1
|
Werner H, Laron Z. Insulin-like growth factors and aging: lessons from Laron syndrome. Front Endocrinol (Lausanne) 2023; 14:1291812. [PMID: 37941907 PMCID: PMC10628706 DOI: 10.3389/fendo.2023.1291812] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Accepted: 10/10/2023] [Indexed: 11/10/2023] Open
Abstract
The growth hormone (GH)-insulin-like growth factor-1 (IGF1) signaling pathway emerged in recent years as a key determinant of aging and longevity. Disruption of this network in different animal species, including flies, nematodes and mouse, was consistently associated with an extended lifespan. Epidemiological analyses have shown that patients with Laron syndrome (LS), the best-characterized disease under the umbrella of the congenital IGF1 deficiencies, seem to be protected from cancer. While aging and cancer, as a rule, are considered diametrically opposite processes, modern lines of evidence reinforce the notion that aging and cancer might, as a matter of fact, be regarded as divergent manifestations of identical biochemical and cellular underlying processes. While the effect of individual mutations on lifespan and health span is very difficult to assess, genome-wide screenings identified a number of differentially represented aging- and longevity-associated genes in patients with LS. The present review summarizes recent data that emerged from comprehensive analyses of LS patients and portrays a number of previously unrecognized targets for GH-IGF1 action. Our article sheds light on complex aging and longevity processes, with a particular emphasis on the role of the GH-IGF1 network in these mechanisms.
Collapse
Affiliation(s)
- Haim Werner
- Department of Human Molecular Genetics and Biochemistry, School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Zvi Laron
- Endocrinology and Diabetes Research Unit, Schneider Children’s Medical Center, Petah Tikva, Israel
| |
Collapse
|
2
|
Galow AM, Brenmoehl J, Hoeflich A. Synergistic effects of hormones on structural and functional maturation of cardiomyocytes and implications for heart regeneration. Cell Mol Life Sci 2023; 80:240. [PMID: 37541969 PMCID: PMC10403476 DOI: 10.1007/s00018-023-04894-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 07/18/2023] [Accepted: 07/22/2023] [Indexed: 08/06/2023]
Abstract
The limited endogenous regenerative capacity of the human heart renders cardiovascular diseases a major health threat, thus motivating intense research on in vitro heart cell generation and cell replacement therapies. However, so far, in vitro-generated cardiomyocytes share a rather fetal phenotype, limiting their utility for drug testing and cell-based heart repair. Various strategies to foster cellular maturation provide some success, but fully matured cardiomyocytes are still to be achieved. Today, several hormones are recognized for their effects on cardiomyocyte proliferation, differentiation, and function. Here, we will discuss how the endocrine system impacts cardiomyocyte maturation. After detailing which features characterize a mature phenotype, we will contemplate hormones most promising to induce such a phenotype, the routes of their action, and experimental evidence for their significance in this process. Due to their pleiotropic effects, hormones might be not only valuable to improve in vitro heart cell generation but also beneficial for in vivo heart regeneration. Accordingly, we will also contemplate how the presented hormones might be exploited for hormone-based regenerative therapies.
Collapse
Affiliation(s)
- Anne-Marie Galow
- Institute of Genome Biology, Research Institute for Farm Animal Biology (FBN), 18196, Dummerstorf, Germany.
| | - Julia Brenmoehl
- Institute of Genome Biology, Research Institute for Farm Animal Biology (FBN), 18196, Dummerstorf, Germany
| | - Andreas Hoeflich
- Institute of Genome Biology, Research Institute for Farm Animal Biology (FBN), 18196, Dummerstorf, Germany
| |
Collapse
|
3
|
Murphy A, Vyavahare S, Kumar S, Lee TJ, Sharma A, Adusumilli S, Hamrick M, Isales CM, Fulzele S. Dietary interventions and molecular mechanisms for healthy musculoskeletal aging. Biogerontology 2022; 23:681-698. [PMID: 35727468 DOI: 10.1007/s10522-022-09970-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 05/18/2022] [Indexed: 12/13/2022]
Abstract
Over the past decade, extensive efforts have focused on understanding age-associated diseases and how to prolong a healthy lifespan. The induction of dietary protocols such as caloric restriction (CR) and protein restriction (PR) has positively affected a healthy lifespan. These intervention ideas (nutritional protocols) have been the subject of human cohort studies and clinical trials to evaluate their effectiveness in alleviating age-related diseases (such as type II diabetes, cardiovascular disease, obesity, and musculoskeletal fragility) and promoting human longevity. This study summarizes the literature on the nutritional protocols, emphasizing their impacts on bone and muscle biology. In addition, we analyzed several CR studies using Gene Expression Omnibus (GEO) database and identified common transcriptome changes to understand the signaling pathway involved in musculoskeletal tissue. We identified nine novel common genes, out of which five were upregulated (Emc3, Fam134b, Fbxo30, Pip5k1a, and Retsat), and four were downregulated (Gstm2, Per2, Fam78a, and Sel1l3) with CR in muscles. Gene Ontology enrichment analysis revealed that CR regulates several signaling pathways (e.g., circadian gene regulation and rhythm, energy reserve metabolic process, thermogenesis) involved in energy metabolism. In conclusion, this study summarizes the beneficiary role of CR and identifies novel genes and signaling pathways involved in musculoskeletal biology.
Collapse
Affiliation(s)
- Andrew Murphy
- Department of Medicine, Augusta University, Augusta, GA, 30912, USA
| | - Sagar Vyavahare
- Department of Cell Biology and Anatomy, Augusta University, Augusta, GA, USA
| | - Sandeep Kumar
- Department of Cell Biology and Anatomy, Augusta University, Augusta, GA, USA
| | - Tae Jin Lee
- Center for Biotechnology and Genomic Medicine, Augusta University, Augusta, GA, 30912, USA
| | - Ashok Sharma
- Center for Biotechnology and Genomic Medicine, Augusta University, Augusta, GA, 30912, USA
| | | | - Mark Hamrick
- Department of Cell Biology and Anatomy, Augusta University, Augusta, GA, USA.,Center for Healthy Aging, Augusta University, Augusta, GA, USA
| | - Carlos M Isales
- Department of Medicine, Augusta University, Augusta, GA, 30912, USA.,Center for Healthy Aging, Augusta University, Augusta, GA, USA
| | - Sadanand Fulzele
- Department of Medicine, Augusta University, Augusta, GA, 30912, USA. .,Department of Cell Biology and Anatomy, Augusta University, Augusta, GA, USA. .,Center for Healthy Aging, Augusta University, Augusta, GA, USA.
| |
Collapse
|
4
|
Zakirova AN, Zakirova NE, Nizamova DF. Insulin-like Growth Factor-1 and Myocardial Remodeling in Patients with Chronic Heart Failure of Ischemic Origin. RATIONAL PHARMACOTHERAPY IN CARDIOLOGY 2022. [DOI: 10.20996/1819-6446-2022-10-01] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Aim. To study the presence and nature of correlations between the level of Insulin-like growth factor-1 (IGF-1) and structural and functional parameters of the heart in the development of myocardial remodeling and fibrosis in patients with chronic heart failure (CHF) of ischemic origin.Material and methods. The study included 120 men with class II-IV CHF who have history of myocardial infarction, which are divided into 3 groups depending on the CHF class. The control group included 25 healthy men. Assessment of left ventricular (LV) structural-functional state was carried out by echocardiography. Investigation of IGF-1 and N-terminal precursor indices of cerebral natriuretic peptide (NT-pro BNP) was performed by enzyme immunoassay.Results. Patients with class II CHF were hyperexpression of IGF-1, with class III CHF were registered low-normal level, with class IV CHF was established a deficiency of IGF-1. The most significant structural-geometric rearrangement of LV and significant deficit of IGF-1 recorded in patients with class IV CHF (95,6±7,02 ng/ml with class IV CHF versus 178,3±11,36 ng/ml and 124,3±9,14 ng/ml with class II and III CHF; р<0,05). In patients of class III-IV CHF, correlation relationships between IGF-1 level and echocardiographic parameters (LV myocardial mass index are established: r=-0,59, p=0,05; end systolic volume index: r=-0,55, p=0,05; value of LV ejection fraction: r=0,61, p=0,05). Significant negative correlation are established in patients with class III-IV CHF between IGF-1 level and NT-pro BNP levels (r=-0,51; р=0,05).Conclusion. The intensity of myocardial remodeling and fibrosis processes in patients with a progressive course of CHF is related to deficit of IGF-1 and is associated with a high level of activity of natriuretic peptides.
Collapse
|
5
|
Yurova MN, Tyndyk ML, Popovich IG, Golubev AG, Anisimov VN. Gender Specificity of the Effect of Neonatal Melatonin Administration on Lifespan and Age-Associated Pathology in 129/Sv Mice. ADVANCES IN GERONTOLOGY 2019. [DOI: 10.1134/s2079057019030184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
6
|
Anisimov VN, Labunets IF, Popovich IG, Tyndyk ML, Yurova MN, Golubev AG. In mice transgenic for IGF1 under keratin-14 promoter, lifespan is decreased and the rates of aging and thymus involution are accelerated. Aging (Albany NY) 2019; 11:2098-2110. [PMID: 30981207 PMCID: PMC6503882 DOI: 10.18632/aging.101903] [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: 12/12/2018] [Accepted: 03/31/2019] [Indexed: 11/25/2022]
Abstract
IGF1 signaling is supposedly a key lifespan determinant in metazoans. However, controversial lifespan data were obtained with different means used to modify IGF1 or its receptor (IGF1R) expression in mice. The emerging puzzle lacks pieces of evidence needed to construct a coherent picture. We add to the available evidence by using the Gompertz model (GM), with account for the artifactual component of the Strehler-Mildvan correlation between its parameters, to compare the survival patterns of female FVB/N and FVB/N-derived K14/mIGF1 mice. In K14/mIGF1 vs. FVB/N mice, the rate of aging (γ) is markedly increased without concomitant changes in the initial mortality (μ0). In published cases where IGF1 signaling was altered by modifying liver or muscle IGF1 or whole body IGF1R expression, lifespan changes are attributable to μ0. The accelerated aging and associated tumor yield in K14/mIGF1 mice are consistent with the finding that the age-associated decreases in thymus weight and serum thymulin are accelerated in K14/mIGF1 mice. Our results underscore the importance of accounting for the mathematical artifacts of data fitting to GM in attempts to resolve discrepancies in survival data and to differentiate the contributions of the initial mortality and the rate of aging to changes in lifespan.
Collapse
Affiliation(s)
- Vladimir N Anisimov
- Department of Carcinogenesis and Oncogerontology, N.N. Petrov National Medical Research Center of Oncology, Saint Petersburg 197758, Russia
| | - Irina F Labunets
- Laboratory of Experimental Models, State Institute of Genetic and Regenerative Medicine, National Academy of Medical Sciences of Ukraine, Kiev 04114, Ukraine
| | - Irina G Popovich
- Department of Carcinogenesis and Oncogerontology, N.N. Petrov National Medical Research Center of Oncology, Saint Petersburg 197758, Russia
| | - Margarita L Tyndyk
- Department of Carcinogenesis and Oncogerontology, N.N. Petrov National Medical Research Center of Oncology, Saint Petersburg 197758, Russia
| | - Maria N Yurova
- Department of Carcinogenesis and Oncogerontology, N.N. Petrov National Medical Research Center of Oncology, Saint Petersburg 197758, Russia
| | - Alexey G Golubev
- Department of Carcinogenesis and Oncogerontology, N.N. Petrov National Medical Research Center of Oncology, Saint Petersburg 197758, Russia
| |
Collapse
|
7
|
Evans LW, Ferguson BS. Food Bioactive HDAC Inhibitors in the Epigenetic Regulation of Heart Failure. Nutrients 2018; 10:E1120. [PMID: 30126190 PMCID: PMC6115944 DOI: 10.3390/nu10081120] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 08/15/2018] [Accepted: 08/15/2018] [Indexed: 12/21/2022] Open
Abstract
Approximately 5.7 million U.S. adults have been diagnosed with heart failure (HF). More concerning is that one in nine U.S. deaths included HF as a contributing cause. Current HF drugs (e.g., β-blockers, ACEi) target intracellular signaling cascades downstream of cell surface receptors to prevent cardiac pump dysfunction. However, these drugs fail to target other redundant intracellular signaling pathways and, therefore, limit drug efficacy. As such, it has been postulated that compounds designed to target shared downstream mediators of these signaling pathways would be more efficacious for the treatment of HF. Histone deacetylation has been linked as a key pathogenetic element for the development of HF. Lysine residues undergo diverse and reversible post-translational modifications that include acetylation and have historically been studied as epigenetic modifiers of histone tails within chromatin that provide an important mechanism for regulating gene expression. Of recent, bioactive compounds within our diet have been linked to the regulation of gene expression, in part, through regulation of the epi-genome. It has been reported that food bioactives regulate histone acetylation via direct regulation of writer (histone acetyl transferases, HATs) and eraser (histone deacetylases, HDACs) proteins. Therefore, bioactive food compounds offer unique therapeutic strategies as epigenetic modifiers of heart failure. This review will highlight food bio-actives as modifiers of histone deacetylase activity in the heart.
Collapse
Affiliation(s)
- Levi W Evans
- Department of Agriculture, Nutrition, & Veterinary Sciences, University of Nevada, Reno, NV 89557, USA.
- Center for Cardiovascular Research, University of Nevada, Reno, NV 89557, USA.
- Environmental Science & Health, University of Nevada, Reno, NV 89557, USA.
| | - Bradley S Ferguson
- Department of Agriculture, Nutrition, & Veterinary Sciences, University of Nevada, Reno, NV 89557, USA.
- Center for Cardiovascular Research, University of Nevada, Reno, NV 89557, USA.
| |
Collapse
|
8
|
Angelini F, Pagano F, Bordin A, Picchio V, De Falco E, Chimenti I. Getting Old through the Blood: Circulating Molecules in Aging and Senescence of Cardiovascular Regenerative Cells. Front Cardiovasc Med 2017; 4:62. [PMID: 29057227 PMCID: PMC5635266 DOI: 10.3389/fcvm.2017.00062] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Accepted: 09/19/2017] [Indexed: 12/11/2022] Open
Abstract
Global aging is a hallmark of our century. The natural multifactorial process resulting in aging involves structural and functional changes, affecting molecules, cells, and tissues. As the western population is getting older, we are witnessing an increase in the burden of cardiovascular events, some of which are known to be directly linked to cellular senescence and dysfunction. In this review, we will focus on the description of a few circulating molecules, which have been correlated to life span, aging, and cardiovascular homeostasis. We will review the current literature concerning the circulating levels and related signaling pathways of selected proteins (insulin-like growth factor 1, growth and differentiation factor-11, and PAI-1) and microRNAs of interest (miR-34a, miR-146a, miR-21), whose bloodstream levels have been associated to aging in different organisms. In particular, we will also discuss their potential role in the biology and senescence of cardiovascular regenerative cell types, such as endothelial progenitor cells, mesenchymal stromal cells, and cardiac progenitor cells.
Collapse
Affiliation(s)
- Francesco Angelini
- Department of Medical Surgical Sciences and Biotechnologies, "La Sapienza" University of Rome, Latina, Italy
| | - Francesca Pagano
- Department of Medical Surgical Sciences and Biotechnologies, "La Sapienza" University of Rome, Latina, Italy
| | - Antonella Bordin
- Department of Medical Surgical Sciences and Biotechnologies, "La Sapienza" University of Rome, Latina, Italy
| | - Vittorio Picchio
- Department of Medical Surgical Sciences and Biotechnologies, "La Sapienza" University of Rome, Latina, Italy
| | - Elena De Falco
- Department of Medical Surgical Sciences and Biotechnologies, "La Sapienza" University of Rome, Latina, Italy
| | - Isotta Chimenti
- Department of Medical Surgical Sciences and Biotechnologies, "La Sapienza" University of Rome, Latina, Italy
| |
Collapse
|
9
|
Ben-Avraham D, Govindaraju DR, Budagov T, Fradin D, Durda P, Liu B, Ott S, Gutman D, Sharvit L, Kaplan R, Bougnères P, Reiner A, Shuldiner AR, Cohen P, Barzilai N, Atzmon G. The GH receptor exon 3 deletion is a marker of male-specific exceptional longevity associated with increased GH sensitivity and taller stature. SCIENCE ADVANCES 2017; 3:e1602025. [PMID: 28630896 PMCID: PMC5473676 DOI: 10.1126/sciadv.1602025] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Accepted: 04/26/2017] [Indexed: 05/26/2023]
Abstract
Although both growth hormone (GH) and insulin-like growth factor 1 (IGF-1) signaling were shown to regulate life span in lower organisms, the role of GH signaling in human longevity remains unclear. Because a GH receptor exon 3 deletion (d3-GHR) appears to modulate GH sensitivity in humans, we hypothesized that this polymorphism could play a role in human longevity. We report a linear increased prevalence of d3-GHR homozygosity with age in four independent cohorts of long-lived individuals: 841 participants [567 of the Longevity Genes Project (LGP) (8% increase; P = 0.01), 152 of the Old Order Amish (16% increase; P = 0.02), 61 of the Cardiovascular Health Study (14.2% increase; P = 0.14), and 61 of the French Long-Lived Study (23.5% increase; P = 0.02)]. In addition, mega analysis of males in all cohorts resulted in a significant positive trend with age (26% increase; P = 0.007), suggesting sexual dimorphism for GH action in longevity. Further, on average, LGP d3/d3 homozygotes were 1 inch taller than the wild-type (WT) allele carriers (P = 0.05) and also showed lower serum IGF-1 levels (P = 0.003). Multivariate regression analysis indicated that the presence of d3/d3 genotype adds approximately 10 years to life span. The LGP d3/d3-GHR transformed lymphocytes exhibited superior growth and extracellular signal-regulated kinase activation, to GH treatment relative to WT GHR lymphocytes (P < 0.01), indicating a GH dose response. The d3-GHR variant is a common genetic polymorphism that modulates GH responsiveness throughout the life span and positively affects male longevity.
Collapse
Affiliation(s)
- Danny Ben-Avraham
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Institute of Aging Research, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Diddahally R. Govindaraju
- Institute of Aging Research, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Department of Human Evolutionary Biology, Harvard University, Cambridge, MA 02138, USA
| | - Temuri Budagov
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Institute of Aging Research, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Delphine Fradin
- INSERM U986, Pincus Building, Bicêtre Hospital, Paris Sud University, 94275 Le Kremlin Bicêtre, France
| | - Peter Durda
- Department of Pathology University of Vermont, 208 South Park Drive, Colchester, VT 05446, USA
| | - Bing Liu
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, USA
| | - Sandy Ott
- University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Danielle Gutman
- Department of Human Biology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel
| | - Lital Sharvit
- Department of Human Biology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel
| | - Robert Kaplan
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Pierre Bougnères
- INSERM U986, Pincus Building, Bicêtre Hospital, Paris Sud University, 94275 Le Kremlin Bicêtre, France
- INSERM U1169 and Department of Pediatric Endocrinology and Diabetes, Bicêtre Hospital, Pôle I3E, Paris Sud University, 94275 Le Kremlin Bicêtre, France
| | - Alex Reiner
- Department of Epidemiology, University of Washington School of Public Health, Seattle, WA 98195, USA
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Alan R. Shuldiner
- University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Geriatrics Research and Education Clinical Center, Veterans Administration Medical Center, Baltimore, MD 20420, USA
| | - Pinchas Cohen
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, USA
| | - Nir Barzilai
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Institute of Aging Research, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Gil Atzmon
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Institute of Aging Research, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Department of Human Biology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel
| |
Collapse
|
10
|
Shirakabe A, Ikeda Y, Sciarretta S, Zablocki DK, Sadoshima J. Aging and Autophagy in the Heart. Circ Res 2016; 118:1563-76. [PMID: 27174950 PMCID: PMC4869999 DOI: 10.1161/circresaha.116.307474] [Citation(s) in RCA: 316] [Impact Index Per Article: 35.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Accepted: 02/15/2016] [Indexed: 12/15/2022]
Abstract
The aging population is increasing in developed countries. Because the incidence of cardiac disease increases dramatically with age, it is important to understand the molecular mechanisms through which the heart becomes either more or less susceptible to stress. Cardiac aging is characterized by the presence of hypertrophy, fibrosis, and accumulation of misfolded proteins and dysfunctional mitochondria. Macroautophagy (hereafter referred to as autophagy) is a lysosome-dependent bulk degradation mechanism that is essential for intracellular protein and organelle quality control. Autophagy and autophagic flux are generally decreased in aging hearts, and murine autophagy loss-of-function models develop exacerbated cardiac dysfunction that is accompanied by the accumulation of misfolded proteins and dysfunctional organelles. On the contrary, stimulation of autophagy generally improves cardiac function in mouse models of protein aggregation by removing accumulated misfolded proteins, dysfunctional mitochondria, and damaged DNA, thereby improving the overall cellular environment and alleviating aging-associated pathology in the heart. Increasing lines of evidence suggest that autophagy is required for many mechanisms that mediate lifespan extension, such as caloric restriction, in various organisms. These results raise the exciting possibility that autophagy may play an important role in combating the adverse effects of aging in the heart. In this review, we discuss the role of autophagy in the heart during aging, how autophagy alleviates age-dependent changes in the heart, and how the level of autophagy in the aging heart can be restored.
Collapse
Affiliation(s)
- Akihiro Shirakabe
- From the Department of Cell Biology and Molecular Medicine, Rutgers-New Jersey Medical School, Newark (A.S., Y.I., S.S., D.K.Z., J.S.); Department of Cardiovascular Medicine and Hypertension, Graduate School of Medical and Dental Science, Kagoshima University, Japan (Y.I.); Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Rome, Italy (S.S.); and Department of AngioCardioNeurology, IRCCS Neuromed, Pozzilli, Italy (S.S.)
| | - Yoshiyuki Ikeda
- From the Department of Cell Biology and Molecular Medicine, Rutgers-New Jersey Medical School, Newark (A.S., Y.I., S.S., D.K.Z., J.S.); Department of Cardiovascular Medicine and Hypertension, Graduate School of Medical and Dental Science, Kagoshima University, Japan (Y.I.); Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Rome, Italy (S.S.); and Department of AngioCardioNeurology, IRCCS Neuromed, Pozzilli, Italy (S.S.)
| | - Sebastiano Sciarretta
- From the Department of Cell Biology and Molecular Medicine, Rutgers-New Jersey Medical School, Newark (A.S., Y.I., S.S., D.K.Z., J.S.); Department of Cardiovascular Medicine and Hypertension, Graduate School of Medical and Dental Science, Kagoshima University, Japan (Y.I.); Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Rome, Italy (S.S.); and Department of AngioCardioNeurology, IRCCS Neuromed, Pozzilli, Italy (S.S.)
| | - Daniela K Zablocki
- From the Department of Cell Biology and Molecular Medicine, Rutgers-New Jersey Medical School, Newark (A.S., Y.I., S.S., D.K.Z., J.S.); Department of Cardiovascular Medicine and Hypertension, Graduate School of Medical and Dental Science, Kagoshima University, Japan (Y.I.); Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Rome, Italy (S.S.); and Department of AngioCardioNeurology, IRCCS Neuromed, Pozzilli, Italy (S.S.)
| | - Junichi Sadoshima
- From the Department of Cell Biology and Molecular Medicine, Rutgers-New Jersey Medical School, Newark (A.S., Y.I., S.S., D.K.Z., J.S.); Department of Cardiovascular Medicine and Hypertension, Graduate School of Medical and Dental Science, Kagoshima University, Japan (Y.I.); Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Rome, Italy (S.S.); and Department of AngioCardioNeurology, IRCCS Neuromed, Pozzilli, Italy (S.S.).
| |
Collapse
|
11
|
|
12
|
Sharples AP, Hughes DC, Deane CS, Saini A, Selman C, Stewart CE. Longevity and skeletal muscle mass: the role of IGF signalling, the sirtuins, dietary restriction and protein intake. Aging Cell 2015; 14:511-23. [PMID: 25866088 PMCID: PMC4531066 DOI: 10.1111/acel.12342] [Citation(s) in RCA: 145] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/03/2015] [Indexed: 12/11/2022] Open
Abstract
Advancing age is associated with a progressive loss of skeletal muscle (SkM) mass and function. Given the worldwide aging demographics, this is a major contributor to morbidity, escalating socio-economic costs and ultimately mortality. Previously, it has been established that a decrease in regenerative capacity in addition to SkM loss with age coincides with suppression of insulin/insulin-like growth factor signalling pathways. However, genetic or pharmacological modulations of these highly conserved pathways have been observed to significantly enhance life and healthspan in various species, including mammals. This therefore provides a controversial paradigm in which reduced regenerative capacity of skeletal muscle tissue with age potentially promotes longevity of the organism. This paradox will be assessed and considered in the light of the following: (i) the genetic knockout, overexpression and pharmacological models that induce lifespan extension (e.g. IRS-1/s6K KO, mTOR inhibition) versus the important role of these signalling pathways in SkM growth and adaptation; (ii) the role of the sirtuins (SIRTs) in longevity versus their emerging role in SkM regeneration and survival under catabolic stress; (iii) the role of dietary restriction and its impact on longevity versus skeletal muscle mass regulation; (iv) the crosstalk between cellular energy metabolism (AMPK/TSC2/SIRT1) and survival (FOXO) versus growth and repair of SkM (e.g. AMPK vs. mTOR); and (v) the impact of protein feeding in combination with dietary restriction will be discussed as a potential intervention to maintain SkM mass while increasing longevity and enabling healthy aging.
Collapse
Affiliation(s)
- Adam P. Sharples
- Stem Cells, Ageing & Molecular Physiology Unit; Research Institute for Sport and Exercise Sciences (RISES); Exercise Metabolism and Adaptation Research Group (EMARG); Liverpool John Moores University; Tom Reilly Building Liverpool L3 3AF UK
| | - David C. Hughes
- Stem Cells, Ageing & Molecular Physiology Unit; Research Institute for Sport and Exercise Sciences (RISES); Exercise Metabolism and Adaptation Research Group (EMARG); Liverpool John Moores University; Tom Reilly Building Liverpool L3 3AF UK
- Department of Neurobiology, Physiology and Behavior; University of California; Davis California CA 95616 USA
| | - Colleen S. Deane
- MRC/ARUK Centre of Excellence for Musculoskeletal Ageing Research; School of Medicine; University of Nottingham; Royal Derby Hospital; Derby DE22 3DT UK
- School of Health and Social Care; Bournemouth University; Bournemouth BH12 5BB UK
| | - Amarjit Saini
- Department of Physiology and Pharmacology; Karolinska Institutet; Stockholm 171 77 Sweden
| | - Colin Selman
- Glasgow Ageing Research Network (GARNER); Institute of Biodiversity, Animal Health and Comparative Medicine; College of Medicine, Veterinary and Life Sciences; University of Glasgow; Glasgow G12 8QQ UK
| | - Claire E. Stewart
- Stem Cells, Ageing & Molecular Physiology Unit; Research Institute for Sport and Exercise Sciences (RISES); Exercise Metabolism and Adaptation Research Group (EMARG); Liverpool John Moores University; Tom Reilly Building Liverpool L3 3AF UK
| |
Collapse
|
13
|
Ren J, Anversa P. The insulin-like growth factor I system: physiological and pathophysiological implication in cardiovascular diseases associated with metabolic syndrome. Biochem Pharmacol 2014; 93:409-17. [PMID: 25541285 DOI: 10.1016/j.bcp.2014.12.006] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2014] [Revised: 12/11/2014] [Accepted: 12/11/2014] [Indexed: 12/31/2022]
Abstract
Metabolic syndrome is a cluster of risk factors including obesity, dyslipidemia, hypertension, and insulin resistance. A number of theories have been speculated for the pathogenesis of metabolic syndrome including impaired glucose and lipid metabolism, lipotoxicity, oxidative stress, interrupted neurohormonal regulation and compromised intracellular Ca(2+) handling. Recent evidence has revealed that adults with severe growth hormone (GH) and insulin-like growth factor I (IGF-1) deficiency such as Laron syndrome display increased risk of stroke and cardiovascular diseases. IGF-1 signaling may regulate contractility, metabolism, hypertrophy, apoptosis, autophagy, stem cell regeneration and senescence in the heart to maintain cardiac homeostasis. An inverse relationship between plasma IGF-1 levels and prevalence of metabolic syndrome as well as associated cardiovascular complications has been identified, suggesting the clinical promises of IGF-1 analogues or IGF-1 receptor activation in the management of metabolic and cardiovascular diseases. However, the underlying pathophysiological mechanisms between IGF-1 and metabolic syndrome are still poorly understood. This mini-review will discuss the role of IGF-1 signaling cascade in the prevalence of metabolic syndrome in particular the susceptibility to overnutrition and sedentary life style-induced obesity, dyslipidemia, insulin resistance and other features of metabolic syndrome. Special attention will be dedicated in IGF-1-associated changes in cardiac responses in various metabolic syndrome components such as insulin resistance, obesity, hypertension and dyslipidemia. The potential risk of IGF-1 and IGF-1R stimulation such as tumorigenesis is discussed. Therapeutic promises of IGF-1 and IGF-1 analogues including mecasermin, mecasermin rinfabate and PEGylated IGF-1 will be discussed.
Collapse
Affiliation(s)
- Jun Ren
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Center for Cardiovascular Research and Alternative Medicine, University of Wyoming College of Health Sciences, Laramie, WY 82071, USA.
| | - Piero Anversa
- Departments of Anesthesia and Medicine and Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
14
|
Papatheodorou SI, Rohrmann S, Lopez DS, Bradwin G, Joshu CE, Kanarek N, Nelson WG, Rifai N, Platz EA, Tsilidis KK. Association between endogenous sex steroid hormones and insulin-like growth factor proteins in US men. Cancer Causes Control 2014; 25:353-63. [PMID: 24395140 DOI: 10.1007/s10552-013-0336-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2013] [Accepted: 12/27/2013] [Indexed: 11/25/2022]
Abstract
PURPOSE Sex steroid hormone concentrations and insulin-like growth factor (IGF) proteins have been independently associated with risk of cancer, chronic diseases, and mortality. However, studies that evaluated the inter-relation between the sex hormones and IGF pathways have provided mixed results. We examined the association between endogenous sex hormones and sex hormone-binding globulin (SHBG) with IGF-1 and IGF-binding protein 3 (IGFBP-3) in a population-based sample of US men. METHODS Data from 1,135 men aged 20 years or older participating in the third National Health and Nutrition Examination Survey (NHANES III) were analyzed. Weighted linear regression was used to estimate geometric means and 95 % confidence intervals for IGF-1 and IGFBP-3 concentrations by sex steroid hormones and SHBG after adjusting for age, race/ethnicity, body mass index, waist circumference, alcohol consumption, cigarette smoking, physical activity, diabetes, and mutually adjusting for other sex hormones and SHBG. RESULTS No significant association was observed between sex steroid hormones, SHBG, and IGF-1 concentrations. Total estradiol (% difference in Q5 - Q1 geometric means -9.7 %; P-trend 0.05) and SHBG (% difference -7.3 %; P-trend 0.02) were modestly inversely associated with IGFBP-3. Total testosterone was modestly inversely associated with IGFBP-3 (% difference -6.2 %; P-trend 0.01), but this association disappeared after adjustment for total estradiol and SHBG (% difference 2.6 %; P-trend 0.23). Androstanediol glucuronide was not associated with IGFBP-3. CONCLUSIONS These findings suggest that there may be inter-relationships between circulating total estradiol, SHBG, and IGFBP-3 concentrations. Future research may consider these inter-relationships when evaluating potential joint effects of the sex hormones and IGF pathways.
Collapse
Affiliation(s)
- Stefania I Papatheodorou
- Cyprus International Institute for Environmental and Public Health, Cyprus University of Technology, Limassol, Cyprus
| | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Maggio M, Cattabiani C, Lauretani F, Bandinelli S, De Vita F, Dall'Aglio E, Corsonello A, Lattanzio F, Paolisso G, Ferrucci L, Ceda GP. Insulin-like growth factor-1 bioactivity plays a prosurvival role in older participants. J Gerontol A Biol Sci Med Sci 2013; 68:1342-50. [PMID: 23671288 DOI: 10.1093/gerona/glt045] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
The aim of this study was to address the intriguing issue of the role of the insulin-like growth factor (IGF)-1 system in longevity looking at the role of different components of IGF system. Vital status was ascertained in 1,197 men and women aged greater than or equal to 65 years from the InCHIANTI study. Hormonal levels were categorized into quartiles, and ratio of IGF-1 to IGF-binding protein (IGFBP)-1 was calculated. The relationship between hormones and mortality was tested by Cox proportional hazard models adjusted for age, sex, and confounders. During the 8-year follow-up period, 240 died and 957 survived. Lowest quartiles of IGF-1 and IGFBP-1 were considered as reference. Compared with the lowest quartiles, IGF-1 in upper quartiles was a negative predictor of mortality independent of age and sex (p = .01) but not independent of IGFBP-1 and other confounders. IGFBP-1 in second-third quartiles was negatively associated and that in the fourth quartiles was positively associated with risk of death. IGF-1/IGFBP-1 ratio in the lowest quartiles was a strong positive predictor of mortality, in age- and sex-adjusted model (p = .005), and independent of additional confounders (p = .037). High IGFBP-1 and low IGF-1/IGFBP-1 ratio are associated with all-cause mortality in older population.
Collapse
Affiliation(s)
- Marcello Maggio
- Section of Geriatrics, Department of Clinical and Experimental Medicine, University of Parma, via Gramsci 14, 43100 Parma, Italy.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
|
17
|
Jong HL, Mustafa MR, Vanhoutte PM, AbuBakar S, Wong PF. MicroRNA 299-3p modulates replicative senescence in endothelial cells. Physiol Genomics 2013; 45:256-67. [PMID: 23362143 DOI: 10.1152/physiolgenomics.00071.2012] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
MicroRNAs (miRNAs) regulate various cellular processes. While several genes associated with replicative senescence have been described in endothelial cells, miRNAs that regulate these genes remain largely unknown. The present study was designed to identify miRNAs associated with replicative senescence and their target genes in human umbilical vein endothelial cells (HUVECs). An integrated miRNA and gene profiling approach revealed that hsa-miR-299-3p is upregulated in senescent HUVECs compared with the young cells, and one of its target genes could be IGF1. IGF1 was upregulated in senescent compared with young HUVECs, and knockdown of hsa-miR-299-3p dose-dependently increased the mRNA expression of IGF1, more significantly observed in the presenescent cells (passage 19) compared with the senescent cells (passage 25). Knockdown of hsa-miR-299-3p also resulted in significant reduction in the percentage of cells positively stained for senescence-associated β-galactosidase and increases in cell viability measured by MTT assay but marginal increases in cell proliferation and cell migration capacity measured by real-time growth kinetics analysis. Moreover, knockdown of hsa-miR-299-3p also increased proliferation of cells treated with H2O2 to induce senescence. These findings suggest that hsa-miR-299-3p may delay or protect against replicative senescence by improving the metabolic activity of the senesced cells but does not stimulate growth of the remaining cells in senescent cultures. Hence, these findings provide an early insight into the role of hsa-miR-299-3p in the modulation of replicative senescence in HUVECs.
Collapse
Affiliation(s)
- Hui-Lan Jong
- Department of Pharmacology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | | | | | | | | |
Collapse
|
18
|
Ibarra C, Vicencio JM, Estrada M, Lin Y, Rocco P, Rebellato P, Munoz JP, Garcia-Prieto J, Quest AFG, Chiong M, Davidson SM, Bulatovic I, Grinnemo KH, Larsson O, Szabadkai G, Uhlén P, Jaimovich E, Lavandero S. Local control of nuclear calcium signaling in cardiac myocytes by perinuclear microdomains of sarcolemmal insulin-like growth factor 1 receptors. Circ Res 2012; 112:236-45. [PMID: 23118311 DOI: 10.1161/circresaha.112.273839] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
RATIONALE The ability of a cell to independently regulate nuclear and cytosolic Ca(2+) signaling is currently attributed to the differential distribution of inositol 1,4,5-trisphosphate receptor channel isoforms in the nucleoplasmic versus the endoplasmic reticulum. In cardiac myocytes, T-tubules confer the necessary compartmentation of Ca(2+) signals, which allows sarcomere contraction in response to plasma membrane depolarization, but whether there is a similar structure tunneling extracellular stimulation to control nuclear Ca(2+) signals locally has not been explored. OBJECTIVE To study the role of perinuclear sarcolemma in selective nuclear Ca(2+) signaling. METHODS AND RESULTS We report here that insulin-like growth factor 1 triggers a fast and independent nuclear Ca(2+) signal in neonatal rat cardiac myocytes, human embryonic cardiac myocytes, and adult rat cardiac myocytes. This fast and localized response is achieved by activation of insulin-like growth factor 1 receptor signaling complexes present in perinuclear invaginations of the plasma membrane. The perinuclear insulin-like growth factor 1 receptor pool connects extracellular stimulation to local activation of nuclear Ca(2+) signaling and transcriptional upregulation through the perinuclear hydrolysis of phosphatidylinositol 4,5-biphosphate inositol 1,4,5-trisphosphate production, nuclear Ca(2+) release, and activation of the transcription factor myocyte-enhancing factor 2C. Genetically engineered Ca(2+) buffers--parvalbumin--with cytosolic or nuclear localization demonstrated that the nuclear Ca(2+) handling system is physically and functionally segregated from the cytosolic Ca(2+) signaling machinery. CONCLUSIONS These data reveal the existence of an inositol 1,4,5-trisphosphate-dependent nuclear Ca(2+) toolkit located in direct apposition to the cell surface, which allows the local control of rapid and independent activation of nuclear Ca(2+) signaling in response to an extracellular ligand.
Collapse
Affiliation(s)
- Cristian Ibarra
- Centro de Estudios Moleculares de la Célula, Facultad de Ciencias Quimicas y Farmaceuticas, Universidad de Chile, Santiago, Chile
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Li J, Pei M. Cell Senescence: A Challenge in Cartilage Engineering and Regeneration. TISSUE ENGINEERING PART B-REVIEWS 2012; 18:270-87. [PMID: 22273114 DOI: 10.1089/ten.teb.2011.0583] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- Jingting Li
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, West Virginia
- Division of Exercise Physiology, West Virginia University, Morgantown, West Virginia
| | - Ming Pei
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, West Virginia
- Division of Exercise Physiology, West Virginia University, Morgantown, West Virginia
- Mechanical and Aerospace Engineering, West Virginia University, Morgantown, West Virginia
| |
Collapse
|
20
|
Stem cells as a tool for breast imaging. JOURNAL OF ONCOLOGY 2012; 2012:814014. [PMID: 22848220 PMCID: PMC3405672 DOI: 10.1155/2012/814014] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/14/2012] [Accepted: 06/18/2012] [Indexed: 12/16/2022]
Abstract
Stem cells are a scientific field of interest due to their therapeutic potential. There are different groups, depending on the differentiation state. We can find lonely stem cells, but generally they distribute in niches. Stem cells don't survive forever. They are affected for senescence. Cancer stem cells are best defined functionally, as a subpopulation of tumor cells that can enrich for tumorigenic property and can regenerate heterogeneity of the original tumor. Circulating tumor cells are cells that have detached from a primary tumor and circulate in the bloodstream. They may constitute seeds for subsequent growth of additional tumors (metastasis) in different tissues. Advances in molecular imaging have allowed a deeper understanding of the in vivo behavior of stem cells and have proven to be indispensable in preclinical and clinical studies. One of the first imaging modalities for monitoring pluripotent stem cells in vivo, magnetic resonance imaging (MRI) offers high spatial and temporal resolution to obtain detailed morphological and functional information. Advantages of radioscintigraphic techniques include their picomolar sensitivity, good tissue penetration, and translation to clinical applications. Radionuclide imaging is the sole direct labeling technique used thus far in human studies, involving both autologous bone marrow derived and peripheral stem cells.
Collapse
|
21
|
Friedrich N, Schneider HJ, Haring R, Nauck M, Völzke H, Kroemer HK, Dörr M, Klotsche J, Jung-Sievers C, Pittrow D, Lehnert H, März W, Pieper L, Wittchen HU, Wallaschofski H, Stalla GK. Improved prediction of all-cause mortality by a combination of serum total testosterone and insulin-like growth factor I in adult men. Steroids 2012; 77:52-8. [PMID: 22037276 DOI: 10.1016/j.steroids.2011.10.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2011] [Revised: 10/11/2011] [Accepted: 10/11/2011] [Indexed: 11/18/2022]
Abstract
OBJECTIVE Lower levels of anabolic hormones in older age are well documented. Several studies suggested that low insulin-like growth factor I (IGF-I) or testosterone levels were related to increased mortality. The aim of the present study was to investigate the combined influence of low IGF-I and low testosterone on all-cause mortality in men. METHODS AND RESULTS From two German prospective cohort studies, the DETECT study and SHIP, 3942 men were available for analyses. During 21,838 person-years of follow-up, 8.4% (n=330) of men died. Cox model analyses with age as timescale and adjusted for potential confounders revealed that men with levels below the 10th percentile of at least one hormone [hazard ratio (HR) 1.38 (95% confidence-interval (CI) 1.06-1.78), p=0.02] and two hormones [HR 2.88 (95% CI 1.32-6.29), p<0.01] showed a higher risk of all-cause mortality compared to men with non-low hormones. The associations became non-significant by using the 20th percentile as cut-off showing that the specificity increased with lower cut-offs for decreased hormone levels. The inclusion of both IGF-I and total testosterone in a mortality prediction model with common risk factors resulted in a significant integrated discrimination improvement of 0.5% (95% CI 0.3-0.7%, p=0.03). CONCLUSIONS Our results prove that multiple anabolic deficiencies have a higher impact on mortality than a single anabolic deficiency and suggest that assessment of more than one anabolic hormone as a biomarker improve the prediction of all-cause mortality.
Collapse
Affiliation(s)
- Nele Friedrich
- Institute of Clinical Chemistry and Laboratory Medicine, Ernst Moritz Arndt University, Greifswald, Germany.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Hu J, Li C, Wang L, Zhang X, Zhang M, Gao H, Yu X, Wang F, Zhao W, Yan S, Wang Y. Long term effects of the implantation of autologous bone marrow mononuclear cells for type 2 diabetes mellitus. Endocr J 2012; 59:1031-9. [PMID: 22814142 DOI: 10.1507/endocrj.ej12-0092] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Previous studies have shown that several types of stem cells can differentiate into insulin-secreting islet beta-cells and that these cells can reduce blood glucose in some trials, but there has been no report of a long-term follow-up. We assessed the long-term effects of the use of autologous bone marrow mononuclear cells in the treatment of type 2 diabetes mellitus (T2DM). Based on the willingness to receive implantation of bone marrow mononuclear cells, One hundred and eighteen patients with T2DM were divided into two groups; the patients in group I were treated with autologous bone marrow mononuclear cells and patients in group II were treated with insulin intensification therapy. Mononuclear cells from bone marrow were injected back into the patient's pancreas via a catheter. Patients were followed-up after the operation at monthly intervals for the first 3 months and thereafter every 3 months for the next 33 months, the occurrence of any side effects and the results of laboratory examinations were evaluated. There were no reported acute or chronic side effects in group I and both the HbA1c and C-peptide in group I patients were significantly better than either pretherapy values or group II patients during the follow-up period. These data suggested that the implantation of autologous bone marrow mononuclear cells for the treatment of T2DM is safe and effective. This therapy can partially restore the function of islet beta-cells and maintain blood glucose homeostasis in a longer time.
Collapse
Affiliation(s)
- Jianxia Hu
- Stem Cell Research Center, the Affiliated Hospital of Medical College, Qingdao University, Qingdao 266003, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Abstract
IGF-1 (insulin-like growth factor-1) plays a unique role in the cell protection of multiple systems, where its fine-tuned signal transduction helps to preserve tissues from hypoxia, ischaemia and oxidative stress, thus mediating functional homoeostatic adjustments. In contrast, its deprivation results in apoptosis and dysfunction. Many prospective epidemiological surveys have associated low IGF-1 levels with late mortality, MI (myocardial infarction), HF (heart failure) and diabetes. Interventional studies suggest that IGF-1 has anti-atherogenic actions, owing to its multifaceted impact on cardiovascular risk factors and diseases. The metabolic ability of IGF-1 in coupling vasodilation with improved function plays a key role in these actions. The endothelial-protective, anti-platelet and anti-thrombotic activities of IGF-1 exert critical effects in preventing both vascular damage and mechanisms that lead to unstable coronary plaques and syndromes. The pro-survival and anti-inflammatory short-term properties of IGF-1 appear to reduce infarct size and improve LV (left ventricular) remodelling after MI. An immune-modulatory ability, which is able to suppress 'friendly fire' and autoreactivity, is a proposed important additional mechanism explaining the anti-thrombotic and anti-remodelling activities of IGF-1. The concern of cancer risk raised by long-term therapy with IGF-1, however, deserves further study. In the present review, we discuss the large body of published evidence and review data on rhIGF-1 (recombinant human IGF-1) administration in cardiovascular disease and diabetes, with a focus on dosage and safety issues. Perhaps the time has come for the regenerative properties of IGF-1 to be assessed as a new pharmacological tool in cardiovascular medicine.
Collapse
|
24
|
Madonna R, Renna FV, Cellini C, Cotellese R, Picardi N, Francomano F, Innocenti P, De Caterina R. Age-dependent impairment of number and angiogenic potential of adipose tissue-derived progenitor cells. Eur J Clin Invest 2011; 41:126-33. [PMID: 20874854 DOI: 10.1111/j.1365-2362.2010.02384.x] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND Adipose tissue-derived stromal cells (ADSCs) are being recognized as a source of stem cells potentially useful for cardiovascular repair. We analysed the abundance and angiogenic activity of adipose tissue-derived progenitor cells (PCs) in elderly patients most likely to benefit from this novel source of stem cells. MATERIALS AND METHODS Fifty-two subjects (aged 68 ± 13 years) with variable degrees of cardiovascular risk underwent abdominal surgery for intercurrent diseases. Visceral adipose tissue (3 ± 1 g visceral fat per patient) was processed with type-1 collagenase to obtain ADSCs from the stromal-vascular fraction. Adipose tissue-derived PCs were quantified by flow cytometry as %CD45(-)/CD34(+)/CD133(+) cells of total ADSCs. Matrigel angiogenesis assay was used to analyse the ability of ADSCs to form tubes or networks. RESULTS We found no correlations between number of CD45(-)/CD34(+)/CD133(+) or total ADSCs and quantitative risk parameters including total cholesterol, low density lipoprotein cholesterol, high density lipoprotein cholesterol, waist circumference, body mass index, and systolic and diastolic arterial pressure. However, increasing age (r = -0·31, P < 0·05) significantly and inversely correlated with levels of adipose tissue-derived CD45(-)/CD34(+)/CD133(+) cells in Matrigel angiogenesis assays; increasing age (r = -0·29, P < 0·05) was related to a reduction of ADSC-derived tubulization. CONCLUSIONS Ageing may alter the availability of adipose tissue-derived CD45(-)/CD34(+)/CD133(+) cells and their angiogenic functional capacity. Such changes may impair the use of adipose tissue as source of autologous PCs in elderly patients.
Collapse
Affiliation(s)
- Rosalinda Madonna
- Department of Cardiology and Center of Excellence on Aging, G. d'Annunzio University-Chieti, Chieti, Italy
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Khan M, Akhtar S, Mohsin S, N Khan S, Riazuddin S. Growth factor preconditioning increases the function of diabetes-impaired mesenchymal stem cells. Stem Cells Dev 2010; 20:67-75. [PMID: 20446810 DOI: 10.1089/scd.2009.0397] [Citation(s) in RCA: 84] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Bone marrow-derived mesenchymal stem cells (MSCs) possess multilineage differentiation potential and can be used for the treatment of diabetic heart failure. However, hyperglycemia can affect the function of MSCs adversely and merits the requirement for a strategy to correct this anomaly. MSCs were isolated from the tibias and femurs of C57BL/6 wild-type mice at 60 days after induction of diabetes by streptozotocin. MSCs were characterized by flow cytometry for CD44 (97.7%), CD90 (95.4%), and CD105 (92.3%) markers and were preconditioned with insulin-like growth factor-1 (IGF-1) (50 ng/mL) and fibroblast growth factor-2 (FGF-2) (50 ng/mL) in combination for 1 h in serum-free Iscove's modified Dulbecco's medium. This was followed by hypoxic and high glucose insults to mimic diabetic heart microenvironment and to study the effect of preconditioning. Diabetic MSCs after treatment showed upregulation of IGF-1, FGF-2, Akt, GATA-4, and Nkx 2.5 and downregulation of p16(INK4a), p66(shc), p53, Bax, and Bak. Under hypoxic stress, preconditioned diabetic MSCs showed high superoxide dismutase activity (52.3%) compared with untreated cells (36.9%). This was concomitant with low numbers of annexin-V-positive cells, high in vitro tube-forming ability, and high chemotactic mobility to stromal cell-derived factor-1α after preconditioning in diabetic MSCs. Upregulation of Ang-I and VEGF and downregulation of p16(INK4a) were also observed in preconditioned cells under conditions of high glucose insult. Therefore, preconditioning with IGF-1 and FGF-2 in combination represents a novel strategy to augment MSC function affected by diabetes and holds significance for future strategies to treat diabetic heart failure.
Collapse
Affiliation(s)
- Mohsin Khan
- National Center of Excellence in Molecular Biology, University of the Punjab, Lahore, Pakistan
| | | | | | | | | |
Collapse
|
26
|
Gläser S, Friedrich N, Ewert R, Schäper C, Nauck M, Dörr M, Völzke H, Felix SB, Krebs A, Wallaschofski H, Koch B. Association between serum insulin-like growth factor (IGF) I and IGF binding protein-3 and lung function. J Clin Endocrinol Metab 2009; 94:2452-8. [PMID: 19401363 DOI: 10.1210/jc.2008-2662] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
BACKGROUND There is strong evidence that IGF-I and IGF binding protein 3 (IGFBP-3), as central mediators of endocrine and finally metabolic or anabolic effects of GH, were associated with increased lung size in acromegaly or a decrease of respiratory muscle pressures in patients with GH deficiency. The aim of the present study was to further clarify the impact of IGF-I and IGFBP-3 levels on lung volumes and respiratory pressures in a general adult population. MATERIAL AND METHODS From the Study of Health in Pomerania, 1326 subjects aged 25 to 85 yr participated in standardized pulmonary function testing. IGF-I and IGFBP-3 levels were measured with the Immulite 2500 system. Multivariable linear regression analyses adjusted for age, sex, body mass index, physical activity, and smoking were performed. RESULTS In men, positive linear associations between IGF-I and IGF-I/IGFBP-3 ratio with forced expiratory volume in 1 sec (FEV1) as well as with forced vital capacity (FVC) were detected across all ages, whereas in women this positive association was only detectable above 50 yr. Furthermore, the analyses indicated positive linear relations of IGF-I/IGFBP-3 ratio with FEV1 and FVC, respectively. No significant relations between IGF-I or IGFBP-3 and maximal inspiratory pressure was detectable in both sexes. CONCLUSION In conclusion, higher IGF-I levels were associated with higher lung volumes in men, whereas in women this association was only detectable in subjects older than 50 yr. Higher IGF-I values were not associated with increased respiratory muscle strength measured as maximal inspiratory pressure.
Collapse
Affiliation(s)
- Sven Gläser
- Department of Internal Medicine B, Pulmonary Medicine and Infectious Diseases, Ernst-Moritz-Arndt University Greifswald, D-17475 Greifswald, Germany.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Karbownik-Lewinska M, Kokoszko A, Lewandowski KC, Shalet SM, Lewinski A. GH replacement reduces increased lipid peroxidation in GH-deficient adults. Clin Endocrinol (Oxf) 2008; 68:957-64. [PMID: 18031310 DOI: 10.1111/j.1365-2265.2007.03142.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
BACKGROUND GH replacement improves numerous metabolic abnormalities in GH-deficient patients; increased lipid peroxidation (LPO) has been observed in GH-deficient patients; however, it is unknown if LPO is influenced by GH replacement. AIM AND METHODS To evaluate the extent to which GH replacement might reverse the increased LPO in GH-deficient adults and to analyse if this phenomenon might be involved in the improvement of metabolic disturbances due to GH treatment. Serum concentrations of malondialdehyde + 4-hydroxyalkenals (MDA + 4-HDA), as an index of LPO, were measured at baseline, and after 12 and 24 months of GH replacement in 40 adult patients with severe GH deficiency (both in adult- and childhood-onset) and in 40 healthy volunteers, matched for sex, age and body mass index (BMI). Correlations were evaluated between LPO and lipids, IGF-I, metalloproteinase-2 and -9 (MMP-2, -9), vascular endothelial growth factor (VEGF), BMI and GH dose. RESULTS LPO values in GH-deficient patients were several-fold higher than in controls [55.36 +/- 2.27 vs. 4.19 +/- 0.42 nmol/mg protein (mean +/- SEM), P < 0.0001] and decreased significantly over time with GH replacement to 38.61 +/- 2.15 nmol/mg protein (i.e. by approximately 30%), though still remaining markedly elevated compared with controls (P < 0.0001). The proatherogenic lipid profile parameters correlated positively with LPO in the childhood-onset subgroup before GH replacement. GH replacement restored the positive correlation between LPO and age in male patients (r = 0.57, P = 0.013; r = 0.8, P < 0.001, at 12 and 24 months of GH replacement, respectively). CONCLUSIONS GH replacement partially reverses the grossly abnormal LPO in GH-deficient adults. It is highly probable, therefore, that oxidative mechanisms are involved in the overall improvement of metabolic changes due to GH replacement.
Collapse
|
28
|
Hsu CP, Odewale I, Alcendor RR, Sadoshima J. Sirt1 protects the heart from aging and stress. Biol Chem 2008; 389:221-31. [DOI: 10.1515/bc.2008.032] [Citation(s) in RCA: 85] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Abstract
The prevalence of heart diseases, such as coronary artery disease and congestive heart failure, increases with age. Optimal therapeutic interventions that antagonize aging may reduce the occurrence and mortality of adult heart diseases. We discuss here how molecular mechanisms mediating life span extension affect aging of the heart and its resistance to pathological insults. In particular, we review our recent findings obtained from transgenic mice with cardiac-specific overexpression of Sirt1, which demonstrated delayed aging and protection against oxidative stress in the heart. We propose that activation of known longevity mechanisms in the heart may represent a novel cardioprotection strategy against aging and certain types of cardiac stress, such as oxidative stress.
Collapse
|
29
|
McMullen JR. ROLE OF INSULIN-LIKE GROWTH FACTOR 1 AND PHOSPHOINOSITIDE 3-KINASE IN A SETTING OF HEART DISEASE. Clin Exp Pharmacol Physiol 2008; 35:349-54. [DOI: 10.1111/j.1440-1681.2007.04873.x] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
30
|
Hinkal G, Donehower LA. How does suppression of IGF-1 signaling by DNA damage affect aging and longevity? Mech Ageing Dev 2008; 129:243-53. [PMID: 18374391 DOI: 10.1016/j.mad.2008.02.005] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2008] [Revised: 02/11/2008] [Accepted: 02/13/2008] [Indexed: 01/01/2023]
Abstract
Long-lived animals have evolved a robust set of defenses to maintain genomic integrity over their entire lifespan. The DNA damage response and DNA repair pathways are critical pillars of organismal defenses, minimizing somatic mutations in both post-mitotic and mitotic cells. These genomic maintenance systems not only prevent the premature emergence of cancers but may also maintain normal tissue function and organismal longevity. Genetic defects in a number of DNA repair and DNA damage response genes often leads to a dramatic increase in cancer incidence; in other cases, premature aging or progeroid syndromes may be induced. In this review, we discuss two recent reports of two nucleotide excision repair-deficient models that exhibit dramatic premature aging and shortened longevity. The DNA repair defects were also associated with a significant inhibition of the growth hormone/insulin-like growth factor 1 (GH/IGF-1) axis, an endocrine signaling pathway shown to influence aging and longevity in both vertebrates and invertebrates. Potential mechanisms of how DNA damage might affect IGF-1 signaling and aging are discussed, with a particular emphasis on the role of such signaling alterations in the adult tissue stem cell compartments.
Collapse
Affiliation(s)
- George Hinkal
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA
| | | |
Collapse
|
31
|
Li M, Naqvi N, Yahiro E, Liu K, Powell PC, Bradley WE, Martin DIK, Graham RM, Dell'Italia LJ, Husain A. c-kit is required for cardiomyocyte terminal differentiation. Circ Res 2008; 102:677-85. [PMID: 18258857 DOI: 10.1161/circresaha.107.161737] [Citation(s) in RCA: 75] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
c-kit, the transmembrane tyrosine kinase receptor for stem cell factor, is required for melanocyte and mast cell development, hematopoiesis, and differentiation of spermatogonial stem cells. We show here that in the heart, c-kit is expressed not only by cardiac stem cells but also by cardiomyocytes, commencing immediately after birth and terminating a few days later, coincident with the onset of cardiomyocyte terminal differentiation. To examine the function of c-kit in cardiomyocyte terminal differentiation, we used compound heterozygous mice carrying the W (null) and W(v) (dominant negative) mutations of c-kit. In vivo, adult W/W(v) cardiomyocytes are phenotypically indistinguishable from their wild-type counterparts. After acute pressure overload adult W/W(v) cardiomyocytes reenter the cell cycle and proliferate, leading to left ventricular growth; furthermore in transgenic mice with cardiomyocyte-restricted overexpression of the dominant negative W(v) mutant, pressure overload causes cardiomyocytes to reenter the cell cycle. In contrast, in wild-type mice left ventricular growth after pressure overload results mainly from cardiomyocyte hypertrophy. Importantly, W/W(v) mice with pressure overload-induced cardiomyocyte hyperplasia had improved left ventricular function and survival. In W/W(v) mice, c-kit dysfunction also resulted in an approximately 14-fold decrease (P<0.01) in the number of c-kit(+)/GATA4(+) cardiac progenitors. These findings identify novel functions for c-kit: promotion of cardiac stem cell differentiation and regulation of cardiomyocyte terminal differentiation.
Collapse
Affiliation(s)
- Ming Li
- Departments of Physiology and Biophysics, University of Alabama at Birmingham, AL 35294, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Maggio M, Lauretani F, Ceda GP, Bandinelli S, Ling SM, Metter EJ, Artoni A, Carassale L, Cazzato A, Ceresini G, Guralnik JM, Basaria S, Valenti G, Ferrucci L. Relationship between low levels of anabolic hormones and 6-year mortality in older men: the aging in the Chianti Area (InCHIANTI) study. ACTA ACUST UNITED AC 2007; 167:2249-54. [PMID: 17998499 DOI: 10.1001/archinte.167.20.2249] [Citation(s) in RCA: 142] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
BACKGROUND Aging in men is characterized by a progressive decline in levels of anabolic hormones, such as testosterone, insulinlike growth factor 1 (IGF-1), and dehydroepiandrosterone sulfate (DHEA-S). We hypothesized that in older men a parallel age-associated decline in bioavailable testosterone, IGF-1, and DHEA-S secretion is associated with higher mortality independent of potential confounders. METHODS Testosterone, IGF-1, DHEA-S, and demographic features were evaluated in a representative sample of 410 men 65 years and older enrolled in the Aging in the Chianti Area (InCHIANTI) study. A total of 126 men died during the 6-year follow-up. Thresholds for lowest-quartile definitions were 70 ng/dL (to convert to nanomoles per liter, multiply by 0.0347) for bioavailable testosterone, 63.9 ng/mL (to convert to nanomoles per liter, multiply by 0.131) for total IGF-1, and 50 microg/dL (to convert to micromoles per liter, multiply by 0.027) for DHEA-S. Men were divided into 4 groups: no hormone in the lowest quartile (reference) and 1, 2, and 3 hormones in the lowest quartiles. Kaplan-Meier survival and Cox proportional hazards models adjusted for confounders were used in the analysis. RESULTS Compared with men with levels of all 3 hormones above the lowest quartiles, having 1, 2, and 3 dysregulated hormones was associated with hazard ratios for mortality of 1.47 (95% confidence interval [CI], 0.88-2.44), 1.85 (95% CI, 1.04-3.30), and 2.29 (95% CI, 1.12-4.68), respectively (test for trend, P <.001). In the fully adjusted analysis, only men with 3 anabolic hormone deficiencies had a significant increase in mortality (hazard ratio, 2.44; 95% CI, 1.09-5.46 (test for trend, P <.001). CONCLUSIONS Age-associated decline in anabolic hormone levels is a strong independent predictor of mortality in older men. Having multiple hormonal deficiencies rather than a deficiency in a single anabolic hormone is a robust biomarker of health status in older persons.
Collapse
Affiliation(s)
- Marcello Maggio
- National Institute on Aging, National Institutes of Health, NIA Clinical Research Branch at Harbor Hospital, 3001 S Hanover Street, Baltimore, MD 21225, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Li Q, Ren J. Influence of cardiac-specific overexpression of insulin-like growth factor 1 on lifespan and aging-associated changes in cardiac intracellular Ca2+ homeostasis, protein damage and apoptotic protein expression. Aging Cell 2007; 6:799-806. [PMID: 17973971 DOI: 10.1111/j.1474-9726.2007.00343.x] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
A fall in circulating levels of cardiac survival factor insulin-like growth factor 1 (IGF-1) contributes to cardiac aging. To better understand the role of IGF-1 in cardiac aging, we examined the influence of cardiac IGF-1 overexpression on lifespan, cardiomyocyte intracellular Ca2+ homeostasis, protein damage, apoptosis and expression of pro- and anti-apoptotic proteins in young and old mice. Mouse survival rate was constructed by the Kaplan-Meier curve. Intracellular Ca2+ was evaluated by fura-2 fluorescence. Protein damage was determined by protein carbonyl formation. Apoptosis was assessed by caspase-8 expression, caspase-3 and TUNEL (terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling) assay. Pro- and anti-apoptotic proteins including Bax, p53, pp53, Bcl2, Omi/HtrA2, apoptosis repressor with caspase recruitment domain (ARC) and X-linked inhibitor of apoptosis protein (XIAP) were assessed by Western blot. Aging decreased plasma in IGF-1 levels, elevated myocyte resting intracellular Ca2+ levels, reduced electrically stimulated rise in intracellular Ca2+ and delayed intracellular Ca2+ decay associated with enhanced protein carbonyl formation, caspase-8 expression and caspase-3 activity in FVB mice, all of which with the exception of elevated resting intracellular Ca2+ were attenuated by IGF-1. Aging up-regulated expression of Bax, Bcl2 and ARC, down-regulated XIAP expression and did not affect p53, pp53 and Omi/HtrA2. The IGF-1 transgene attenuated or nullified aging-induced changes in Bax, Bcl2 and XIAP. Our data suggest a beneficial role for IGF-1 in aging-induced survival, cardiac intracellular Ca2+ homeostasis, protein damage and apoptosis possibly related to pro- and anti-apoptotic proteins.
Collapse
Affiliation(s)
- Qun Li
- Division of Pharmaceutical Sciences & Center for Cardiovascular Research and Alternative Medicine, University of Wyoming, Laramie, WY 82071, USA
| | | |
Collapse
|
34
|
Rota M, Hosoda T, De Angelis A, Arcarese ML, Esposito G, Rizzi R, Tillmanns J, Tugal D, Musso E, Rimoldi O, Bearzi C, Urbanek K, Anversa P, Leri A, Kajstura J. The young mouse heart is composed of myocytes heterogeneous in age and function. Circ Res 2007; 101:387-99. [PMID: 17601802 DOI: 10.1161/circresaha.107.151449] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The recognition that the adult heart continuously renews its myocyte compartment raises the possibility that the age and lifespan of myocytes does not coincide with the age and lifespan of the organ and organism. If this were the case, myocyte turnover would result at any age in a myocardium composed by a heterogeneous population of parenchymal cells which are structurally integrated but may contribute differently to myocardial performance. To test this hypothesis, left ventricular myocytes were isolated from mice at 3 months of age and the contractile, electrical, and calcium cycling characteristics of these cells were determined together with the expression of the senescence-associated protein p16(INK4a) and telomere length. The heart was characterized by the coexistence of young, aged, and senescent myocytes. Old nonreplicating, p16(INK4a)-positive, hypertrophied myocytes with severe telomeric shortening were present together with young, dividing, p16(INK4a)-negative, small myocytes with long telomeres. A class of myocytes with intermediate properties was also found. Physiologically, evidence was obtained in favor of the critical role that action potential (AP) duration and I(CaL) play in potentiating Ca(2+) cycling and the mechanical behavior of young myocytes or in decreasing Ca(2+) transients and the performance of senescent hypertrophied cells. The characteristics of the AP appeared to be modulated by the transient outward K(+) current I(to) which was influenced by the different expression of the K(+) channels subunits. Collectively, these observations at the physiological and structural cellular level document that by necessity the heart has to constantly repopulate its myocyte compartment to replace senescent poorly contracting myocytes with younger more efficient cells. Thus, cardiac homeostasis and myocyte turnover regulate cardiac function.
Collapse
Affiliation(s)
- Marcello Rota
- Cardiovascular Research Institute, Department of Medicine, New York Medical College, Valhalla, New York 10595, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Abstract
It is well established that cardiovascular repair mechanisms become progressively impaired with age and that advanced age is itself a significant risk factor for cardiovascular disease. Although therapeutic developments have improved the prognosis for those with cardiovascular disease, mortality rates have nevertheless remained virtually unchanged in the last twenty years. Clearly, there is a need for alternative strategies for the treatment of cardiovascular disease. In recent years, the idea that the heart is capable of regeneration has raised the possibility that cell-based therapies may provide such an alternative to conventional treatments. Cells that have the potential to generate cardiomyocytes and vascular cells have been identified in both the adult heart and peripheral tissues, and in vivo experiments suggest that these cardiovascular stem cells and cardiovascular progenitor cells, including endothelial progenitor cells, are capable of replacing damaged myocardium and vascular tissues. Despite these findings, the endogenous actions of cardiovascular stem cells and cardiovascular progenitor cells appear to be insufficient to protect against cardiovascular disease in older individuals. Because recent evidence suggests that cardiovascular stem cells and cardiovascular progenitor cells are subject to age-associated changes that impair their function, these changes may contribute to the dysregulation of endogenous cardiovascular repair mechanisms in the aging heart and vasculature. Here we present the evidence for the impact of aging on cardiovascular stem cell/cardiovascular progenitor cell function and its potential importance in the increased severity of cardiovascular pathophysiology observed in the geriatric population.
Collapse
Affiliation(s)
- Victoria L T Ballard
- Department of Medicine, Weill Medical College of Cornell University, New York, USA
| | | |
Collapse
|
36
|
Alcendor RR, Gao S, Zhai P, Zablocki D, Holle E, Yu X, Tian B, Wagner T, Vatner SF, Sadoshima J. Sirt1 regulates aging and resistance to oxidative stress in the heart. Circ Res 2007; 100:1512-21. [PMID: 17446436 DOI: 10.1161/01.res.0000267723.65696.4a] [Citation(s) in RCA: 870] [Impact Index Per Article: 48.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Silent information regulator (Sir)2, a class III histone deacetylase, mediates lifespan extension in model organisms and prevents apoptosis in mammalian cells. However, beneficial functions of Sir2 remain to be shown in mammals in vivo at the organ level, such as in the heart. We addressed this issue by using transgenic mice with heart-specific overexpression of Sirt1, a mammalian homolog of Sir2. Sirt1 was significantly upregulated (4- to 8-fold) in response to pressure overload and oxidative stress in nontransgenic adult mouse hearts. Low (2.5-fold) to moderate (7.5-fold) overexpression of Sirt1 in transgenic mouse hearts attenuated age-dependent increases in cardiac hypertrophy, apoptosis/fibrosis, cardiac dysfunction, and expression of senescence markers. In contrast, a high level (12.5-fold) of Sirt1 increased apoptosis and hypertrophy and decreased cardiac function, thereby stimulating the development of cardiomyopathy. Moderate overexpression of Sirt1 protected the heart from oxidative stress induced by paraquat, with increased expression of antioxidants, such as catalase, through forkhead box O (FoxO)-dependent mechanisms, whereas high levels of Sirt1 increased oxidative stress in the heart at baseline. Thus, mild to moderate expression of Sirt1 retards aging of the heart, whereas a high dose of Sirt1 induces cardiomyopathy. Furthermore, although high levels of Sirt1 increase oxidative stress, moderate expression of Sirt1 induces resistance to oxidative stress and apoptosis. These results suggest that Sirt1 could retard aging and confer stress resistance to the heart in vivo, but these beneficial effects can be observed only at low to moderate doses (up to 7.5-fold) of Sirt1.
Collapse
Affiliation(s)
- Ralph R Alcendor
- Cardiovascular Research Institute, Department of Cell Biology and Molecular Medicine, University of Medicine & Dentistry of New Jersey, New Jersey Medical School, Newark, NJ 07103, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Thum T, Hoeber S, Froese S, Klink I, Stichtenoth DO, Galuppo P, Jakob M, Tsikas D, Anker SD, Poole-Wilson PA, Borlak J, Ertl G, Bauersachs J. Age-dependent impairment of endothelial progenitor cells is corrected by growth-hormone-mediated increase of insulin-like growth-factor-1. Circ Res 2007; 100:434-43. [PMID: 17234973 DOI: 10.1161/01.res.0000257912.78915.af] [Citation(s) in RCA: 231] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Aging is associated with an increased risk for atherosclerosis. A possible cause is low numbers and dysfunction of endothelial progenitor cells (EPC) which insufficiently repair damaged vascular walls. We hypothesized that decreased levels of insulin-like growth factor-1 (IGF-1) during age contribute to dysfunctional EPC. We measured the effect of growth hormone (GH), which increases endogenous IGF-1 levels, on EPC in mice and human subjects. We compared EPC number and function in healthy middle-aged male volunteers (57.4+/-1.4 years) before and after a 10 day treatment with recombinant GH (0.4 mg/d) with that of younger and elderly male subjects (27.5+/-0.9 and 74.1+/-0.9 years). Middle-aged and elderly subjects had lower circulating CD133(+)/VEGFR-2(+) EPC with impaired function and increased senescence. GH treatment in middle-aged subjects elevated IGF-1 levels (126.0+/-7.2 ng/mL versus 241.1+/-13.8 ng/mL; P<0.0001), increased circulating EPC with improved colony forming and migratory capacity, enhanced incorporation into tube-like structures, and augmented endothelial nitric oxide synthase expression in EPC comparable to that of the younger group. EPC senescence was attenuated, whereas telomerase activity was increased after GH treatment. Treatment of aged mice with GH (7 days) or IGF-1 increased IGF-1 and EPC levels and improved EPC function, whereas a two day GH treatment did not alter IGF-1 or EPC levels. Ex vivo treatment of EPC from elderly individuals with IGF-1 improved function and attenuated cellular senescence. IGF-1 stimulated EPC differentiation, migratory capacity and the ability to incorporate into forming vascular networks in vitro via the IGF-1 receptor. IGF-1 increased telomerase activity, endothelial nitric oxide synthase expression, phosphorylation and activity in EPC in a phosphoinositide-3-kinase/Akt dependent manner. Small interference RNA-mediated knockdown of endothelial nitric oxide synthase in EPC abolished the IGF-1 effects. Growth hormone-mediated increase in IGF-1 reverses age-related EPC dysfunction and may be a novel therapeutic strategy against vascular disorders with impairment of EPC.
Collapse
Affiliation(s)
- Thomas Thum
- Universität Würzburg, Medizinische Klinik I (Kardiologie), Würzburg, Germany.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Rosenthal N, Santini MP, Musarò A. Growth factor enhancement of cardiac regeneration. Cell Transplant 2006; 15 Suppl 1:S41-5. [PMID: 16826794 DOI: 10.3727/000000006783982287] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
The potential for endogenous or supplementary stem cells to restore the form and function of damaged tissues is particularly promising for overcoming the restricted regenerative capacity of the mammalian heart. To maintain blood circulation, this essential organ needs to launch a rapid response to repair damage of the muscle wall and to prevent muscle loss. The capacity of growth factors to supplement the repair process has been successfully applied to restore the integrity of damaged skeletal muscle, reducing the fibrotic response to injury, and recruiting local populations of self-renewing precursor cells and circulating stem cells. We review the recent evidence that extension of growth factor supplementation to the heart may overcome its inherent regenerative impediments through improvement of the local tissue environment and stimulation of cell replacement, and we speculate on future research directions for treatment of myocardial damage.
Collapse
Affiliation(s)
- Nadia Rosenthal
- Mouse Biology Unit, EMBL-Monterotondo Outstation, Monterotondo (Rome) 00016, Italy.
| | | | | |
Collapse
|