recent years, several genetic pathways and biochemical mechanisms that contribute to physiological ageing have been described, but further research is needed to better characterize this complex biological process. Because premature ageing (progeroid) syndromes, including progeria, mimic many of the characteristics of human ageing, research into these conditions has proven to be very useful not only to identify the underlying causal mechanisms and identify treatments for these pathologies, but also for the study of physiological ageing. In this Review, we summarize the main cellular and animal models used in progeria research, with an emphasis on patient-derived induced pluripotent stem cell models, and define a series of molecular and cellular hallmarks that characterize progeroid syndromes and parallel physiological ageing. Finally, we describe the therapeutic strategies being investigated for the treatment of progeroid syndromes, and their main limitations. Summary: This Review defines the molecular and cellular hallmarks of progeroid syndromes according to the main cellular and animal models, and discusses the therapeutic strategies developed to date.
Collapse
Affiliation(s)
- Dido Carrero
- Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Instituto Universitario de Oncología (IUOPA), Universidad de Oviedo, Oviedo 33006, Spain
| | - Clara Soria-Valles
- Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Instituto Universitario de Oncología (IUOPA), Universidad de Oviedo, Oviedo 33006, Spain
| | - Carlos López-Otín
- Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Instituto Universitario de Oncología (IUOPA), Universidad de Oviedo, Oviedo 33006, Spain
| |
Collapse
|
11
|
Kiernan J, Davies JE, Stanford WL. Concise Review: Musculoskeletal Stem Cells to Treat Age-Related Osteoporosis. Stem Cells Transl Med 2017; 6:1930-1939. [PMID: 28834263 PMCID: PMC6430063 DOI: 10.1002/sctm.17-0054] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Accepted: 06/14/2017] [Indexed: 01/03/2023] Open
Abstract
Age‐related (type‐II) osteoporosis is a common and debilitating condition driven in part by the loss of bone marrow (BM) mesenchymal stromal cells (MSC) and their osteoblast progeny, leading to reduced bone formation. Current pharmacological regiments targeting age‐related osteoporosis do not directly treat the disease by increasing bone formation, but instead use bisphosphonates to reduce bone resorption—a treatment designed for postmenopausal (type‐I) osteoporosis. Recently, the bone regenerative capacity of MSCs has been found within a very rare population of skeletal stem cells (SSCs) residing within the larger heterogeneous BM‐MSC pool. The osteoregenerative potential of SSCs would be an ideal candidate for cell‐based therapies to treat degenerative bone diseases such as osteoporosis. However, to date, clinical and translational studies attempting to improve bone formation through cell transplantation have used the larger, nonspecific, MSC pool. In this review, we will outline the physiological basis of age‐related osteoporosis, as well as discuss relevant preclinical studies that use exogenous MSC transplantation with the aim of treating osteoporosis in murine models. We will also discuss results from specific clinical trials aimed at treating other systemic bone diseases, and how the discovery of SSC could help realize the full regenerative potential of MSC therapy to increase bone formation. Finally, we will outline how ancillary clinical trials could be initiated to assess MSC/SSC‐mediated bone formation gains in existing and potentially unrelated clinical trials, setting the stage for a dedicated clinical investigation to treat age‐related osteoporosis. Stem Cells Translational Medicine2017;6:1930–1939
Collapse
Affiliation(s)
- Jeffrey Kiernan
- Laboratory Medicine Program, University Health Network, Toronto, Ontario, Canada.,Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
| | - John E Davies
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada.,Faculty of Dentistry, University of Toronto, Toronto, Ontario, Canada
| | - William L Stanford
- Sprott Centre for Stem Cell Research, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada.,Departments of Cellular & Molecular Medicine, and Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
12
|
Jin HJ, Lee HJ, Heo J, Lim J, Kim M, Kim MK, Nam HY, Hong GH, Cho YS, Choi SJ, Kim IG, Shin DM, Kim SW. Senescence-Associated MCP-1 Secretion Is Dependent on a Decline in BMI1 in Human Mesenchymal Stromal Cells. Antioxid Redox Signal 2016; 24:471-85. [PMID: 26573462 PMCID: PMC4800271 DOI: 10.1089/ars.2015.6359] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
AIMS Cellular senescence and its secretory phenotype (senescence-associated secretory phenotype [SASP]) develop after long-term expansion of mesenchymal stromal cells (MSCs). Further investigation of this phenotype is required to improve the therapeutic efficacy of MSC-based cell therapies. In this study, we show that positive feedback between SASP and inherent senescence processes plays a crucial role in the senescence of umbilical cord blood-derived MSCs (UCB-MSCs). RESULTS We found that monocyte chemoattractant protein-1 (MCP-1) was secreted as a dominant component of the SASP during expansion of UCB-MSCs and reinforced senescence via its cognate receptor chemokine (c-c motif) receptor 2 (CCR2) by activating the ROS-p38-MAPK-p53/p21 signaling cascade in both an autocrine and paracrine manner. The activated p53 in turn increased MCP-1 secretion, completing a feed-forward loop that triggered the senescence program in UCB-MSCs. Accordingly, knockdown of CCR2 in UCB-MSCs significantly improved their therapeutic ability to alleviate airway inflammation in an experimental allergic asthma model. Moreover, BMI1, a polycomb protein, repressed the expression of MCP-1 by binding to its regulatory elements. The reduction in BMI1 levels during UCB-MSC senescence altered the epigenetic status of MCP-1, including the loss of H2AK119Ub, and resulted in derepression of MCP-1. INNOVATION Our results provide the first evidence supporting the existence of the SASP as a causative contributor to UCB-MSC senescence and reveal a so far unappreciated link between epigenetic regulation and SASP for maintaining a stable senescent phenotype. CONCLUSION Senescence of UCB-MSCs is orchestrated by MCP-1, which is secreted as a major component of the SASP and is epigenetically regulated by BMI1.
Collapse
Affiliation(s)
- Hye Jin Jin
- 1 Department of Biochemistry and Molecular Biology, Asan Medical Center, University of Ulsan College of Medicine , Seoul, Korea.,2 Biomedical Research Institute , MEDIPOST Co., Ltd., Seongnam, Korea
| | - Hyang Ju Lee
- 1 Department of Biochemistry and Molecular Biology, Asan Medical Center, University of Ulsan College of Medicine , Seoul, Korea
| | - Jinbeom Heo
- 3 Department of Biomedical Sciences, Asan Medical Center, University of Ulsan College of Medicine , Seoul, Korea.,4 Department of Physiology, Asan Medical Center, University of Ulsan College of Medicine , Seoul, Korea
| | - Jisun Lim
- 3 Department of Biomedical Sciences, Asan Medical Center, University of Ulsan College of Medicine , Seoul, Korea.,4 Department of Physiology, Asan Medical Center, University of Ulsan College of Medicine , Seoul, Korea.,5 Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine , Seoul, Korea
| | - Miyeon Kim
- 2 Biomedical Research Institute , MEDIPOST Co., Ltd., Seongnam, Korea
| | - Min Kyung Kim
- 1 Department of Biochemistry and Molecular Biology, Asan Medical Center, University of Ulsan College of Medicine , Seoul, Korea
| | - Hae Yun Nam
- 1 Department of Biochemistry and Molecular Biology, Asan Medical Center, University of Ulsan College of Medicine , Seoul, Korea
| | - Gyong Hwa Hong
- 6 Graduate School of Medical Science and Engineering, Biomedical Research Center, KAIST Institute for the BioCentury, Korea Advanced Institute of Science and Technology , Daejeon, Korea
| | - You Sook Cho
- 7 Division of Allergy and Clinical Immunology, Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine , Seoul, Korea
| | - Soo Jin Choi
- 2 Biomedical Research Institute , MEDIPOST Co., Ltd., Seongnam, Korea
| | - In-Gyu Kim
- 5 Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine , Seoul, Korea
| | - Dong-Myung Shin
- 3 Department of Biomedical Sciences, Asan Medical Center, University of Ulsan College of Medicine , Seoul, Korea.,4 Department of Physiology, Asan Medical Center, University of Ulsan College of Medicine , Seoul, Korea
| | - Seong Who Kim
- 1 Department of Biochemistry and Molecular Biology, Asan Medical Center, University of Ulsan College of Medicine , Seoul, Korea
| |
Collapse
|
13
|
Pan H, Guan D, Liu X, Li J, Wang L, Wu J, Zhou J, Zhang W, Ren R, Zhang W, Li Y, Yang J, Hao Y, Yuan T, Yuan G, Wang H, Ju Z, Mao Z, Li J, Qu J, Tang F, Liu GH. SIRT6 safeguards human mesenchymal stem cells from oxidative stress by coactivating NRF2. Cell Res 2016; 26:190-205. [PMID: 26768768 PMCID: PMC4746611 DOI: 10.1038/cr.2016.4] [Citation(s) in RCA: 243] [Impact Index Per Article: 30.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Revised: 10/15/2015] [Accepted: 11/23/2015] [Indexed: 02/06/2023] Open
Abstract
SIRT6 belongs to the mammalian homologs of Sir2 histone NAD(+)-dependent deacylase family. In rodents, SIRT6 deficiency leads to aging-associated degeneration of mesodermal tissues. It remains unknown whether human SIRT6 has a direct role in maintaining the homeostasis of mesodermal tissues. To this end, we generated SIRT6 knockout human mesenchymal stem cells (hMSCs) by targeted gene editing. SIRT6-deficient hMSCs exhibited accelerated functional decay, a feature distinct from typical premature cellular senescence. Rather than compromised chromosomal stability, SIRT6-null hMSCs were predominately characterized by dysregulated redox metabolism and increased sensitivity to the oxidative stress. In addition, we found SIRT6 in a protein complex with both nuclear factor erythroid 2-related factor 2 (NRF2) and RNA polymerase II, which was required for the transactivation of NRF2-regulated antioxidant genes, including heme oxygenase 1 (HO-1). Overexpression of HO-1 in SIRT6-null hMSCs rescued premature cellular attrition. Our study uncovers a novel function of SIRT6 in maintaining hMSC homeostasis by serving as a NRF2 coactivator, which represents a new layer of regulation of oxidative stress-associated stem cell decay.
Collapse
Affiliation(s)
- Huize Pan
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Di Guan
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Xiaomeng Liu
- Biodynamic Optical Imaging Center, College of Life Sciences, Peking University, Beijing 100871, China
| | - Jingyi Li
- Biodynamic Optical Imaging Center, College of Life Sciences, Peking University, Beijing 100871, China
| | - Lixia Wang
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
- FSU-CAS Innovation Institute, Foshan University, Foshan, Guangdong 528000, China
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Jun Wu
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Junzhi Zhou
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Weizhou Zhang
- Department of Pathology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Ruotong Ren
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
- FSU-CAS Innovation Institute, Foshan University, Foshan, Guangdong 528000, China
| | - Weiqi Zhang
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
- FSU-CAS Innovation Institute, Foshan University, Foshan, Guangdong 528000, China
| | - Ying Li
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jiping Yang
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Ying Hao
- FSU-CAS Innovation Institute, Foshan University, Foshan, Guangdong 528000, China
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Tingting Yuan
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Guohong Yuan
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Hu Wang
- Institute of Aging Research, Leibniz Link Partner Group on Stem Cell Aging, Hangzhou Normal University School of Medicine, Hangzhou, Zhejiang 310036, China
| | - Zhenyu Ju
- Institute of Aging Research, Leibniz Link Partner Group on Stem Cell Aging, Hangzhou Normal University School of Medicine, Hangzhou, Zhejiang 310036, China
| | - Zhiyong Mao
- School of life sciences and technology, Tongji University, Shanghai 200092, China
| | - Jian Li
- The Key Laboratory of Geriatrics, Beijing Hospital & Beijing Institute of Geriatrics, Ministry of Health, Beijing 100730, China
| | - Jing Qu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Fuchou Tang
- Biodynamic Optical Imaging Center, College of Life Sciences, Peking University, Beijing 100871, China
- Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, Beijing 100871, China
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China
- Center for Molecular and Translational Medicine, CMTM, Beijing 100101, China
| | - Guang-Hui Liu
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
- FSU-CAS Innovation Institute, Foshan University, Foshan, Guangdong 528000, China
- Center for Molecular and Translational Medicine, CMTM, Beijing 100101, China
- Beijing Institute for Brain Disorders, Beijing 100069, China
| |
Collapse
|
14
|
Karnaukhov AV, Karnaukhova EV, Sergievich LA, Karnaukhova NA, Bogdanenko EV, Manokhina IA, Karnaukhov VN. Informational Theory of Aging: The Life Extension Method Based on the Bone Marrow Transplantation. JOURNAL OF BIOPHYSICS (HINDAWI PUBLISHING CORPORATION : ONLINE) 2015; 2015:686249. [PMID: 26491435 PMCID: PMC4605449 DOI: 10.1155/2015/686249] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/30/2015] [Revised: 08/10/2015] [Accepted: 08/12/2015] [Indexed: 12/25/2022]
Abstract
The method of lifespan extension that is a practical application of the informational theory of aging is proposed. In this theory, the degradation (error accumulation) of the genetic information in cells is considered a main cause of aging. According to it, our method is based on the transplantation of genetically identical (or similar) stem cells with the lower number of genomic errors to the old recipients. For humans and large mammals, this method can be realized by cryopreservation of their own stem cells, taken in a young age, for the later autologous transplantation in old age. To test this method experimentally, we chose laboratory animals of relatively short lifespan (mouse). Because it is difficult to isolate the required amount of the stem cells (e.g., bone marrow) without significant damage for animals, we used the bone marrow transplantation from sacrificed inbred young donors. It is shown that the lifespan extension of recipients depends on level of their genetic similarity (syngeneity) with donors. We have achieved the lifespan increase of the experimental mice by 34% when the transplantation of the bone marrow with high level of genetic similarity was used.
Collapse
Affiliation(s)
- Alexey V. Karnaukhov
- Institute of Cell Biophysics, Russian Academy of Sciences, Institutskaya Street 3, Pushchino, Moscow 142290, Russia
| | - Elena V. Karnaukhova
- Institute of Cell Biophysics, Russian Academy of Sciences, Institutskaya Street 3, Pushchino, Moscow 142290, Russia
| | - Larisa A. Sergievich
- Institute of Cell Biophysics, Russian Academy of Sciences, Institutskaya Street 3, Pushchino, Moscow 142290, Russia
| | - Natalia A. Karnaukhova
- Institute of Cell Biophysics, Russian Academy of Sciences, Institutskaya Street 3, Pushchino, Moscow 142290, Russia
| | - Elena V. Bogdanenko
- Institute of General Pathology and Pathophysiology, Russian Academy of Medical Sciences, Baltiyskaya Street 8, Moscow 125315, Russia
| | - Irina A. Manokhina
- Institute of Cell Biophysics, Russian Academy of Sciences, Institutskaya Street 3, Pushchino, Moscow 142290, Russia
| | - Valery N. Karnaukhov
- Institute of Cell Biophysics, Russian Academy of Sciences, Institutskaya Street 3, Pushchino, Moscow 142290, Russia
| |
Collapse
|
15
|
|
16
|
Liu H, Xia X, Li B. Mesenchymal stem cell aging: Mechanisms and influences on skeletal and non-skeletal tissues. Exp Biol Med (Maywood) 2015; 240:1099-106. [PMID: 26088863 DOI: 10.1177/1535370215591828] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The aging population and the incidence of aging-related diseases such as osteoporosis are on the rise. Aging at the tissue and organ levels usually involves tissue stem cells. Human and animal model studies indicate that aging affects two aspects of mesenchymal stem cell (MSC): a decrease in the bone marrow MSC pool and biased differentiation into adipocyte at the cost of osteoblast, which underlie the etiology of osteoporosis. Aging of MSC cells is also detrimental to some non-skeletal tissues, in particular the hematopoietic system, where MSCs serve as a niche component. In addition, aging compromises the therapeutic potentials of MSC cells, including cells isolated from aged individuals or cells cultured for many passages. Here we discuss the recent progress on our understanding of MSC aging, with a focus on the effects of MSC aging on bone remodeling and hematopoiesis and the mechanisms of MSC aging.
Collapse
Affiliation(s)
- Huijuan Liu
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Xuechun Xia
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Baojie Li
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai 200240, China
| |
Collapse
|
17
|
Karnaukhov AV, Karnaukhova EV, Sergievich LA, Karnaukhova NA, Bogdanenko EV, Smirnov AA, Manokhina IA, Karnaukhov VN. Information theory of ageing: Studying the effect of bone marrow transplantation on the life span of mice. Biophysics (Nagoya-shi) 2014. [DOI: 10.1134/s0006350914040137] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
|
18
|
Brennan TA, Egan KP, Lindborg CM, Chen Q, Sweetwyne MT, Hankenson KD, Xie SX, Johnson FB, Pignolo RJ. Mouse models of telomere dysfunction phenocopy skeletal changes found in human age-related osteoporosis. Dis Model Mech 2014; 7:583-92. [PMID: 24626990 PMCID: PMC4007409 DOI: 10.1242/dmm.014928] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
A major medical challenge in the elderly is osteoporosis and the high risk of fracture. Telomere dysfunction is a cause of cellular senescence and telomere shortening, which occurs with age in cells from most human tissues, including bone. Telomere defects contribute to the pathogenesis of two progeroid disorders characterized by premature osteoporosis, Werner syndrome and dyskeratosis congenital. It is hypothesized that telomere shortening contributes to bone aging. We evaluated the skeletal phenotypes of mice with disrupted telomere maintenance mechanisms as models for human bone aging, including mutants in Werner helicase (Wrn−/−), telomerase (Terc−/−) and Wrn−/−Terc−/− double mutants. Compared with young wild-type (WT) mice, micro-computerized tomography analysis revealed that young Terc−/− and Wrn−/−Terc−/− mice have decreased trabecular bone volume, trabecular number and trabecular thickness, as well as increased trabecular spacing. In cortical bone, young Terc−/− and Wrn−/−Terc−/− mice have increased cortical thinning, and increased porosity relative to age-matched WT mice. These trabecular and cortical changes were accelerated with age in Terc−/− and Wrn−/−Terc−/− mice compared with older WT mice. Histological quantification of osteoblasts in aged mice showed a similar number of osteoblasts in all genotypes; however, significant decreases in osteoid, mineralization surface, mineral apposition rate and bone formation rate in older Terc−/− and Wrn−/−Terc−/− bone suggest that osteoblast dysfunction is a prominent feature of precocious aging in these mice. Except in the Wrn−/− single mutant, osteoclast number did not increase in any genotype. Significant alterations in mechanical parameters (structure model index, degree of anistrophy and moment of inertia) of the Terc−/− and Wrn−/−Terc−/− femurs compared with WT mice were also observed. Young Wrn−/−Terc−/− mice had a statistically significant increase in bone-marrow fat content compared with young WT mice, which remained elevated in aged double mutants. Taken together, our results suggest that Terc−/− and Wrn−/−Terc−/− mutants recapitulate the human bone aging phenotype and are useful models for studying age-related osteoporosis.
Collapse
Affiliation(s)
- Tracy A Brennan
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
19
|
McClelland Descalzo DL, Ehnes DD, zur Nieden NI. Stem cells for osteodegenerative diseases: current studies and future outlook. Regen Med 2014; 9:219-30. [DOI: 10.2217/rme.13.100] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
As the worldwide population grows and life expectancies continue to increase, degenerative diseases of the bones, muscles, and connective tissue are a growing problem for society. Current therapies for osteodegenerative disorders such as hormone replacement therapies, calcium/vitamin D supplements and oral bisphosphonates are often inadequate to stop degeneration and/or have serious negative side effects. Thus, there is an urgent need in the medical community for more effective and safer treatments. Stem cell therapies for osteodegenerative disorders have been rigorously explored over the last decade and are yielding some promising results in animal models and clinical trials. Although much work still needs to be done to ensure the safety and efficacy of these therapies, stem cells represent a new frontier of exciting possibilities for bone and cartilage regeneration.
Collapse
Affiliation(s)
- Darcie L McClelland Descalzo
- Department of Cell Biology & Neuroscience, 1113 Biological Sciences Building, University of California Riverside, Riverside, CA 92521, USA
| | - Devon D Ehnes
- Department of Cell Biology & Neuroscience, 1113 Biological Sciences Building, University of California Riverside, Riverside, CA 92521, USA
| | - Nicole I zur Nieden
- Department of Cell Biology & Neuroscience, 1113 Biological Sciences Building, University of California Riverside, Riverside, CA 92521, USA
| |
Collapse
|
20
|
Bidwell JP, Alvarez MB, Hood M, Childress P. Functional impairment of bone formation in the pathogenesis of osteoporosis: the bone marrow regenerative competence. Curr Osteoporos Rep 2013; 11:117-25. [PMID: 23471774 DOI: 10.1007/s11914-013-0139-2] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The skeleton is a high-renewal organ that undergoes ongoing cycles of remodeling. The regenerative bone formation arm ultimately declines in the aging, postmenopausal skeleton, but current therapies do not adequately address this deficit. Bone marrow is the primary source of the skeletal anabolic response and the mesenchymal stem cells (MSCs), which give rise to bone matrix-producing osteoblasts. The identity of these stem cells is emerging, but it now appears that the term 'MSC' has often been misapplied to the bone marrow stromal cell (BMSC), a progeny of the MSC. Nevertheless, the changes in BMSC phenotype associated with age and estrogen depletion likely contribute to the attenuated regenerative competence of the marrow and may reflect alterations in MSC phenotype. Here we summarize current concepts in bone marrow MSC identity, and within this context, review recent observations on changes in bone marrow population dynamics associated with aging and menopause.
Collapse
Affiliation(s)
- Joseph P Bidwell
- Department of Anatomy and Cell Biology, Indiana University School of Medicine (IUSM), Medical Science Bldg 5035, 635 Barnhill Drive, Indianapolis, IN 46202, USA.
| | | | | | | |
Collapse
|
21
|
Hofmann TJ, Otsuru S, Marino R, Rasini V, Veronesi E, Murgia A, Lahti J, Boyd K, Dominici M, Horwitz EM. Transplanted murine long-term repopulating hematopoietic cells can differentiate to osteoblasts in the marrow stem cell niche. Mol Ther 2013; 21:1224-31. [PMID: 23587920 DOI: 10.1038/mt.2013.36] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Bone marrow transplantation (BMT) can give rise to donor-derived osteopoiesis in mice and humans; however, the source of this activity, whether a primitive osteoprogenitor or a transplantable marrow cell with dual hematopoietic and osteogenic potential, has eluded detection. To address this issue, we fractionated whole BM from mice according to cell surface immunophenotype and assayed the hematopoietic and osteopoietic potentials of the transplanted cells. Here, we show that a donor marrow cell capable of robust osteopoiesis possesses a surface phenotype of c-Kit(+) Lin(-) Sca-1(+) CD34(-/lo), identical to that of the long-term repopulating hematopoietic stem cell (LTR-HSC). Secondary BMT studies demonstrated that a single marrow cell able to contribute to hematopoietic reconstitution in primary recipients also drives robust osteopoiesis and LT hematopoiesis in secondary recipients. These findings indicate that LTR-HSC can give rise to progeny that differentiate to osteoblasts after BMT, suggesting a mechanism for prompt restoration of the osteoblastic HSC niche following BM injury, such as that induced by clinical BMT preparative regimens. An understanding of the mechanisms that regulate this differentiation potential may lead to novel treatments for disorders of bone as well as methods for preserving the integrity of endosteal hematopoietic niches.
Collapse
Affiliation(s)
- Ted J Hofmann
- Division of Oncology/Blood and Marrow Transplantation, The Children's Hospital of Philadelphia and The University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|