201
|
Circulating factors in young blood as potential therapeutic agents for age-related neurodegenerative and neurovascular diseases. Brain Res Bull 2019; 153:15-23. [PMID: 31400495 DOI: 10.1016/j.brainresbull.2019.08.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 07/30/2019] [Accepted: 08/05/2019] [Indexed: 02/07/2023]
Abstract
Recent animal studies on heterochronic parabiosis (a technique combining the blood circulation of two animals) have revealed that young blood has a powerful rejuvenating effect on brain aging. Circulating factors, especially growth differentiation factor 11 (GDF11) and C-C motif chemokine 11 (CCL11), may play a key role in this effect, which inspires hope for novel approaches to treating age-related cerebral diseases in humans, such as neurodegenerative and neurovascular diseases. Recently, attempts have begun to translate these astonishing and exciting findings from mice to humans and from bench to bedside. However, increasing reports have shown contradictory data, questioning the capacity of these circulating factors to reverse age-related brain dysfunction. In this review, we summarize the current research on the role of young blood, as well as the circulating factors GDF11 and CCL11, in the aging brain and age-related cerebral diseases. We highlight recent controversies, discuss related challenges and provide a future outlook.
Collapse
|
202
|
Pearsall RS, Davies MV, Cannell M, Li J, Widrick J, Mulivor AW, Wallner S, Troy ME, Spaits M, Liharska K, Sako D, Castonguay R, Keates S, Grinberg AV, Suragani RNVS, Kumar R. Follistatin-based ligand trap ACE-083 induces localized hypertrophy of skeletal muscle with functional improvement in models of neuromuscular disease. Sci Rep 2019; 9:11392. [PMID: 31388039 PMCID: PMC6684588 DOI: 10.1038/s41598-019-47818-w] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 07/17/2019] [Indexed: 12/13/2022] Open
Abstract
Skeletal muscle is under inhibitory homeostatic regulation by multiple ligands of the transforming growth factor-β (TGFβ) superfamily. Follistatin is a secreted protein that promotes muscle growth and function by sequestering these ligands extracellularly. In the present study, we evaluated the potential of ACE-083 – a locally acting, follistatin-based fusion protein – as a novel therapeutic agent for focal or asymmetric myopathies. Characterization of ACE-083 in vitro revealed its high affinity for heparin and extracellular matrix while surface plasmon resonance and cell-based assays confirmed that ACE-083 binds and potently neutralizes myostatin, activin A, activin B and growth differentiation factor 11 (GDF11). Intramuscular administration of ACE-083 caused localized, dose-dependent hypertrophy of the injected muscle in wild-type mice and mouse models of Charcot-Marie-Tooth disease (CMT) and Duchenne muscular dystrophy, with no evidence of systemic muscle effects or endocrine perturbation. Importantly, ACE-083 also increased the force of isometric contraction in situ by the injected tibialis anterior muscle in wild-type mice and disease models and increased ankle dorsiflexion torque in CMT mice. Our results demonstrate the potential of ACE-083 as a therapeutic agent for patients with CMT, muscular dystrophy and other disorders with focal or asymmetric muscle atrophy or weakness.
Collapse
Affiliation(s)
| | | | - M Cannell
- Acceleron Pharma, Cambridge, MA, USA
| | - J Li
- Acceleron Pharma, Cambridge, MA, USA
| | - J Widrick
- Division of Genetics and Genomics, The Manton Center for Orphan Disease Research, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - A W Mulivor
- Acceleron Pharma, Cambridge, MA, USA.,The Hospital for Sick Children, Toronto, Ontario, Canada
| | - S Wallner
- Acceleron Pharma, Cambridge, MA, USA.,NovaRock Biotherapeutics, Princeton, NJ, USA
| | - M E Troy
- Acceleron Pharma, Cambridge, MA, USA
| | - M Spaits
- Acceleron Pharma, Cambridge, MA, USA
| | - K Liharska
- Acceleron Pharma, Cambridge, MA, USA.,Dragonfly Therapeutics, Waltham, MA, USA
| | - D Sako
- Acceleron Pharma, Cambridge, MA, USA
| | | | - S Keates
- Acceleron Pharma, Cambridge, MA, USA
| | - A V Grinberg
- Acceleron Pharma, Cambridge, MA, USA.,Dragonfly Therapeutics, Waltham, MA, USA
| | | | - R Kumar
- Acceleron Pharma, Cambridge, MA, USA
| |
Collapse
|
203
|
Sun L, Sun S, Zhao X, Zhang J, Guo J, Tang L, Ta D. Inhibition of myostatin signal pathway may be involved in low-intensity pulsed ultrasound promoting bone healing. J Med Ultrason (2001) 2019; 46:377-388. [PMID: 31377938 DOI: 10.1007/s10396-019-00962-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Accepted: 06/12/2019] [Indexed: 12/28/2022]
Abstract
PURPOSE Low-intensity pulsed ultrasound (LIPUS) is effective in promoting bone healing, and a myostatin deficiency also has a positive effect on bone formation. In this study, we evaluated the effects of LIPUS on bone healing in rats in vivo and investigated the mechanisms in vitro, aiming to explore whether LIPUS promotes bone healing through inhibition of the myostatin signaling pathway. METHODS Rats with both drill-hole defects and MC3T3-E1 cells were randomly assigned to a LIPUS group and a control group. The LIPUS group received LIPUS treatment (1.5 MHz, 30 mW/cm2) for 20 min/day. RESULTS After 21 days, the myostatin expression in quadriceps was significantly inhibited in the LIPUS group, and remodeling of the newly formed bone in the drill-hole site was significantly better in the LIPUS group than that in the control group, which was confirmed by micro-CT analysis. After 3 days, LIPUS significantly promoted osteoblast proliferation; inhibited the expression of AcvrIIB (the myostatin receptor), Smad3, p-Smad3, and GSK-3β; and increased Wnt1 and β-catenin expression. Moreover, translocation of β-catenin from the cytolemma to the nucleus was observed in the LIPUS group. However, these effects were blocked by treatment with myostatin recombinant protein. CONCLUSIONS The results indicate that LIPUS may promote bone healing through inhibition of the myostatin signal pathway.
Collapse
Affiliation(s)
- Lijun Sun
- Institute of Sports Biology, Shaanxi Normal University, Xi'an, 710119, China
| | - Shuxin Sun
- Department of Electronic Engineering, Fudan University, Shanghai, 200433, China
| | - Xinjuan Zhao
- Institute of Sports Biology, Shaanxi Normal University, Xi'an, 710119, China
| | - Jing Zhang
- Institute of Sports Biology, Shaanxi Normal University, Xi'an, 710119, China
| | - Jianzhong Guo
- Shaanxi Key Laboratory of Ultrasonics, Shaanxi Normal University, Xi'an, 710119, China
| | - Liang Tang
- Institute of Sports Biology, Shaanxi Normal University, Xi'an, 710119, China.
| | - Dean Ta
- Department of Electronic Engineering, Fudan University, Shanghai, 200433, China. .,Human Phenome Institute, Fudan University, Shanghai, 201203, China. .,Key Laboratory of Medical Imaging Computing and Computer Assisted Intervention (MICCAI) of Shanghai, Shanghai, 200032, China.
| |
Collapse
|
204
|
Wang MJ, Chen J, Chen F, Liu Q, Sun Y, Yan C, Yang T, Bao Y, Hu YP. Rejuvenating Strategies of Tissue-specific Stem Cells for Healthy Aging. Aging Dis 2019; 10:871-882. [PMID: 31440391 PMCID: PMC6675530 DOI: 10.14336/ad.2018.1119] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Accepted: 11/19/2018] [Indexed: 12/22/2022] Open
Abstract
Although aging is a physiological process, it has raised interest in the science of aging and rejuvenation because of the increasing burden on the rapidly aging global population. With advanced age, there is a decline in homeostatic maintenance and regenerative responsiveness to the injury of various tissues, thereby contributing to the incidence of age-related diseases. The primary cause of the functional declines that occur along with aging is considered to be the exhaustion of stem cell functions in their corresponding tissues. Age-related changes in the systemic environment, the niche, and stem cells contribute to this loss. Thus, the reversal of stem cell aging at the cellular level might lead to the rejuvenation of the animal at an organismic level and the prevention of aging, which would be critical for developing new therapies for age-related dysfunction and diseases. Here, we will explore the effects of aging on stem cells in different tissues. The focus of this discussion is on pro-youth interventions that target intrinsic stem cell properties, environmental niche component, systemic factors, and senescent cellular clearance, which are promising for developing strategies related to the reversal of aged stem cell function and optimizing tissue repair processes.
Collapse
Affiliation(s)
- Min-Jun Wang
- 1Department of Cell Biology, Center for Stem Cell and Medicine, Second Military Medical University, Shanghai 200433, China
| | - Jiajia Chen
- 1Department of Cell Biology, Center for Stem Cell and Medicine, Second Military Medical University, Shanghai 200433, China
| | - Fei Chen
- 1Department of Cell Biology, Center for Stem Cell and Medicine, Second Military Medical University, Shanghai 200433, China
| | - Qinggui Liu
- 1Department of Cell Biology, Center for Stem Cell and Medicine, Second Military Medical University, Shanghai 200433, China
| | - Yu Sun
- 1Department of Cell Biology, Center for Stem Cell and Medicine, Second Military Medical University, Shanghai 200433, China
| | - Chen Yan
- 1Department of Cell Biology, Center for Stem Cell and Medicine, Second Military Medical University, Shanghai 200433, China
| | - Tao Yang
- 1Department of Cell Biology, Center for Stem Cell and Medicine, Second Military Medical University, Shanghai 200433, China
| | - Yiwen Bao
- 1Department of Cell Biology, Center for Stem Cell and Medicine, Second Military Medical University, Shanghai 200433, China.,2Department of Diagnostic radiology, University of Hong Kong, Hong Kong 999077, China
| | - Yi-Ping Hu
- 1Department of Cell Biology, Center for Stem Cell and Medicine, Second Military Medical University, Shanghai 200433, China
| |
Collapse
|
205
|
Lee M, Oikawa S, Ushida T, Suzuki K, Akimoto T. Effects of Exercise Training on Growth and Differentiation Factor 11 Expression in Aged Mice. Front Physiol 2019; 10:970. [PMID: 31417428 PMCID: PMC6684741 DOI: 10.3389/fphys.2019.00970] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Accepted: 07/11/2019] [Indexed: 01/05/2023] Open
Abstract
Exercise training is considered an effective way to prevent age-related skeletal muscle loss. However, the molecular mechanism has not been clarified. Growth and differentiation factor 11 (GDF11) has been controversially considered a regulator of skeletal muscle aging. In this study, we examined whether GDF11 is associated with skeletal muscle aging and the effects of exercise training on age-related skeletal muscle loss. First, we observed that Gdf11 mRNA and protein expression levels in young (5-month-old, n = 6) and aged (22-to 26-month-old, n = 5) mice were not significantly different. Aged mice were then divided into sedentary (n = 5) and exercise (n = 6) groups. The exercise group performed moderate-intensity treadmill running for 6 weeks. Treadmill exercise training increased Gdf11 mRNA expression in the soleus muscle, but its protein expression was not altered. In contrast, the GDF11 level in the plantaris muscle was not changed at either the mRNA or protein level. Collectively, our data demonstrate that GDF11 levels do not change during aging, and that treadmill exercise training increased Gdf11 mRNA expression in a predominantly slow-twitch muscle.
Collapse
Affiliation(s)
- Minjung Lee
- Faculty of Sport Sciences, Waseda University, Saitama, Japan.,Division of Regenerative Medical Engineering, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Bunkyo, Japan
| | - Satoshi Oikawa
- Faculty of Sport Sciences, Waseda University, Saitama, Japan
| | - Takashi Ushida
- Division of Regenerative Medical Engineering, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Bunkyo, Japan
| | | | - Takayuki Akimoto
- Faculty of Sport Sciences, Waseda University, Saitama, Japan.,Division of Regenerative Medical Engineering, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Bunkyo, Japan
| |
Collapse
|
206
|
Wallner C, Huber J, Drysch M, Schmidt SV, Wagner JM, Dadras M, Lehnhardt M, Behr B. Myostatin Upregulation in Patients in the Chronic Phase of Severe Burn Injury Leads to Muscle Cell Catabolism. Eur Surg Res 2019; 60:86-96. [PMID: 31302645 DOI: 10.1159/000500760] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Accepted: 05/05/2019] [Indexed: 11/19/2022]
Abstract
BACKGROUND Burn injury leads to a hypercatabolic response and ultimately muscle wasting with drastic implications for recovery of bodily functions, patient's quality of life (QoL), and long-term survival. Several treatment options target the body's initial stress response, but pharmacological approaches to specifically address muscle protein metabolism have only been poorly investigated. OBJECTIVE The aim of this study was to assess the role of myostatin and follistatin in burn injury and its possible implications in muscle wasting syndrome. METHODS We harvested serum from male patients within 48 h and again 9-12 months after severe burn injury (>20% of total body surface area). By means of myoblast cultures, immunohistochemistry, immunoblotting, and scratch assay, the role of myostatin and its implications in post-burn muscle metabolism and myoblast proliferation and differentiation was analyzed. RESULTS We were able to show increased proliferative and myogenic capacity, decreased myostatin, decreased SMAD 2/3, and elevated follistatin concentrations in human skeletal myoblast cultures with serum conditioned medium of patients in the acute phase of burn injury and conversely a reversed situation in patients in the chronic phase of burn injury. Thus, there is a biphasic response to burn trauma, initiated by an anabolic state and followed by long-term hypercatabolism. CONCLUSION We conclude that the myostatin signaling pathway plays an important regulative role in burn-associated muscle wasting and that blockade of myostatin could prove to be a valuable treatment approach improving the rehabilitation process, QoL, and long-term survival after severe burn injury.
Collapse
Affiliation(s)
- Christoph Wallner
- Department of Plastic Surgery, BG University Hospital Bergmannsheil, Ruhr University Bochum, Bochum, Germany
| | - Julika Huber
- Department of Plastic Surgery, BG University Hospital Bergmannsheil, Ruhr University Bochum, Bochum, Germany
| | - Marius Drysch
- Department of Plastic Surgery, BG University Hospital Bergmannsheil, Ruhr University Bochum, Bochum, Germany
| | - Sonja Verena Schmidt
- Department of Plastic Surgery, BG University Hospital Bergmannsheil, Ruhr University Bochum, Bochum, Germany
| | - Johannes Maximilian Wagner
- Department of Plastic Surgery, BG University Hospital Bergmannsheil, Ruhr University Bochum, Bochum, Germany
| | - Mehran Dadras
- Department of Plastic Surgery, BG University Hospital Bergmannsheil, Ruhr University Bochum, Bochum, Germany
| | - Marcus Lehnhardt
- Department of Plastic Surgery, BG University Hospital Bergmannsheil, Ruhr University Bochum, Bochum, Germany
| | - Björn Behr
- Department of Plastic Surgery, BG University Hospital Bergmannsheil, Ruhr University Bochum, Bochum, Germany,
| |
Collapse
|
207
|
Abstract
Age is the primary risk factor for the vast majority of disorders, including neurodegenerative diseases impacting brain function. Whether the consequences of aging at the biological level can be reversed, or age-related changes prevented, to change the trajectory of such disorders is thus of extreme interest and value. Studies using young plasma, the acellular component of blood, have demonstrated that aging is malleable, with the ability to restore functions in old animals. Fascinatingly, this functional improvement is even observed in the brain, despite the blood-brain barrier, indicating that peripheral sources can effectively impact central sites leading to clinically relevant changes such as enhancement of cognitive function. A plasma-based approach is also attractive as aging is inherently complex, with an array of mechanisms dysregulated in diverse cells and organs throughout the body leading to disturbed function. Plasma, containing a natural mixture of components, has the ability to act multimodally, modulating diverse mechanisms that can converge to change the trajectory of age-related diseases. Here we review the evidence that plasma modulates aging processes in the brain and consider the therapeutic applications that derive from these observations. Plasma and plasma-derived therapeutics are an attractive translation of this concept, requiring critical consideration of benefits, risks, and ethics. Ultimately, knowledge derived from this science will drive a comprehensive molecular understanding to deliver optimized therapeutics. The potential of highly differentiated, multimodal therapeutics for treatment of age-related brain disorders provides an exciting new clinical approach to address the complex etiology of aging.
Collapse
Affiliation(s)
- Viktoria Kheifets
- Alkahest Inc., 125 Shoreway Road, Suite D, San Carlos, CA, 94070, USA
| | | |
Collapse
|
208
|
Garbern J, Kristl AC, Bassaneze V, Vujic A, Schoemaker H, Sereda R, Peng L, Ricci-Blair EM, Goldstein JM, Walker RG, Bhasin S, Wagers AJ, Lee RT. Analysis of Cre-mediated genetic deletion of Gdf11 in cardiomyocytes of young mice. Am J Physiol Heart Circ Physiol 2019; 317:H201-H212. [PMID: 31125255 PMCID: PMC6692736 DOI: 10.1152/ajpheart.00615.2018] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 05/09/2019] [Accepted: 05/10/2019] [Indexed: 01/19/2023]
Abstract
Administration of active growth differentiation factor 11 (GDF11) to aged mice can reduce cardiac hypertrophy, and low serum levels of GDF11 measured together with the related protein, myostatin (also known as GDF8), predict future morbidity and mortality in coronary heart patients. Using mice with a loxP-flanked ("floxed") allele of Gdf11 and Myh6-driven expression of Cre recombinase to delete Gdf11 in cardiomyocytes, we tested the hypothesis that cardiac-specific Gdf11 deficiency might lead to cardiac hypertrophy in young adulthood. We observed that targeted deletion of Gdf11 in cardiomyocytes does not cause cardiac hypertrophy but rather leads to left ventricular dilation when compared with control mice carrying only the Myh6-cre or Gdf11-floxed alleles, suggesting a possible etiology for dilated cardiomyopathy. However, the mechanism underlying this finding remains unclear because of multiple confounding effects associated with the selected model. First, whole heart Gdf11 expression did not decrease in Myh6-cre; Gdf11-floxed mice, possibly because of upregulation of Gdf11 in noncardiomyocytes in the heart. Second, we observed Cre-associated toxicity, with lower body weights and increased global fibrosis, in Cre-only control male mice compared with flox-only controls, making it challenging to infer which changes in Myh6-cre;Gdf11-floxed mice were the result of Cre toxicity versus deletion of Gdf11. Third, we observed differential expression of cre mRNA in Cre-only controls compared with the cardiomyocyte-specific knockout mice, also making comparison between these two groups difficult. Thus, targeted Gdf11 deletion in cardiomyocytes may lead to left ventricular dilation without hypertrophy, but alternative animal models are necessary to understand the mechanism for these findings. NEW & NOTEWORTHY We observed that targeted deletion of growth differentiation factor 11 in cardiomyocytes does not cause cardiac hypertrophy but rather leads to left ventricular dilation compared with control mice carrying only the Myh6-cre or growth differentiation factor 11-floxed alleles. However, the mechanism underlying this finding remains unclear because of multiple confounding effects associated with the selected mouse model.
Collapse
Affiliation(s)
- Jessica Garbern
- Department of Stem Cell and Regenerative Biology and the Harvard Stem Cell Institute, Harvard University , Cambridge, Massachusetts
- Department of Cardiology, Boston Children's Hospital , Boston, Massachusetts
| | - Amy C Kristl
- Department of Stem Cell and Regenerative Biology and the Harvard Stem Cell Institute, Harvard University , Cambridge, Massachusetts
| | - Vinicius Bassaneze
- Department of Stem Cell and Regenerative Biology and the Harvard Stem Cell Institute, Harvard University , Cambridge, Massachusetts
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School , Boston, Massachusetts
| | - Ana Vujic
- Department of Stem Cell and Regenerative Biology and the Harvard Stem Cell Institute, Harvard University , Cambridge, Massachusetts
| | - Henk Schoemaker
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School , Boston, Massachusetts
| | - Rebecca Sereda
- Department of Stem Cell and Regenerative Biology and the Harvard Stem Cell Institute, Harvard University , Cambridge, Massachusetts
| | - Liming Peng
- Brigham Research Assay Core, Brigham and Women's Hospital , Boston, Massachusetts
| | - Elisabeth M Ricci-Blair
- Department of Stem Cell and Regenerative Biology and the Harvard Stem Cell Institute, Harvard University , Cambridge, Massachusetts
| | - Jill M Goldstein
- Department of Stem Cell and Regenerative Biology and the Harvard Stem Cell Institute, Harvard University , Cambridge, Massachusetts
| | - Ryan G Walker
- Department of Stem Cell and Regenerative Biology and the Harvard Stem Cell Institute, Harvard University , Cambridge, Massachusetts
| | - Shalender Bhasin
- Brigham Research Assay Core, Brigham and Women's Hospital , Boston, Massachusetts
| | - Amy J Wagers
- Department of Stem Cell and Regenerative Biology and the Harvard Stem Cell Institute, Harvard University , Cambridge, Massachusetts
- Paul F. Glenn Center for the Biology of Aging, Harvard Medical School , Boston, Massachusetts
- Section on Islet Cell and Regenerative Biology, Joslin Diabetes Center , Boston, Massachusetts
| | - Richard T Lee
- Department of Stem Cell and Regenerative Biology and the Harvard Stem Cell Institute, Harvard University , Cambridge, Massachusetts
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School , Boston, Massachusetts
| |
Collapse
|
209
|
Añón-Hidalgo J, Catalán V, Rodríguez A, Ramírez B, Idoate-Bayón A, Silva C, Mugueta C, Galofré JC, Salvador J, Frühbeck G, Gómez-Ambrosi J. Circulating Concentrations of GDF11 are Positively Associated with TSH Levels in Humans. J Clin Med 2019; 8:jcm8060878. [PMID: 31248139 PMCID: PMC6617068 DOI: 10.3390/jcm8060878] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 06/12/2019] [Accepted: 06/18/2019] [Indexed: 12/16/2022] Open
Abstract
Growth differentiation factor 11 (GDF11) is a member of the transforming growth factor (TGF)-β superfamily which declines with age and has been proposed as an anti-aging factor with regenerative effects in skeletal muscle in mice. However, recent data in humans and mice are conflicting, casting doubts about its true functional actions. The aim of the present study was to analyze the potential involvement of GFD11 in energy homeostasis in particular in relation with thyroid hormones. Serum concentrations of GDF11 were measured by enzyme-linked immunosorbent assay (ELISA) in 287 subjects. A highly significant positive correlation was found between GDF11 and thyroid-stimulating hormone (TSH) concentrations (r = 0.40, p < 0.001). Neither resting energy expenditure (REE) nor REE per unit of fat-free mass (REE/FFM) were significantly correlated (p > 0.05 for both) with GDF11 levels. In a multiple linear regression analysis, the model that best predicted logGDF11 included logTSH, leptin, body mass index (BMI), age, and C-reactive protein (logCRP). This model explained 37% of the total variability of logGDF11 concentrations (p < 0.001), with only logTSH being a significant predictor of logGDF11. After segregating subjects by TSH levels, those within the low TSH group exhibited significantly decreased (p < 0.05) GDF11 concentrations as compared to the normal TSH group or the high TSH group. A significant correlation of GDF11 levels with logCRP (r = 0.19, p = 0.025) was found. GDF11 levels were not related to the presence of hypertension or cardiopathy. In conclusion, our results show that circulating concentrations of GDF11 are closely associated with TSH concentrations and reduced in subjects with low TSH levels. However, GDF11 is not related to the regulation of energy expenditure. Our data also suggest that GDF11 may be involved in the regulation of inflammation, without relation to cardiac function. Further research is needed to elucidate the role of GDF11 in metabolism and its potential involvement in thyroid pathophysiology.
Collapse
Affiliation(s)
- Juan Añón-Hidalgo
- Metabolic Research Laboratory, Clínica Universidad de Navarra, 31008 Pamplona, Spain.
| | - Victoria Catalán
- Metabolic Research Laboratory, Clínica Universidad de Navarra, 31008 Pamplona, Spain.
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, 31008 Pamplona, Spain.
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain.
| | - Amaia Rodríguez
- Metabolic Research Laboratory, Clínica Universidad de Navarra, 31008 Pamplona, Spain.
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, 31008 Pamplona, Spain.
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain.
| | - Beatriz Ramírez
- Metabolic Research Laboratory, Clínica Universidad de Navarra, 31008 Pamplona, Spain.
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, 31008 Pamplona, Spain.
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain.
| | - Adrián Idoate-Bayón
- Metabolic Research Laboratory, Clínica Universidad de Navarra, 31008 Pamplona, Spain.
| | - Camilo Silva
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, 31008 Pamplona, Spain.
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain.
- Department of Endocrinology and Nutrition, Clínica Universidad de Navarra, 31008 Pamplona, Spain.
| | - Carmen Mugueta
- Department of Biochemistry, Clínica Universidad de Navarra, 31008 Pamplona, Spain.
| | - Juan C Galofré
- Department of Endocrinology and Nutrition, Clínica Universidad de Navarra, 31008 Pamplona, Spain.
| | - Javier Salvador
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, 31008 Pamplona, Spain.
- Department of Endocrinology and Nutrition, Clínica Universidad de Navarra, 31008 Pamplona, Spain.
| | - Gema Frühbeck
- Metabolic Research Laboratory, Clínica Universidad de Navarra, 31008 Pamplona, Spain.
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, 31008 Pamplona, Spain.
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain.
- Department of Endocrinology and Nutrition, Clínica Universidad de Navarra, 31008 Pamplona, Spain.
| | - Javier Gómez-Ambrosi
- Metabolic Research Laboratory, Clínica Universidad de Navarra, 31008 Pamplona, Spain.
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, 31008 Pamplona, Spain.
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain.
| |
Collapse
|
210
|
Growth differentiation factor 11 (GDF11) has pronounced effects on skin biology. PLoS One 2019; 14:e0218035. [PMID: 31181098 PMCID: PMC6557520 DOI: 10.1371/journal.pone.0218035] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Accepted: 05/23/2019] [Indexed: 12/22/2022] Open
Abstract
Growth differentiation factor 11 (GDF11) belongs to the TGF-β superfamily of proteins and is closely related to myostatin. Recent findings show that GDF11 has rejuvenating properties with pronounced effects on the cardiovascular system, brain, skeletal muscle, and skeleton in mice. Several human studies were also conducted, some implicating decreasing levels of circulating GDF11 with age. To date, however, there have not been any reports on its role in human skin. This study examined the impact of GDF11 on human skin, specifically related to skin aging. The effect of recombinant GDF11 on the function of various skin cells was examined in human epidermal keratinocytes, dermal fibroblasts, melanocytes, dermal microvascular endothelial cells and 3D skin equivalents, as well as in ex vivo human skin explants. GDF11 had significant effects on the production of dermal matrix components in multiple skin models in vitro and ex vivo. In addition, it had a pronounced effect on expression of multiple skin related genes in full thickness 3D skin equivalents. This work, for the first time, demonstrates an important role for GDF11 in skin biology and a potential impact on skin health and aging.
Collapse
|
211
|
Gerardo-Ramírez M, Lazzarini-Lechuga R, Hernández-Rizo S, Jiménez-Salazar JE, Simoni-Nieves A, García-Ruiz C, Fernández-Checa JC, Marquardt JU, Coulouarn C, Gutiérrez-Ruiz MC, Pérez-Aguilar B, Gomez-Quiroz LE. GDF11 exhibits tumor suppressive properties in hepatocellular carcinoma cells by restricting clonal expansion and invasion. Biochim Biophys Acta Mol Basis Dis 2019; 1865:1540-1554. [PMID: 30890427 DOI: 10.1016/j.bbadis.2019.03.003] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 02/26/2019] [Accepted: 03/14/2019] [Indexed: 02/06/2023]
Abstract
Growth differentiation factor 11 (GDF11) has been characterized as a key regulator of differentiation in cells that retain stemness features, despite some controversies in age-related studies. GDF11 has been poorly investigated in cancer, particularly in those with stemness capacity, such as hepatocellular carcinoma (HCC), one of the most aggressive cancers worldwide. Here, we focused on investigating the effects of GDF11 in liver cancer cells. GDF11 treatment significantly reduced proliferation, colony and spheroid formation in HCC cell lines. Consistently, down-regulation of CDK6, cyclin D1, cyclin A, and concomitant upregulation of p27 was observed after 24 h of treatment. Interestingly, cell viability was unchanged, but cell functionality was compromised. These effects were potentially induced by the expression of E-cadherin and occludin, as well as Snail and N-cadherin repression, in a time-dependent manner. Furthermore, GDF11 treatment for 72 h induced that cells were incapable of sustaining colony and sphere capacity in the absent of GDF11, up to 5 days, indicating that the effect of GDF11 on self-renewal capacity is not transient. Finally, in vivo invasion studies revealed a significant decrease in cell migration of hepatocellular carcinoma cells treated with GDF11 associated to a decreased proliferation judged by Ki67 staining. Data show that exogenous GDF11 displays tumor suppressor properties in HCC cells.
Collapse
MESH Headings
- Animals
- Antigens, CD/genetics
- Antigens, CD/metabolism
- Bone Morphogenetic Proteins/genetics
- Bone Morphogenetic Proteins/metabolism
- Bone Morphogenetic Proteins/pharmacology
- Cadherins/genetics
- Cadherins/metabolism
- Cell Differentiation/drug effects
- Cell Line, Tumor
- Cell Movement/drug effects
- Cell Proliferation/drug effects
- Chick Embryo
- Chorioallantoic Membrane/blood supply
- Chorioallantoic Membrane/drug effects
- Cyclin A/genetics
- Cyclin A/metabolism
- Cyclin D1/genetics
- Cyclin D1/metabolism
- Cyclin-Dependent Kinase 6/genetics
- Cyclin-Dependent Kinase 6/metabolism
- Cyclin-Dependent Kinase Inhibitor p27/genetics
- Cyclin-Dependent Kinase Inhibitor p27/metabolism
- Gene Expression Regulation, Neoplastic
- Growth Differentiation Factors/genetics
- Growth Differentiation Factors/metabolism
- Growth Differentiation Factors/pharmacology
- Hep G2 Cells
- Humans
- Neovascularization, Pathologic/genetics
- Neovascularization, Pathologic/metabolism
- Neovascularization, Pathologic/pathology
- Neovascularization, Pathologic/prevention & control
- Occludin/genetics
- Occludin/metabolism
- Signal Transduction
- Snail Family Transcription Factors/genetics
- Snail Family Transcription Factors/metabolism
- Spheroids, Cellular/drug effects
- Spheroids, Cellular/metabolism
- Spheroids, Cellular/pathology
Collapse
Affiliation(s)
- Monserrat Gerardo-Ramírez
- Posgrado en Biología Experimental, DCBS, Universidad Autónoma Metropolitana-Iztapalapa, Mexico City, Mexico; Departamento de Ciencias de la Salud, Universidad Autónoma Metropolitana-Iztapalapa, Mexico City, Mexico
| | - Roberto Lazzarini-Lechuga
- Departamento de Biología de la Reproducción, Universidad Autónoma Metropolitana-Iztapalapa, Mexico City, Mexico
| | - Sharik Hernández-Rizo
- Posgrado en Biología Experimental, DCBS, Universidad Autónoma Metropolitana-Iztapalapa, Mexico City, Mexico; Departamento de Ciencias de la Salud, Universidad Autónoma Metropolitana-Iztapalapa, Mexico City, Mexico
| | | | - Arturo Simoni-Nieves
- Posgrado en Biología Experimental, DCBS, Universidad Autónoma Metropolitana-Iztapalapa, Mexico City, Mexico; Departamento de Ciencias de la Salud, Universidad Autónoma Metropolitana-Iztapalapa, Mexico City, Mexico
| | - Carmen García-Ruiz
- Department of Cell Death and Proliferation, Instituto de Investigaciones Biomédicas de Barcelona, CSIC, Liver Unit, Hospital Clinic, IDIBPAS and CIBERehd, Barcelona, Spain
| | - José Carlos Fernández-Checa
- Department of Cell Death and Proliferation, Instituto de Investigaciones Biomédicas de Barcelona, CSIC, Liver Unit, Hospital Clinic, IDIBPAS and CIBERehd, Barcelona, Spain
| | - Jens U Marquardt
- 1st Department of Medicine, University Medical Center, Johannes Gutenberg University Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany
| | - Cedric Coulouarn
- INSERM, Inra, University of Rennes, UMR 1241, Nutrition Metabolisms and Cancer, Rennes, France
| | - María Concepción Gutiérrez-Ruiz
- Departamento de Ciencias de la Salud, Universidad Autónoma Metropolitana-Iztapalapa, Mexico City, Mexico; Laboratorio de Medicina Experimental, Unidad de Medicina Translacional, Instituto de Investigaciones Biomédicas, UNAM/Instituto Nacional de Cardiología Ignacio Chavez, Mexico City, Mexico
| | - Benjamín Pérez-Aguilar
- Departamento de Ciencias de la Salud, Universidad Autónoma Metropolitana-Iztapalapa, Mexico City, Mexico
| | - Luis E Gomez-Quiroz
- Departamento de Ciencias de la Salud, Universidad Autónoma Metropolitana-Iztapalapa, Mexico City, Mexico; Laboratorio de Medicina Experimental, Unidad de Medicina Translacional, Instituto de Investigaciones Biomédicas, UNAM/Instituto Nacional de Cardiología Ignacio Chavez, Mexico City, Mexico.
| |
Collapse
|
212
|
Zhang H, Cherian R, Jin K. Systemic milieu and age-related deterioration. GeroScience 2019; 41:275-284. [PMID: 31152364 DOI: 10.1007/s11357-019-00075-1] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Accepted: 05/21/2019] [Indexed: 01/11/2023] Open
Abstract
Aging is a fundamental biological process accompanied by a general decline in tissue function and an increased risk for age-related disease. The risk for cardiovascular, stroke, cancer, and neurodegenerative diseases significantly increases with aging, especially in people aged 60 years and older in the USA. Although the cellular and molecular mechanisms underlying aging and age-related disease are beginning to be unraveled, the role of the systemic milieu remains unknown. Recent studies have shown that systemic factors in young blood can revise age-related impairments and extend organismal lifespan, suggesting that the systemic milieu contains pro-aging and rejuvenating factors that play a critical role in the health and aging phenotype. In this review, we summarize the current knowledge of systemic milieu changes during the aging process and its link to age-related deterioration.
Collapse
Affiliation(s)
- Hongxia Zhang
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX, 76107, USA
| | - Ryan Cherian
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX, 76107, USA
| | - Kunlin Jin
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX, 76107, USA.
| |
Collapse
|
213
|
Egerman MA, Glass DJ. The role of GDF11 in aging and skeletal muscle, cardiac and bone homeostasis. Crit Rev Biochem Mol Biol 2019; 54:174-183. [DOI: 10.1080/10409238.2019.1610722] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Marc A. Egerman
- Age-Related Disorders, Department of Chemical Biology and Therapeutics, Novartis Institutes for Biomedical Research, Cambridge, MA, USA
| | - David J. Glass
- Age-Related Disorders, Department of Chemical Biology and Therapeutics, Novartis Institutes for Biomedical Research, Cambridge, MA, USA
| |
Collapse
|
214
|
Jang YJ, Ahn J, Son HJ, Jung CH, Ahn J, Ha TY. Hydrangea serrata
Tea Enhances Running Endurance and Skeletal Muscle Mass. Mol Nutr Food Res 2019; 63:e1801149. [DOI: 10.1002/mnfr.201801149] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Revised: 04/29/2019] [Indexed: 11/11/2022]
Affiliation(s)
- Young Jin Jang
- Research Group of Natural Materials and MetabolismKorea Food Research Institute Wanju‐gun 55365 Republic of Korea
| | - Jisong Ahn
- Research Group of Natural Materials and MetabolismKorea Food Research Institute Wanju‐gun 55365 Republic of Korea
- Department of Food Science and TechnologyChonbuk National University Jeonju‐si 54896 Republic of Korea
| | - Hyo Jeong Son
- Research Group of Natural Materials and MetabolismKorea Food Research Institute Wanju‐gun 55365 Republic of Korea
| | - Chang Hwa Jung
- Research Group of Natural Materials and MetabolismKorea Food Research Institute Wanju‐gun 55365 Republic of Korea
- Division of Food BiotechnologyUniversity of Science and Technology Daejeon 34113 Republic of Korea
| | - Jiyun Ahn
- Research Group of Natural Materials and MetabolismKorea Food Research Institute Wanju‐gun 55365 Republic of Korea
- Division of Food BiotechnologyUniversity of Science and Technology Daejeon 34113 Republic of Korea
| | - Tae Youl Ha
- Research Group of Natural Materials and MetabolismKorea Food Research Institute Wanju‐gun 55365 Republic of Korea
- Division of Food BiotechnologyUniversity of Science and Technology Daejeon 34113 Republic of Korea
| |
Collapse
|
215
|
Jin Q, Qiao C, Li J, Xiao B, Li J, Xiao X. A GDF11/myostatin inhibitor, GDF11 propeptide-Fc, increases skeletal muscle mass and improves muscle strength in dystrophic mdx mice. Skelet Muscle 2019; 9:16. [PMID: 31133057 PMCID: PMC6537384 DOI: 10.1186/s13395-019-0197-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 04/10/2019] [Indexed: 01/27/2023] Open
Abstract
Background Growth differentiation factor 11 (GDF11) is a member of the transforming growth factor β superfamily. The GDF11 propeptide, which is derived from the GDF11 precursor protein, blocks the activity of GDF11 and its homolog, myostatin, which are both potent inhibitors of muscle growth. Thus, treatment with GDF11 propeptide may be a potential therapeutic strategy for diseases associated with muscle atrophy like sarcopenia and the muscular dystrophies. Here, we evaluate the impact of GDF11 propeptide-Fc (GDF11PRO-Fc) gene delivery on skeletal muscle in normal and dystrophic adult mice. Methods A pull-down assay was used to obtain physical confirmation of a protein-protein interaction between GDF11PRO-Fc and GDF11 or myostatin. Next, differentiated C2C12 myotubes were treated with AAV6-GDF11PRO-Fc and challenged with GDF11 or myostatin to determine if GDF11PRO-Fc could block GDF11/myostatin-induced myotube atrophy. Localized expression of GDF11PRO-Fc was evaluated via a unilateral intramuscular injection of AAV9-GDF11PRO-Fc into the hindlimb of C57BL/6J mice. In mdx mice, intravenous injection of AAV9-GDF11PRO-Fc was used to achieve systemic expression. The impact of GDF11PRO-Fc on muscle mass, function, and pathological features were assessed. Results GDF11PRO-Fc was observed to bind both GDF11 and myostatin. In C2C12 myotubes, expression of GDF11PRO-Fc was able to mitigate GDF11/myostatin-induced atrophy. Following intramuscular injection in C57BL/6J mice, increased grip strength and localized muscle hypertrophy were observed in the injected hindlimb after 10 weeks. In mdx mice, systemic expression of GDF11PRO-Fc resulted in skeletal muscle hypertrophy without a significant change in cardiac mass after 12 weeks. In addition, grip strength and rotarod latency time were improved. Intramuscular fibrosis was also reduced in treated mdx mice; however, there was no change seen in central nucleation, membrane permeability to serum IgG or serum creatine kinase levels. Conclusions GDF11PRO-Fc induces skeletal muscle hypertrophy and improvements in muscle strength via inhibition of GDF11/myostatin signaling. However, GDF11PRO-Fc does not significantly improve the dystrophic pathology in mdx mice. Electronic supplementary material The online version of this article (10.1186/s13395-019-0197-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Quan Jin
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, USA.
| | - Chunping Qiao
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, USA
| | - Jianbin Li
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, USA
| | - Bin Xiao
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, USA
| | - Juan Li
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, USA
| | - Xiao Xiao
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, USA
| |
Collapse
|
216
|
Takahara Y, Miyachi N, Nawa M, Matsuoka M. Calmodulin‐like skin protein suppresses the increase in senescence‐associated β‐galactosidase induced by hydrogen peroxide or ultraviolet irradiation in keratinocytes. Cell Biol Int 2019; 43:835-843. [DOI: 10.1002/cbin.11159] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Accepted: 04/28/2019] [Indexed: 11/11/2022]
Affiliation(s)
- Yusuke Takahara
- Department of PharmacologyTokyo Medical University 6‐1‐1 Shinjuku Shinjuku‐ku Tokyo 160‐8402 Japan
- Department of Dermatological NeuroscienceTokyo Medical University6‐1‐1 Shinjuku Shinjuku‐ku Tokyo 160‐8402 Japan
- Groupwide Research and Development Tokyo Research Laboratory Noevir Co., Ltd., C‐333 R&D KSP 3‐2‐1 Sakado Takatsuku, Kawasaki Kanagawa Japan
| | - Nobuyuki Miyachi
- Department of Dermatological NeuroscienceTokyo Medical University6‐1‐1 Shinjuku Shinjuku‐ku Tokyo 160‐8402 Japan
- Groupwide Research and Development Tokyo Research Laboratory Noevir Co., Ltd., C‐333 R&D KSP 3‐2‐1 Sakado Takatsuku, Kawasaki Kanagawa Japan
| | - Mikiro Nawa
- Department of PharmacologyTokyo Medical University 6‐1‐1 Shinjuku Shinjuku‐ku Tokyo 160‐8402 Japan
| | - Masaaki Matsuoka
- Department of PharmacologyTokyo Medical University 6‐1‐1 Shinjuku Shinjuku‐ku Tokyo 160‐8402 Japan
- Department of Dermatological NeuroscienceTokyo Medical University6‐1‐1 Shinjuku Shinjuku‐ku Tokyo 160‐8402 Japan
| |
Collapse
|
217
|
Piccirillo R. Exercise-Induced Myokines With Therapeutic Potential for Muscle Wasting. Front Physiol 2019; 10:287. [PMID: 30984014 PMCID: PMC6449478 DOI: 10.3389/fphys.2019.00287] [Citation(s) in RCA: 88] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 03/04/2019] [Indexed: 12/26/2022] Open
Abstract
Skeletal muscle is a highly vascularized tissue that can secrete proteins called myokines. These muscle-secreted factors exert biological functions in muscle itself (autocrine effect) or on short- or long-distant organs (paracrine/endocrine effects) and control processes such as metabolism, angiogenesis, or inflammation. Widely differing diseases ranging from genetic myopathies to cancers are emerging as causing dysregulated secretion of myokines from skeletal muscles. Myokines are also involved in the control of muscle size and may be important to be restored to normal levels to alleviate muscle wasting in various conditions, such as cancer, untreated diabetes, chronic obstructive pulmonary disease, aging, or heart failure. Interestingly, many myokines are induced by exercise (muscle-derived exerkines) and some even by specific types of physical activity, but more studies are needed on this issue. Most exercise-induced myokines travel throughout the body by means of extracellular vesicles. Restoring myokines by physical activity may be added to the list of mechanisms by which exercise exerts preventative or curative effects against a large number of diseases, including the deleterious muscle wasting they may cause. Extending our understanding about which myokines could be usefully restored in certain diseases might help in prescribing more tailored exercise or myokine-based drugs.
Collapse
Affiliation(s)
- Rosanna Piccirillo
- Department of Neurosciences, Mario Negri Institute for Pharmacological Research IRCCS, Milan, Italy
| |
Collapse
|
218
|
Naro F, Venturelli M, Monaco L, Toniolo L, Muti E, Milanese C, Zhao J, Richardson RS, Schena F, Reggiani C. Skeletal Muscle Fiber Size and Gene Expression in the Oldest-Old With Differing Degrees of Mobility. Front Physiol 2019; 10:313. [PMID: 30971947 PMCID: PMC6443969 DOI: 10.3389/fphys.2019.00313] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Accepted: 03/07/2019] [Indexed: 12/22/2022] Open
Abstract
The oldest-old, in the ninth and tenth decades of their life, represent a population characterized by neuromuscular impairment, which often implies a loss of mobility and independence. As recently documented by us and others, muscle atrophy and weakness are accompanied by an unexpected preservation of the size and contractile function of skeletal muscle fibers. This suggests that, while most fibers are likely lost with their respective motoneurons, the surviving fibers are well preserved. Here, we investigated the mechanisms behind this fiber preservation and the relevance of physical activity, by comparing a group of 6 young healthy controls (YG: 22-28 years) with two groups of oldest-old (81-96 years), one able to walk (OW: n = 6, average 86 years) and one confined to a wheelchair (ONW n = 9, average 88 years). We confirmed previous results of fiber preservation and, additionally, observed a shift in fiber type, toward slow predominance in OW and fast predominance in ONW. Myonuclear density was increased in muscles of ONW, compared to YG and OW, potentially indicative of an ongoing atrophy process. We analyzed, by RT-qPCR, the expression of genes relevant for fiber size and type regulation in a biopsy sample from the vastus lateralis. In all oldest-old both myostatin and IGF-1 expression were attenuated compared to YG, however, in ONW two specific IGF-1 isoforms, IGF-1EA and MGF, demonstrated a further significant decrease compared to OW. Surprisingly, atrogenes (MURF1 and atrogin) expression was also significantly reduced compared to YG and this was accompanied by a close to statistically significantly attenuated marker of autophagy, LC3. Among the determinants of the metabolic fiber type, PGC1α was significantly reduced in both OW and ONW compared to YG, while AMPK was down-regulated only in ONW. We conclude that, in contrast to the shift of the balance in favor of pro-atrophy factors found by other studies in older adults (decreased IGF-1, increase of myostatin, increase of atrogenes), in the oldest-old the pro-atrophy factors also appear to be down-regulated, allowing a partial recovery of the proteostasis balance. Furthermore, the impact of muscle activity, as a consequence of lost or preserved walking ability, is limited.
Collapse
Affiliation(s)
- Fabio Naro
- Department of Anatomical, Histological, Forensic and Orthopedic Sciences, Sapienza University of Rome, Rome, Italy
| | - Massimo Venturelli
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Lucia Monaco
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy
| | - Luana Toniolo
- Department of Biomedical Sciences, University of Padova, Padua, Italy
| | - Ettore Muti
- Monsignor Arrigo Mazzali Foundation, Mantova, Italy
| | - Chiara Milanese
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Jia Zhao
- Division of Geriatrics, Department of Internal Medicine, The University of Utah, Salt Lake City, UT, United States.,Department of Nutrition and Integrative Physiology, The University of Utah, Salt Lake City, UT, United States.,Geriatric Research, Education, and Clinical Center, George E. Whalen VA Medical Center, Salt Lake City, UT, United States
| | - Russell S Richardson
- Division of Geriatrics, Department of Internal Medicine, The University of Utah, Salt Lake City, UT, United States.,Department of Nutrition and Integrative Physiology, The University of Utah, Salt Lake City, UT, United States.,Geriatric Research, Education, and Clinical Center, George E. Whalen VA Medical Center, Salt Lake City, UT, United States
| | - Federico Schena
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Carlo Reggiani
- Department of Biomedical Sciences, University of Padova, Padua, Italy.,Institute for Kinesiology Research, Science and Research Center of Koper, Koper, Slovenia
| |
Collapse
|
219
|
Zhang XJ, Tan H, Shi ZF, Li N, Jia Y, Hao Z. Growth differentiation factor 11 is involved in isoproterenol‑induced heart failure. Mol Med Rep 2019; 19:4109-4118. [PMID: 30942402 PMCID: PMC6471622 DOI: 10.3892/mmr.2019.10077] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Accepted: 02/02/2019] [Indexed: 12/14/2022] Open
Abstract
The present study aimed to investigate the potential effects of growth differentiation factor 11 (GDF11) on isoproterenol (ISO)-induced heart failure (HF) and identify the underlying molecular mechanisms. A rat model of HF was induced in vivo by intraperitoneally administering ISO (5 mg/kg/day) for 7 days. After 4 weeks following establishment of the HF model, hemodynamic analysis demonstrated that ISO induced a significant increase in the left ventricular end-diastolic pressure and a decrease in the left ventricular systolic pressure and maximum contraction velocity. The plasma levels of myocardial injury markers, including lactate dehydrogenase (LDH), creatine kinase (CK), CK-muscle/brain which were determined using the corresponding assay kits and plasma brain natriuretic peptide which was detected by an ELISA kit, an important biomarker of HF, increased following ISO treatment. Furthermore, levels of GDF11 expression and protein, which were estimated using reverse transcription-quantitative polymerase chain reaction and an ELISA kit in plasma and western blotting in the heart tissue, respectively, significantly increased following ISO treatment. To demonstrate the effects of ISO on GDF11 production in cardiomyocytes, H9C2 cells (a cardiomyoblast cell line derived from embryonic rat heart tissue) were treated with ISO (50 nM) for 24 h in vitro; it was revealed that GDF11 protein and mRNA expression levels significantly increased following ISO treatment. In addition, recombinant GDF11 (rGDF11) administered to ISO-treated H9C2 cells resulted in decreased proliferation, which was detected via a CCK-8 assay, and increased LDH levels and cell apoptosis of cells, which was determined using Caspase-3 activity and Hoechst 33258 staining. Additionally, rGDF11 increased the levels of reactive oxygen species and malondialdehyde due to the upregulation of nicotinamide adenine dinucleotide phosphate oxidase 4 (Nox4) following rGDF11 treatment. Conversely, GDF11 knockdown reduced ISO-induced apoptosis by inhibiting oxidative stress injury. The results suggested that GDF11 production was upregulated in ISO-induced rats with HF and in ISO-treated H9C2 cells, and that rGDF11 treatment increased ISO-induced oxidative stress injury by upregulating Nox4 in H9C2 cells.
Collapse
Affiliation(s)
- Xiu-Jing Zhang
- The First Department of Cadres Health Care, The Third Hospital of Shijiazhuang, Shijiazhuang, Hebei 050011, P.R. China
| | - Hua Tan
- The First Department of Cadres Health Care, The Third Hospital of Shijiazhuang, Shijiazhuang, Hebei 050011, P.R. China
| | - Zhi-Fang Shi
- The Second Department of Cadres Health Care, The Third Hospital of Shijiazhuang, Shijiazhuang, Hebei 050011, P.R. China
| | - Na Li
- The First Department of Cadres Health Care, The Third Hospital of Shijiazhuang, Shijiazhuang, Hebei 050011, P.R. China
| | - Ying Jia
- The First Department of Cadres Health Care, The Third Hospital of Shijiazhuang, Shijiazhuang, Hebei 050011, P.R. China
| | - Zhe Hao
- The First Department of Cadres Health Care, The Third Hospital of Shijiazhuang, Shijiazhuang, Hebei 050011, P.R. China
| |
Collapse
|
220
|
Roh JD, Hobson R, Chaudhari V, Quintero P, Yeri A, Benson M, Xiao C, Zlotoff D, Bezzerides V, Houstis N, Platt C, Damilano F, Lindman BR, Elmariah S, Biersmith M, Lee SJ, Seidman CE, Seidman JG, Gerszten RE, Lach-Trifilieff E, Glass DJ, Rosenzweig A. Activin type II receptor signaling in cardiac aging and heart failure. Sci Transl Med 2019; 11:eaau8680. [PMID: 30842316 PMCID: PMC7124007 DOI: 10.1126/scitranslmed.aau8680] [Citation(s) in RCA: 112] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 02/15/2019] [Indexed: 01/14/2023]
Abstract
Activin type II receptor (ActRII) ligands have been implicated in muscle wasting in aging and disease. However, the role of these ligands and ActRII signaling in the heart remains unclear. Here, we investigated this catabolic pathway in human aging and heart failure (HF) using circulating follistatin-like 3 (FSTL3) as a potential indicator of systemic ActRII activity. FSTL3 is a downstream regulator of ActRII signaling, whose expression is up-regulated by the major ActRII ligands, activin A, circulating growth differentiation factor-8 (GDF8), and GDF11. In humans, we found that circulating FSTL3 increased with aging, frailty, and HF severity, correlating with an increase in circulating activins. In mice, increasing circulating activin A increased cardiac ActRII signaling and FSTL3 expression, as well as impaired cardiac function. Conversely, ActRII blockade with either clinical-stage inhibitors or genetic ablation reduced cardiac ActRII signaling while restoring or preserving cardiac function in multiple models of HF induced by aging, sarcomere mutation, or pressure overload. Using unbiased RNA sequencing, we show that activin A, GDF8, and GDF11 all induce a similar pathologic profile associated with up-regulation of the proteasome pathway in mammalian cardiomyocytes. The E3 ubiquitin ligase, Smurf1, was identified as a key downstream effector of activin-mediated ActRII signaling, which increased proteasome-dependent degradation of sarcoplasmic reticulum Ca2+ ATPase (SERCA2a), a critical determinant of cardiomyocyte function. Together, our findings suggest that increased activin/ActRII signaling links aging and HF pathobiology and that targeted inhibition of this catabolic pathway holds promise as a therapeutic strategy for multiple forms of HF.
Collapse
Affiliation(s)
- Jason D Roh
- Corrigan Minehan Heart Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Ryan Hobson
- Corrigan Minehan Heart Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Vinita Chaudhari
- Corrigan Minehan Heart Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Pablo Quintero
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | - Ashish Yeri
- Corrigan Minehan Heart Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Mark Benson
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | - Chunyang Xiao
- Corrigan Minehan Heart Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Daniel Zlotoff
- Corrigan Minehan Heart Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Vassilios Bezzerides
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Nicholas Houstis
- Corrigan Minehan Heart Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Colin Platt
- Corrigan Minehan Heart Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Federico Damilano
- Corrigan Minehan Heart Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Brian R Lindman
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN 37203, USA
| | - Sammy Elmariah
- Corrigan Minehan Heart Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Michael Biersmith
- Division of Cardiovascular Medicine, Wexner Medical Center, Ohio State University, Columbus, OH 43210, USA
| | - Se-Jin Lee
- The Jackson Laboratory, Farmington, CT 06032, USA
- Department of Genetics and Genome Sciences, University of Connecticut School of Medicine, Farmington, CT 06032, USA
| | - Christine E Seidman
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
- Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Boston, MA 02114, USA
| | | | - Robert E Gerszten
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | | | - David J Glass
- Novartis Institutes for Biomedical Research, Cambridge, MA 02139, USA
| | - Anthony Rosenzweig
- Corrigan Minehan Heart Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA.
| |
Collapse
|
221
|
Castonguay R, Lachey J, Wallner S, Strand J, Liharska K, Watanabe AE, Cannell M, Davies MV, Sako D, Troy ME, Krishnan L, Mulivor AW, Li H, Keates S, Alexander MJ, Pearsall RS, Kumar R. Follistatin-288-Fc Fusion Protein Promotes Localized Growth of Skeletal Muscle. J Pharmacol Exp Ther 2019; 368:435-445. [PMID: 30563942 DOI: 10.1124/jpet.118.252304] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 12/06/2018] [Indexed: 12/19/2022] Open
Abstract
Follistatin is an endogenous glycoprotein that promotes growth and repair of skeletal muscle by sequestering inhibitory ligands of the transforming growth factor-β superfamily and may therefore have therapeutic potential for neuromuscular diseases. Here, we sought to determine the suitability of a newly engineered follistatin fusion protein (FST288-Fc) to promote localized, rather than systemic, growth of skeletal muscle by capitalizing on the intrinsic heparin-binding ability of the follistatin-288 isoform. As determined by surface plasmon resonance and cell-based assays, FST288-Fc binds to activin A, activin B, myostatin (growth differentiation factor GDF8), and GDF11 with high affinity and neutralizes their activity in vitro. Intramuscular administration of FST288-Fc in mice induced robust, dose-dependent growth of the targeted muscle but not of surrounding or contralateral muscles, in contrast to the systemic effects of a locally administered fusion protein incorporating activin receptor type IIB (ActRIIB-Fc). Furthermore, systemic administration of FST288-Fc in mice did not alter muscle mass or body composition as determined by NMR, which again contrasts with the pronounced systemic activity of ActRIIB-Fc when administered by the same route. Subsequent analysis revealed that FST288-Fc in the circulation undergoes rapid proteolysis, thereby restricting its activity to individual muscles targeted by intramuscular administration. These results indicate that FST288-Fc can produce localized growth of skeletal muscle in a targeted manner with reduced potential for undesirable systemic effects. Thus, FST288-Fc and similar agents may be beneficial in the treatment of disorders with muscle atrophy that is focal, asymmetric, or otherwise heterogeneous.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | - Huiming Li
- Acceleron Pharma, Cambridge, Massachusetts
| | | | | | | | - Ravi Kumar
- Acceleron Pharma, Cambridge, Massachusetts
| |
Collapse
|
222
|
Dong X, Han Y, Abeysekera IR, Shao Z, Wang H. GDF11 is increased in patients with aplastic anemia. ACTA ACUST UNITED AC 2019; 24:331-336. [PMID: 30727851 DOI: 10.1080/16078454.2019.1574386] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
OBJECT To explore the critical role of growth differentiation factor 11 (GDF11) in the pathobiology of aplastic anemia (AA). METHODS We have examined the serum GDF11 levels for 79 AA patients and 30 healthy controls. A total of 79 AA patients, which included 29 new diagnosed (untreated) cases, 14 cases with no response, 21 partial remission (PR) cases and 15 complete remission (CR) cases after immunosuppressive therapy (IST). GDF11 serum levels were assessed by an enzyme-linked immunosorbent assay. GDF11 mRNA expression in peripheral blood mononuclear (PBMNC) was detected through real time polymerase chain reaction. The correlation between GDF11 expression and erythropoietic function was evaluated. RESULTS The serum GDF11 levels in untreated AA patients were higher than that of the control group. The serum GDF11 levels of PR patients or CR patients after IST was decreased, compared with untreated patients, but did not recover back to the normal levels. GDF11 levels had a negative correlation with hemoglobin (Hb) levels and reticulocyte counts in AA patients. GDF11 levels did not correlate with age, sex and severity of in AA patients. CONCLUSION Serum GDF11 levels were increased and negatively correlated with Hb levels and reticulocyte counts in AA patients. This suggests an impaired GDF11 response contributing to anemia in AA patients.
Collapse
Affiliation(s)
- Xifeng Dong
- a Department of Hematology, General Hospital , Tianjin Medical University , Tianjin , People's Republic of China
| | - Yu Han
- b Department of Hematology , Huaihe Hospital of Henan University , Kaifeng , People's Republic of China
| | - Iruni Roshanie Abeysekera
- c Department of Physiology and Pathophysiology, School of Basic Medical Sciences , Tianjin Medical University , Tianjin , People's Republic of China
| | - Zonghong Shao
- a Department of Hematology, General Hospital , Tianjin Medical University , Tianjin , People's Republic of China
| | - Huaquan Wang
- a Department of Hematology, General Hospital , Tianjin Medical University , Tianjin , People's Republic of China
| |
Collapse
|
223
|
Faber MS, Whitehead TA. Data-driven engineering of protein therapeutics. Curr Opin Biotechnol 2019; 60:104-110. [PMID: 30822697 DOI: 10.1016/j.copbio.2019.01.015] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 11/16/2018] [Accepted: 01/21/2019] [Indexed: 12/26/2022]
Abstract
Protein therapeutics requires a series of properties beyond biochemical activity, including serum stability, low immunogenicity, and manufacturability. Mutations that improve one property often decrease one or more of the other essential requirements for therapeutic efficacy, making the protein engineering challenge difficult. The past decade has seen an explosion of new techniques centered around cheaply reading and writing DNA. This review highlights the recent use of such high throughput technologies for engineering protein therapeutics. Examples include the use of human antibody repertoire sequence data to pair antibody heavy and light chains, comprehensive mutational analysis for engineering antibody specificity, and the use of ancestral and inter-species sequence data to engineer simultaneous improvements in enzyme catalytic efficiency and stability. We conclude with a perspective on further ways to integrate mature protein engineering pipelines with the exponential increases in the volume of sequencing data expected in the forthcoming decade.
Collapse
Affiliation(s)
- Matthew S Faber
- Dept. Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI 48824, United States
| | - Timothy A Whitehead
- Dept. of Chemical Engineering & Materials Science, Michigan State University, East Lansing, MI 48824, United States; Dept. of Biosystems Engineering, Michigan State University, East Lansing, MI 48824, United States; Dept. of Biomedical Engineering, Michigan State University, East Lansing, MI 48824, United States; Institute for Quantitative Biology, Michigan State University, East Lansing, MI 48824, United States.
| |
Collapse
|
224
|
The influence of GDF11 on brain fate and function. GeroScience 2019; 41:1-11. [PMID: 30729414 DOI: 10.1007/s11357-019-00054-6] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Accepted: 01/18/2019] [Indexed: 10/27/2022] Open
Abstract
Growth differentiation factor 11 (GDF11) is a transforming growth factor β (TGFβ) protein that regulates aspects of central nervous system (CNS) formation and health throughout the lifespan. During development, GDF11 influences CNS patterning and the genesis, differentiation, maturation, and activity of new cells, which may be primarily dependent on local production and action. In the aged brain, exogenous, peripherally delivered GDF11 may enhance neurogenesis and angiogenesis, as well as improve neuropathological outcomes. This is in contrast to a predominantly negative influence on neurogenesis in the developing CNS. Seemingly antithetical effects may correspond to the cell types and mechanisms activated by local versus circulating concentrations of GDF11. Yet undefined, distinct mechanisms of action in young and aged brains may also play a role, which could include differential receptor and binding partner interactions. Exogenously increasing circulating GDF11 concentrations may be a viable approach for improving deleterious aspects of brain aging and neuropathology. Caution is warranted, however, since GDF11 appears to negatively influence muscle health and body composition. Nevertheless, an expanding understanding of GDF11 biology suggests that it is an important regulator of CNS formation and fate, and its manipulation may improve aspects of brain health in older organisms.
Collapse
|
225
|
Administration of rGDF11 retards the aging process in male mice via action of anti-oxidant system. Biogerontology 2019; 20:433-443. [PMID: 30726519 DOI: 10.1007/s10522-019-09799-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Accepted: 01/29/2019] [Indexed: 10/27/2022]
Abstract
One of the most studied and widely accepted conjectures of aging process is the oxidative stress theory. Current studies have generated disputes on the effects of GDF11 and GDF8, a closely related member of GDF11, on rejuvenation and anti-aging properties. In this study, we first demonstrated that when recombinant GDF8 (rGDF8) and GDF11 (rGDF11) of the fish Nothobranchius guentheri were injected into 20-month-old male mice, their serum GDF8 and GDF11 levels were clearly increased. We also showed that injection of rGDF8 and rGDF11 had little influences on the body weight and serological parameters of the mice, indicating their general condition and physiology were not affected. Based on these findings, we started to test the effects of administration of piscine rGDF11 and rGDF8 on the aging process of male mice and to explore the underlying mechanisms. It was found that rGDF11 was able to reduce the levels of AGEs, protein oxidation and lipid peroxidation, and to slow down the accumulation of age-related histological markers, while rGDF8 was not. Moreover, rGDF11 significantly prevented the decrease in CAT, GPX and SOD activities, but rGDF8 did not. Collectively, these results suggest that it is GDF11 but not GDF8 that can exert rejuvenation and anti-aging activities via the action of antioxidant system. It is also the first report that shows the activity of GDF11 is not species-specific, implicating potential usefulness of piscine GDF11 in prolonging the lifespan of the elderly.
Collapse
|
226
|
Yuan X, Bhat OM, Lohner H, Li N, Zhang Y, Li PL. Inhibitory effects of growth differentiation factor 11 on autophagy deficiency-induced dedifferentiation of arterial smooth muscle cells. Am J Physiol Heart Circ Physiol 2019; 316:H345-H356. [PMID: 30462553 PMCID: PMC6397385 DOI: 10.1152/ajpheart.00342.2018] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Revised: 10/24/2018] [Accepted: 11/15/2018] [Indexed: 12/27/2022]
Abstract
Growth differentiation factor (GDF)11 has been reported to reverse age-related cardiac hypertrophy in mice and cause youthful regeneration of cardiomyocytes. The present study attempted to test a hypothesis that GDF11 counteracts the pathologic dedifferentiation of mouse carotid arterial smooth muscle cells (CASMCs) due to deficient autophagy. By real-time RT-PCR and Western blot analysis, exogenously administrated GDF11 was found to promote CASMC differentiation with increased expression of various differentiation markers (α-smooth muscle actin, myogenin, myogenic differentiation, and myosin heavy chain) as well as decreased expression of dedifferentiation markers (vimentin and proliferating cell nuclear antigen). Upregulation of the GDF11 gene by trichostatin A (TSA) or CRISPR-cas9 activating plasmids also stimulated the differentiation of CASMCs. Either GDF11 or TSA treatment blocked 7-ketocholesterol-induced CASMC dedifferentiation and autophagosome accumulation as well as lysosome inhibitor bafilomycin-induced dedifferentiation and autophagosome accumulation. Moreover, in CASMCs from mice lacking the CD38 gene, an autophagy deficiency model in CASMCs, GDF11 also inhibited its phenotypic transition to dedifferentiation status. Correspondingly, TSA treatment was shown to decrease GDF11 expression and reverse CASMC dedifferentiation in the partial ligated carotid artery of mice. The inhibitory effects of TSA on dedifferentiation of CASMCs were accompanied by reduced autophagosome accumulation in the arterial wall, which was accompanied by attenuated neointima formation in partial ligated carotid arteries. We concluded that GDF11 promotes CASMC differentiation and prevents the phenotypic transition of these cells induced by autophagosome accumulation during different pathological stimulations, such as Western diet, lysosome function deficiency, and inflammation. NEW & NOTEWORTHY The present study demonstrates that growth differentiation factor (GDF)11 promotes autophagy and subsequent differentiation in carotid arterial smooth muscle cells. Upregulation of GDF11 counteracts dedifferentiation under different pathological conditions. These findings provide novel insights into the regulatory role of GDF11 in the counteracting of sclerotic arterial diseases and also suggest that activation or induction of GDF11 may be a new therapeutic strategy for the treatment or prevention of these diseases.
Collapse
Affiliation(s)
- Xinxu Yuan
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University , Richmond, Virginia
| | - Owais M Bhat
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University , Richmond, Virginia
| | - Hannah Lohner
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University , Richmond, Virginia
| | - Ningjun Li
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University , Richmond, Virginia
| | - Yang Zhang
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston , Houston, Texas
| | - Pin-Lan Li
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University , Richmond, Virginia
| |
Collapse
|
227
|
Magga J, Vainio L, Kilpiö T, Hulmi JJ, Taponen S, Lin R, Räsänen M, Szabó Z, Gao E, Rahtu-Korpela L, Alakoski T, Ulvila J, Laitinen M, Pasternack A, Koch WJ, Alitalo K, Kivelä R, Ritvos O, Kerkelä R. Systemic Blockade of ACVR2B Ligands Protects Myocardium from Acute Ischemia-Reperfusion Injury. Mol Ther 2019; 27:600-610. [PMID: 30765322 PMCID: PMC6404100 DOI: 10.1016/j.ymthe.2019.01.013] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Revised: 01/16/2019] [Accepted: 01/16/2019] [Indexed: 02/07/2023] Open
Abstract
Activin A and myostatin, members of the transforming growth factor (TGF)-β superfamily of secreted factors, are potent negative regulators of muscle growth, but their contribution to myocardial ischemia-reperfusion (IR) injury is not known. The aim of this study was to investigate if activin 2B (ACVR2B) receptor ligands contribute to myocardial IR injury. Mice were treated with soluble ACVR2B decoy receptor (ACVR2B-Fc) and subjected to myocardial ischemia followed by reperfusion for 6 or 24 h. Systemic blockade of ACVR2B ligands by ACVR2B-Fc was protective against cardiac IR injury, as evidenced by reduced infarcted area, apoptosis, and autophagy and better preserved LV systolic function following IR. ACVR2B-Fc modified cardiac metabolism, LV mitochondrial respiration, as well as cardiac phenotype toward physiological hypertrophy. Similar to its protective role in IR injury in vivo, ACVR2B-Fc antagonized SMAD2 signaling and cell death in cardiomyocytes that were subjected to hypoxic stress. ACVR2B ligand myostatin was found to exacerbate hypoxic stress. In addition to acute cardioprotection in ischemia, ACVR2B-Fc provided beneficial effects on cardiac function in prolonged cardiac stress in cardiotoxicity model. By blocking myostatin, ACVR2B-Fc potentially reduces cardiomyocyte death and modifies cardiomyocyte metabolism for hypoxic conditions to protect the heart from IR injury.
Collapse
Affiliation(s)
- Johanna Magga
- Research Unit of Biomedicine, Department of Pharmacology and Toxicology, University of Oulu, 90220 Oulu, Finland; Biocenter Oulu, University of Oulu, 90220 Oulu, Finland.
| | - Laura Vainio
- Research Unit of Biomedicine, Department of Pharmacology and Toxicology, University of Oulu, 90220 Oulu, Finland
| | - Teemu Kilpiö
- Research Unit of Biomedicine, Department of Pharmacology and Toxicology, University of Oulu, 90220 Oulu, Finland
| | - Juha J Hulmi
- Neuromuscular Research Center, Biology of Physical Activity, Faculty of Sport and Health Sciences, University of Jyväskylä, 40014 Jyväskylä, Finland; Department of Physiology, Faculty of Medicine, University of Helsinki, 00290 Helsinki, Finland
| | - Saija Taponen
- Research Unit of Biomedicine, Department of Pharmacology and Toxicology, University of Oulu, 90220 Oulu, Finland
| | - Ruizhu Lin
- Research Unit of Biomedicine, Department of Pharmacology and Toxicology, University of Oulu, 90220 Oulu, Finland; Medical Research Center Oulu, Oulu University Hospital and University of Oulu, 90220 Oulu, Finland
| | - Markus Räsänen
- Wihuri Research Institute and Translational Cancer Biology Program, Faculty of Medicine, University of Helsinki, 00290 Helsinki, Finland
| | - Zoltán Szabó
- Research Unit of Biomedicine, Department of Pharmacology and Toxicology, University of Oulu, 90220 Oulu, Finland
| | - Erhe Gao
- Center for Translational Medicine, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | - Lea Rahtu-Korpela
- Research Unit of Biomedicine, Department of Pharmacology and Toxicology, University of Oulu, 90220 Oulu, Finland
| | - Tarja Alakoski
- Research Unit of Biomedicine, Department of Pharmacology and Toxicology, University of Oulu, 90220 Oulu, Finland
| | - Johanna Ulvila
- Research Unit of Biomedicine, Department of Pharmacology and Toxicology, University of Oulu, 90220 Oulu, Finland
| | - Mika Laitinen
- Department of Medicine, University of Helsinki, 00029 Helsinki, Finland; Department of Medicine, Helsinki University Hospital, 00029 Helsinki, Finland
| | - Arja Pasternack
- Department of Physiology, Faculty of Medicine, University of Helsinki, 00290 Helsinki, Finland
| | - Walter J Koch
- Center for Translational Medicine, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | - Kari Alitalo
- Wihuri Research Institute and Translational Cancer Biology Program, Faculty of Medicine, University of Helsinki, 00290 Helsinki, Finland
| | - Riikka Kivelä
- Wihuri Research Institute and Translational Cancer Biology Program, Faculty of Medicine, University of Helsinki, 00290 Helsinki, Finland
| | - Olli Ritvos
- Department of Physiology, Faculty of Medicine, University of Helsinki, 00290 Helsinki, Finland
| | - Risto Kerkelä
- Research Unit of Biomedicine, Department of Pharmacology and Toxicology, University of Oulu, 90220 Oulu, Finland; Medical Research Center Oulu, Oulu University Hospital and University of Oulu, 90220 Oulu, Finland
| |
Collapse
|
228
|
Opstad TB, Kalstad AA, Pettersen AÅ, Arnesen H, Seljeflot I. Novel biomolecules of ageing, sex differences and potential underlying mechanisms of telomere shortening in coronary artery disease. Exp Gerontol 2019; 119:53-60. [PMID: 30684534 DOI: 10.1016/j.exger.2019.01.020] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Revised: 01/08/2019] [Accepted: 01/21/2019] [Indexed: 10/27/2022]
Abstract
Telomere length (TL), growth differentiate factor (GDF)11, insulin growth factor (IGF)1, sirtuin (SIRT)1 and inflammatory processes have been related to ageing and age-related diseases, like coronary artery disease (CAD). We aimed to investigate the associations between leukocyte TLs (LTLs), chronological age, sex and comorbidities in CAD patients. Any covariations between LTL, GDF11, IGF1, SIRT-1 and pro-inflammatory cytokines were further assessed. METHODS In 300 patients with stable CAD (age 36-81 years, 20% females), DNA and RNA were isolated from whole blood for PCR analysis and relative quantification of LTLs and gene-expression of GDF11, IGF1,SIRT1, IL-12, IL-18 and IFNƴ, respectively. Serum was prepared for the analyses of circulating IL-18, IL-12, IL-6 and TNFα. RESULTS Patients with previous myocardial infarction (MI) presented with 20% shorter LTLs vs. patients without (p = 0.019) indicating LTLs to be of importance for CAD severity. The observation however, was only observed in men (p = 0.009, n = 115), in which the upper LTL quartile associated with 64% lower frequency of previous MI compared to quartile 1-3 (p = 0.005, adjusted). LTLs were not differently distributed according to sex or comorbidities such as hypertension, diabetes type 2 and metabolic syndrome. LTLs and GDF11 were inversely correlated to age (r = -0.17; p = 0.007 and r = -0.16; p = 0.010, respectively), however, separated in gender, LTL only in women (r = -0.37) and GDF11 only in men (r = -0.19) (p = 0.006, both). GDF11 and SIRT1 were strongly inter-correlated (r = 0.56, p ≤ 0.001), suggesting common upstream regulators. LTLs were moderately correlated to GDF11 and SIRT1 in overweight women (BMI ≥ 25 kg/m2) (r = 0.41; p = 0.027 and 0.43; p = 0.020, respectively), which may reflect common life-style influences on LTLs and these markers. In all women, we observed further that the highest LTL quartile associated with higher GDF11 and SIRT expression and lower circulating levels of IL-12, IL-18 and TNFα, as compared to quartile 1, which may indicate lifestyle influences on female LTLs. In men, the highest LTL quartile associated with lower IFNƴ expression and lower circulating TNFα. Overall, the results indicate an association between chronic low-grade inflammation and LTLs. CONCLUSIONS Shorter LTLs in CAD patients with previously suffered MI may indicate telomere attrition as part of its pathophysiology in men. The inverse association between LTLs and age exclusively in women underpins the previously reported decline in attrition rate in men with increasing age. As elevated GDF11 and SIRT1 along with attenuated pro-inflammatory cytokines seem to positively affect LTL in women, we hypothesize a potential sex-dimorphism in LTL regulation, which may implicate sex- adjusted health-preventive therapies.
Collapse
Affiliation(s)
- Trine B Opstad
- Center for Clinical Heart Research, Department of Cardiology, Oslo University Hospital, Ullevål, Norway; Center for Heart Failure Research, Oslo University Hospital, Norway; Faculty of Medicine, University of Oslo, Norway.
| | - Are A Kalstad
- Center for Clinical Heart Research, Department of Cardiology, Oslo University Hospital, Ullevål, Norway; Center for Heart Failure Research, Oslo University Hospital, Norway; Faculty of Medicine, University of Oslo, Norway
| | - Alf Åge Pettersen
- Center for Clinical Heart Research, Department of Cardiology, Oslo University Hospital, Ullevål, Norway; Center for Heart Failure Research, Oslo University Hospital, Norway; Ringerike Hospital, Vestre Viken HF, Norway
| | - Harald Arnesen
- Center for Clinical Heart Research, Department of Cardiology, Oslo University Hospital, Ullevål, Norway; Center for Heart Failure Research, Oslo University Hospital, Norway; Faculty of Medicine, University of Oslo, Norway
| | - Ingebjørg Seljeflot
- Center for Clinical Heart Research, Department of Cardiology, Oslo University Hospital, Ullevål, Norway; Center for Heart Failure Research, Oslo University Hospital, Norway; Faculty of Medicine, University of Oslo, Norway
| |
Collapse
|
229
|
The Growth Differentiation Factor 11 is Involved in Skin Fibroblast Ageing and is Induced by a Preparation of Peptides and Sugars Derived from Plant Cell Cultures. Mol Biotechnol 2019; 61:209-220. [DOI: 10.1007/s12033-019-00154-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
230
|
Lau A, Kennedy BK, Kirkland JL, Tullius SG. Mixing old and young: enhancing rejuvenation and accelerating aging. J Clin Invest 2019; 129:4-11. [PMID: 30601138 DOI: 10.1172/jci123946] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Donor age and recipient age are factors that influence transplantation outcomes. Aside from age-associated differences in intrinsic graft function and alloimmune responses, the ability of young and old cells to exert either rejuvenating or aging effects extrinsically may also apply to the transplantation of hematopoietic stem cells or solid organ transplants. While the potential for rejuvenation mediated by the transfer of youthful cells is currently being explored for therapeutic applications, aspects that relate to accelerating aging are no less clinically significant. Those effects may be particularly relevant in transplantation with an age discrepancy between donor and recipient. Here, we review recent advances in understanding the mechanisms by which young and old cells modify their environments to promote rejuvenation- or aging-associated phenotypes. We discuss their relevance to clinical transplantation and highlight potential opportunities for therapeutic intervention.
Collapse
Affiliation(s)
- Ashley Lau
- Division of Transplant Surgery, Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Brian K Kennedy
- Departments of Biochemistry and Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Singapore Institute for Clinical Sciences, Singapore.,Agency for Science, Technology and Research (A*STAR), Singapore.,Buck Institute for Research on Aging, Novato, California, USA
| | - James L Kirkland
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, Minnesota, USA
| | - Stefan G Tullius
- Division of Transplant Surgery, Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
231
|
Mahmoudi S, Xu L, Brunet A. Turning back time with emerging rejuvenation strategies. Nat Cell Biol 2019; 21:32-43. [PMID: 30602763 DOI: 10.1038/s41556-018-0206-0] [Citation(s) in RCA: 111] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Accepted: 08/24/2018] [Indexed: 01/10/2023]
Abstract
Ageing is associated with the functional decline of all tissues and a striking increase in many diseases. Although ageing has long been considered a one-way street, strategies to delay and potentially even reverse the ageing process have recently been developed. Here, we review four emerging rejuvenation strategies-systemic factors, metabolic manipulations, senescent cell ablation and cellular reprogramming-and discuss their mechanisms of action, cellular targets, potential trade-offs and application to human ageing.
Collapse
Affiliation(s)
- Salah Mahmoudi
- Department of Genetics, Stanford University, Stanford, CA, USA
| | - Lucy Xu
- Department of Genetics, Stanford University, Stanford, CA, USA.,Department of Biology, Stanford University, Stanford, CA, USA
| | - Anne Brunet
- Department of Genetics, Stanford University, Stanford, CA, USA. .,Glenn Laboratories for the Biology of Aging, Stanford University, Stanford, CA, USA.
| |
Collapse
|
232
|
Larsson L, Degens H, Li M, Salviati L, Lee YI, Thompson W, Kirkland JL, Sandri M. Sarcopenia: Aging-Related Loss of Muscle Mass and Function. Physiol Rev 2019; 99:427-511. [PMID: 30427277 PMCID: PMC6442923 DOI: 10.1152/physrev.00061.2017] [Citation(s) in RCA: 962] [Impact Index Per Article: 160.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Revised: 05/14/2018] [Accepted: 06/15/2018] [Indexed: 12/11/2022] Open
Abstract
Sarcopenia is a loss of muscle mass and function in the elderly that reduces mobility, diminishes quality of life, and can lead to fall-related injuries, which require costly hospitalization and extended rehabilitation. This review focuses on the aging-related structural changes and mechanisms at cellular and subcellular levels underlying changes in the individual motor unit: specifically, the perikaryon of the α-motoneuron, its neuromuscular junction(s), and the muscle fibers that it innervates. Loss of muscle mass with aging, which is largely due to the progressive loss of motoneurons, is associated with reduced muscle fiber number and size. Muscle function progressively declines because motoneuron loss is not adequately compensated by reinnervation of muscle fibers by the remaining motoneurons. At the intracellular level, key factors are qualitative changes in posttranslational modifications of muscle proteins and the loss of coordinated control between contractile, mitochondrial, and sarcoplasmic reticulum protein expression. Quantitative and qualitative changes in skeletal muscle during the process of aging also have been implicated in the pathogenesis of acquired and hereditary neuromuscular disorders. In experimental models, specific intervention strategies have shown encouraging results on limiting deterioration of motor unit structure and function under conditions of impaired innervation. Translated to the clinic, if these or similar interventions, by saving muscle and improving mobility, could help alleviate sarcopenia in the elderly, there would be both great humanitarian benefits and large cost savings for health care systems.
Collapse
Affiliation(s)
- Lars Larsson
- Department of Physiology and Pharmacology, Basic and Clinical Muscle Biology Group, Karolinska Institutet , Stockholm , Sweden ; Section of Clinical Neurophysiology, Department of Clinical Neuroscience, Karolinska Institutet , Stockholm , Sweden ; Department of Biobehavioral Health, The Pennsylvania State University , University Park, Pennsylvania ; School of Healthcare Science, Metropolitan University , Manchester , United Kingdom ; Institute of Sport Science and Innovations, Lithuanian Sports University , Kaunas , Lithuania ; Clinical Genetics Unit, Department of Woman and Child Health, University of Padova , Padova , Italy ; IRP Città della Speranza, Padova , Italy ; Department of Biology, Texas A&M University , College Station, Texas ; Robert and Arlene Kogod Center on Aging, Mayo Clinic , Rochester, Minnesota ; Department of Biomedical Science, Venetian Institute of Molecular Medicine, University of Padova , Padova , Italy
| | - Hans Degens
- Department of Physiology and Pharmacology, Basic and Clinical Muscle Biology Group, Karolinska Institutet , Stockholm , Sweden ; Section of Clinical Neurophysiology, Department of Clinical Neuroscience, Karolinska Institutet , Stockholm , Sweden ; Department of Biobehavioral Health, The Pennsylvania State University , University Park, Pennsylvania ; School of Healthcare Science, Metropolitan University , Manchester , United Kingdom ; Institute of Sport Science and Innovations, Lithuanian Sports University , Kaunas , Lithuania ; Clinical Genetics Unit, Department of Woman and Child Health, University of Padova , Padova , Italy ; IRP Città della Speranza, Padova , Italy ; Department of Biology, Texas A&M University , College Station, Texas ; Robert and Arlene Kogod Center on Aging, Mayo Clinic , Rochester, Minnesota ; Department of Biomedical Science, Venetian Institute of Molecular Medicine, University of Padova , Padova , Italy
| | - Meishan Li
- Department of Physiology and Pharmacology, Basic and Clinical Muscle Biology Group, Karolinska Institutet , Stockholm , Sweden ; Section of Clinical Neurophysiology, Department of Clinical Neuroscience, Karolinska Institutet , Stockholm , Sweden ; Department of Biobehavioral Health, The Pennsylvania State University , University Park, Pennsylvania ; School of Healthcare Science, Metropolitan University , Manchester , United Kingdom ; Institute of Sport Science and Innovations, Lithuanian Sports University , Kaunas , Lithuania ; Clinical Genetics Unit, Department of Woman and Child Health, University of Padova , Padova , Italy ; IRP Città della Speranza, Padova , Italy ; Department of Biology, Texas A&M University , College Station, Texas ; Robert and Arlene Kogod Center on Aging, Mayo Clinic , Rochester, Minnesota ; Department of Biomedical Science, Venetian Institute of Molecular Medicine, University of Padova , Padova , Italy
| | - Leonardo Salviati
- Department of Physiology and Pharmacology, Basic and Clinical Muscle Biology Group, Karolinska Institutet , Stockholm , Sweden ; Section of Clinical Neurophysiology, Department of Clinical Neuroscience, Karolinska Institutet , Stockholm , Sweden ; Department of Biobehavioral Health, The Pennsylvania State University , University Park, Pennsylvania ; School of Healthcare Science, Metropolitan University , Manchester , United Kingdom ; Institute of Sport Science and Innovations, Lithuanian Sports University , Kaunas , Lithuania ; Clinical Genetics Unit, Department of Woman and Child Health, University of Padova , Padova , Italy ; IRP Città della Speranza, Padova , Italy ; Department of Biology, Texas A&M University , College Station, Texas ; Robert and Arlene Kogod Center on Aging, Mayo Clinic , Rochester, Minnesota ; Department of Biomedical Science, Venetian Institute of Molecular Medicine, University of Padova , Padova , Italy
| | - Young Il Lee
- Department of Physiology and Pharmacology, Basic and Clinical Muscle Biology Group, Karolinska Institutet , Stockholm , Sweden ; Section of Clinical Neurophysiology, Department of Clinical Neuroscience, Karolinska Institutet , Stockholm , Sweden ; Department of Biobehavioral Health, The Pennsylvania State University , University Park, Pennsylvania ; School of Healthcare Science, Metropolitan University , Manchester , United Kingdom ; Institute of Sport Science and Innovations, Lithuanian Sports University , Kaunas , Lithuania ; Clinical Genetics Unit, Department of Woman and Child Health, University of Padova , Padova , Italy ; IRP Città della Speranza, Padova , Italy ; Department of Biology, Texas A&M University , College Station, Texas ; Robert and Arlene Kogod Center on Aging, Mayo Clinic , Rochester, Minnesota ; Department of Biomedical Science, Venetian Institute of Molecular Medicine, University of Padova , Padova , Italy
| | - Wesley Thompson
- Department of Physiology and Pharmacology, Basic and Clinical Muscle Biology Group, Karolinska Institutet , Stockholm , Sweden ; Section of Clinical Neurophysiology, Department of Clinical Neuroscience, Karolinska Institutet , Stockholm , Sweden ; Department of Biobehavioral Health, The Pennsylvania State University , University Park, Pennsylvania ; School of Healthcare Science, Metropolitan University , Manchester , United Kingdom ; Institute of Sport Science and Innovations, Lithuanian Sports University , Kaunas , Lithuania ; Clinical Genetics Unit, Department of Woman and Child Health, University of Padova , Padova , Italy ; IRP Città della Speranza, Padova , Italy ; Department of Biology, Texas A&M University , College Station, Texas ; Robert and Arlene Kogod Center on Aging, Mayo Clinic , Rochester, Minnesota ; Department of Biomedical Science, Venetian Institute of Molecular Medicine, University of Padova , Padova , Italy
| | - James L Kirkland
- Department of Physiology and Pharmacology, Basic and Clinical Muscle Biology Group, Karolinska Institutet , Stockholm , Sweden ; Section of Clinical Neurophysiology, Department of Clinical Neuroscience, Karolinska Institutet , Stockholm , Sweden ; Department of Biobehavioral Health, The Pennsylvania State University , University Park, Pennsylvania ; School of Healthcare Science, Metropolitan University , Manchester , United Kingdom ; Institute of Sport Science and Innovations, Lithuanian Sports University , Kaunas , Lithuania ; Clinical Genetics Unit, Department of Woman and Child Health, University of Padova , Padova , Italy ; IRP Città della Speranza, Padova , Italy ; Department of Biology, Texas A&M University , College Station, Texas ; Robert and Arlene Kogod Center on Aging, Mayo Clinic , Rochester, Minnesota ; Department of Biomedical Science, Venetian Institute of Molecular Medicine, University of Padova , Padova , Italy
| | - Marco Sandri
- Department of Physiology and Pharmacology, Basic and Clinical Muscle Biology Group, Karolinska Institutet , Stockholm , Sweden ; Section of Clinical Neurophysiology, Department of Clinical Neuroscience, Karolinska Institutet , Stockholm , Sweden ; Department of Biobehavioral Health, The Pennsylvania State University , University Park, Pennsylvania ; School of Healthcare Science, Metropolitan University , Manchester , United Kingdom ; Institute of Sport Science and Innovations, Lithuanian Sports University , Kaunas , Lithuania ; Clinical Genetics Unit, Department of Woman and Child Health, University of Padova , Padova , Italy ; IRP Città della Speranza, Padova , Italy ; Department of Biology, Texas A&M University , College Station, Texas ; Robert and Arlene Kogod Center on Aging, Mayo Clinic , Rochester, Minnesota ; Department of Biomedical Science, Venetian Institute of Molecular Medicine, University of Padova , Padova , Italy
| |
Collapse
|
233
|
Galkin F, Zhang B, Dmitriev SE, Gladyshev VN. Reversibility of irreversible aging. Ageing Res Rev 2019; 49:104-114. [PMID: 30513346 DOI: 10.1016/j.arr.2018.11.008] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2018] [Revised: 11/14/2018] [Accepted: 11/30/2018] [Indexed: 12/18/2022]
Abstract
Most multicellular organisms are known to age, due to accumulation of damage and other deleterious changes over time. These changes are often irreversible, as organisms, humans included, evolved fully differentiated, irreplaceable cells (e.g. neurons) and structures (e.g. skeleton). Hence, deterioration or loss of at least some cells and structures should lead to inevitable aging of these organisms. Yet, some cells may escape this fate: adult somatic cells may be converted to partially reprogrammed cells or induced pluripotent stem cells (iPSCs). By their nature, iPSCs are the cells representing the early stages of life, indicating a possibility of reversing the age of cells within the organism. Reprogramming strategies may be accomplished both in vitro and in vivo, offering opportunities for rejuvenation in the context of whole organisms. Similarly, older organs may be replaced with the younger ones prepared ex vivo, or grown within other organisms or even other species. How could the irreversibility of aging of some parts of the organism be reconciled with the putative reversal of aging of the other parts of the same organism? Resolution of this question holds promise for dramatically extending lifespan, which is currently not possible with traditional genetic, dietary and pharmacological approaches. Critical issues in this challenge are the nature of aging, relationship between aging of an organism and aging of its parts, relationship between cell dedifferentiation and rejuvenation, and increased risk of cancer that goes hand in hand with rejuvenation approaches.
Collapse
Affiliation(s)
- Fedor Galkin
- Belozersky Institute of Physico-Chemical Biology, Moscow State University, Moscow, 119234, Russia; Insilico Medicine, Rockville, Maryland 20850, United States
| | - Bohan Zhang
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Sergey E Dmitriev
- Belozersky Institute of Physico-Chemical Biology, Moscow State University, Moscow, 119234, Russia
| | - Vadim N Gladyshev
- Belozersky Institute of Physico-Chemical Biology, Moscow State University, Moscow, 119234, Russia; Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
234
|
Wu Y, Qu J, Li H, Yuan H, Guo Q, Ouyang Z, Lu Q. Relationship between serum level of growth differentiation factors 8, 11 and bone mineral density in girls with anorexia nervosa. Clin Endocrinol (Oxf) 2019; 90:88-93. [PMID: 30281844 DOI: 10.1111/cen.13871] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Revised: 08/30/2018] [Accepted: 09/21/2018] [Indexed: 12/13/2022]
Abstract
BACKGROUND Adolescents with anorexia nervosa (AN) have low body mass and low bone mineral density (BMD). Growth differentiation factor 8 (Myostatin, GDF8) and its homologue growth differentiation factor 11 (GDF11), members of the TGF-β super-family, play an important role in muscle regeneration and bone metabolism in healthy individuals. However, their association with BMD in AN is unknown. The present study was undertaken to investigate the relationship between GDF8, GDF11 and BMD in adolescent girls with AN. METHODS Serum GDF8, GDF11 and BMD were determined in 25 girls (12-16 years old) with AN and 31 healthy girls (12-16 years old). RESULTS Growth differentiation factor 8 levels were lower in AN subjects. On the contrary, GDF11 levels were higher in AN subjects than controls. There was no relationship between GDF8 and BMD. A significant negative correlation between GDF11 and BMD was found. In multiple linear stepwise regression analysis, BMI, 25-hydroxyvitamin D, GDF11, or lean mass, but not fat mass and GDF8, were independent predictors of BMD in the AN and control groups separately. CONCLUSIONS Growth differentiation factor 11 was independent predictor of BMD in girls with AN. It suggested that GDF11 exerts a negative effect on bone mass.
Collapse
Affiliation(s)
- Yali Wu
- Department of Pharmacy, The Second Xiangya Hospital of Central South University, Changsha, China
- Department of Science and Education, Hunan Provincial Maternal and Child Health Care Hospital, Changsha, China
| | - Jian Qu
- Department of Pharmacy, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Huabing Li
- Department of Radiology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Haiyan Yuan
- Department of Pharmacy, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Qi Guo
- Endocrinology Research Center, The Xiangya Hospital of Central South University, Changsha, China
| | - Zhanbo Ouyang
- Department of Pharmacy, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Qiong Lu
- Department of Pharmacy, The Second Xiangya Hospital of Central South University, Changsha, China
| |
Collapse
|
235
|
Kemp PR, Griffiths M, Polkey MI. Muscle wasting in the presence of disease, why is it so variable? Biol Rev Camb Philos Soc 2018; 94:1038-1055. [PMID: 30588725 DOI: 10.1111/brv.12489] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Revised: 11/22/2018] [Accepted: 11/27/2018] [Indexed: 12/16/2022]
Abstract
Skeletal muscle wasting is a common clinical feature of many chronic diseases and also occurs in response to single acute events. The accompanying loss of strength can lead to significant disability, increased care needs and have profound negative effects on quality of life. As muscle is the most abundant source of amino acids in the body, it appears to function as a buffer for fuel and substrates that can be used to repair damage elsewhere and to feed the immune system. In essence, the fundamentals of muscle wasting are simple: less muscle is made than is broken down. However, although well-described mechanisms modulate muscle protein turnover, significant individual differences in the amount of muscle lost in the presence of a given severity of disease complicate the understanding of underlying mechanisms and suggest that individuals have different sensitivities to signals for muscle loss. Furthermore, the rate at which muscle protein is turned over under normal conditions means that clinically significant muscle loss can occur with changes in the rate of protein synthesis and/or breakdown that are too small to be measurable. Consequently, the changes in expression of factors regulating muscle turnover required to cause a decline in muscle mass are small and, except in cases of rapid wasting, there is no consistent pattern of change in the expression of factors that regulate muscle mass. MicroRNAs are fine tuners of cell phenotype and are therefore ideally suited to cause the subtle changes in proteome required to tilt the balance between synthesis and degradation in a way that causes clinically significant wasting. Herein we present a model in which muscle loss as a consequence of disease in non-muscle tissue is modulated by a set of microRNAs, the muscle expression of which is associated with severity of disease in the non-muscle tissue. These microRNAs alter fundamental biological processes including the synthesis of ribosomes and mitochondria leading to reduced protein synthesis and increased protein breakdown, thereby freeing amino acids from the muscle. We argue that the variability in muscle loss observed in the human population arises from at least two sources. The first is from pre-existing or disease-induced variation in the expression of microRNAs controlling the sensitivity of muscle to the atrophic signal and the second is from the expression of microRNAs from imprinted loci (i.e. only expressed from the maternally or paternally inherited allele) and may control the rate of myonuclear recruitment. In the absence of disease, these factors do not correlate with muscle mass, since there is no challenge to the established balance. However, in the presence of such a challenge, these microRNAs determine the rate of decline for a given disease severity. Together these mechanisms provide novel insight into the loss of muscle mass and its variation in the human population. The involvement of imprinted loci also suggests that genes that regulate early development also contribute to the ability of individuals to resist muscle loss in response to disease.
Collapse
Affiliation(s)
- Paul R Kemp
- National Heart & Lung Institute, Imperial College London, South Kensington Campus, London, SW7 2AZ, U.K
| | - Mark Griffiths
- National Heart & Lung Institute, Imperial College London, South Kensington Campus, London, SW7 2AZ, U.K
| | - Michael I Polkey
- National Institute for Health Research Respiratory Biomedical Research Unit, Royal Brompton and Harefield NHS Foundation Trust and Imperial College London, Sydney Street, London SW3 6NP, U.K
| |
Collapse
|
236
|
Zheng R, Xie L, Liu W, Guo Y, Wang Y, Wu Y, Liu Y, Luo H, Kang N, Yuan Q. Recombinant growth differentiation factor 11 impairs fracture healing through inhibiting chondrocyte differentiation. Ann N Y Acad Sci 2018; 1440:54-66. [PMID: 30575056 DOI: 10.1111/nyas.13994] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2018] [Revised: 10/05/2018] [Accepted: 11/14/2018] [Indexed: 02/05/2023]
Abstract
Growth differentiation factor 11 (GDF11), a secreted member of the transforming growth factor-β (TGF-β) superfamily, has been reported to have the capacity to reverse age-related pathologic changes and regulate organ regeneration after injury; however, the role of GDF11 in fracture healing and bone repair is still unclear. Here, we established a fracture model in 12-week-old male mice to observe two healing states: the cartilaginous callus and bony callus formation phases. Our results showed that recombinant GDF11 (rGDF11) injection inhibits cartilaginous callus maturation and hard callus formation, thereby impairing fracture healing in vivo. In vitro, rGDF11 administration inhibited chondrocyte differentiation and maturation by phosphorylating SMAD2/3 protein and inhibiting RUNX2 expression. Notably, inhibition of TGF-β activity by a SMAD-specific inhibitor attenuated GDF11 effects. Thus, our study demonstrates that, rather than acting as a rejuvenating agent, rGDF11 impairs fracture healing by inhibiting chondrocyte differentiation and maturation.
Collapse
Affiliation(s)
- Rixin Zheng
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Liang Xie
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Weiqing Liu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yuchen Guo
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yuan Wang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yunshu Wu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yuting Liu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Hongke Luo
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Ning Kang
- Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Quan Yuan
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
237
|
Evano B, Tajbakhsh S. Skeletal muscle stem cells in comfort and stress. NPJ Regen Med 2018; 3:24. [PMID: 30588332 PMCID: PMC6303387 DOI: 10.1038/s41536-018-0062-3] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Accepted: 11/28/2018] [Indexed: 12/21/2022] Open
Abstract
Investigations on developmental and regenerative myogenesis have led to major advances in decrypting stem cell properties and potential, as well as their interactions within the evolving niche. As a consequence, regenerative myogenesis has provided a forum to investigate intrinsic regulators of stem cell properties as well as extrinsic factors, including stromal cells, during normal growth and following injury and disease. Here we review some of the latest advances in the field that have exposed fundamental processes including regulation of stress following trauma and ageing, senescence, DNA damage control and modes of symmetric and asymmetric cell divisions. Recent studies have begun to explore the nature of the niche that is distinct in different muscle groups, and that is altered from prenatal to postnatal stages, and during ageing. We also discuss heterogeneities among muscle stem cells and how distinct properties within the quiescent and proliferating cell states might impact on homoeostasis and regeneration. Interestingly, cellular quiescence, which was thought to be a passive cell state, is regulated by multiple mechanisms, many of which are deregulated in various contexts including ageing. These and other factors including metabolic activity and genetic background can impact on the efficiency of muscle regeneration.
Collapse
Affiliation(s)
- Brendan Evano
- Stem Cells and Development, Department of Developmental & Stem Cell Biology, Institut Pasteur, 75015 Paris, France
- CNRS UMR 3738, Institut Pasteur, 75015 Paris, France
| | - Shahragim Tajbakhsh
- Stem Cells and Development, Department of Developmental & Stem Cell Biology, Institut Pasteur, 75015 Paris, France
- CNRS UMR 3738, Institut Pasteur, 75015 Paris, France
| |
Collapse
|
238
|
Rochette L, Meloux A, Rigal E, Zeller M, Cottin Y, Malka G, Vergely C. Regenerative Capacity of Endogenous Factor: Growth Differentiation Factor 11; a New Approach of the Management of Age-Related Cardiovascular Events. Int J Mol Sci 2018; 19:ijms19123998. [PMID: 30545044 PMCID: PMC6321079 DOI: 10.3390/ijms19123998] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Revised: 12/05/2018] [Accepted: 12/05/2018] [Indexed: 12/17/2022] Open
Abstract
Aging is a complicated pathophysiological process accompanied by a wide array of biological adaptations. The physiological deterioration correlates with the reduced regenerative capacity of tissues. The rejuvenation of tissue regeneration in aging organisms has also been observed after heterochronic parabiosis. With this model, it has been shown that exposure to young blood can rejuvenate the regenerative capacity of peripheral tissues and brain in aged animals. An endogenous compound called growth differentiation factor 11 (GDF11) is a circulating negative regulator of cardiac hypertrophy, suggesting that raising GDF11 levels could potentially treat or prevent cardiac diseases. The protein GDF11 is found in humans as well as animals. The existence of endogenous regulators of regenerative capacity, such as GDF11, in peripheral tissues and brain has now been demonstrated. It will be important to investigate the mechanisms with therapeutic promise that induce the regenerative effects of GDF11 for a variety of age-related diseases.
Collapse
Affiliation(s)
- Luc Rochette
- Equipe d'Accueil (EA 7460): Physiopathologie et Epidémiologie Cérébro-Cardiovasculaires (PEC2), Université de Bourgogne-Franche Comté, Faculté des Sciences de Santé, 7 Bd Jeanne d'Arc, 21000 Dijon, France.
| | - Alexandre Meloux
- Equipe d'Accueil (EA 7460): Physiopathologie et Epidémiologie Cérébro-Cardiovasculaires (PEC2), Université de Bourgogne-Franche Comté, Faculté des Sciences de Santé, 7 Bd Jeanne d'Arc, 21000 Dijon, France.
| | - Eve Rigal
- Equipe d'Accueil (EA 7460): Physiopathologie et Epidémiologie Cérébro-Cardiovasculaires (PEC2), Université de Bourgogne-Franche Comté, Faculté des Sciences de Santé, 7 Bd Jeanne d'Arc, 21000 Dijon, France.
| | - Marianne Zeller
- Equipe d'Accueil (EA 7460): Physiopathologie et Epidémiologie Cérébro-Cardiovasculaires (PEC2), Université de Bourgogne-Franche Comté, Faculté des Sciences de Santé, 7 Bd Jeanne d'Arc, 21000 Dijon, France.
| | - Yves Cottin
- Equipe d'Accueil (EA 7460): Physiopathologie et Epidémiologie Cérébro-Cardiovasculaires (PEC2), Université de Bourgogne-Franche Comté, Faculté des Sciences de Santé, 7 Bd Jeanne d'Arc, 21000 Dijon, France.
- Service de Cardiologie-CHU-Dijon, 21 000 Dijon, France.
| | - Gabriel Malka
- Institut de formation en biotechnologie et ingénierie biomédicale (IFR2B), Université Mohammed VI Polytechnique, 43 150 Ben-Guerir, Morocco.
| | - Catherine Vergely
- Equipe d'Accueil (EA 7460): Physiopathologie et Epidémiologie Cérébro-Cardiovasculaires (PEC2), Université de Bourgogne-Franche Comté, Faculté des Sciences de Santé, 7 Bd Jeanne d'Arc, 21000 Dijon, France.
| |
Collapse
|
239
|
Zhou Y, Ni S, Song L, Wang X, Zhang Y, Zhang S. Late-onset administration of GDF11 extends life span and delays development of age-related markers in the annual fish Nothobranchius guentheri. Biogerontology 2018; 20:225-239. [DOI: 10.1007/s10522-018-09789-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Accepted: 11/30/2018] [Indexed: 02/06/2023]
|
240
|
Riuzzi F, Sorci G, Arcuri C, Giambanco I, Bellezza I, Minelli A, Donato R. Cellular and molecular mechanisms of sarcopenia: the S100B perspective. J Cachexia Sarcopenia Muscle 2018; 9:1255-1268. [PMID: 30499235 PMCID: PMC6351675 DOI: 10.1002/jcsm.12363] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Accepted: 09/27/2018] [Indexed: 12/11/2022] Open
Abstract
Primary sarcopenia is a condition of reduced skeletal muscle mass and strength, reduced agility, and increased fatigability and risk of bone fractures characteristic of aged, otherwise healthy people. The pathogenesis of primary sarcopenia is not completely understood. Herein, we review the essentials of the cellular and molecular mechanisms of skeletal mass maintenance; the alterations of myofiber metabolism and deranged properties of muscle satellite cells (the adult stem cells of skeletal muscles) that underpin the pathophysiology of primary sarcopenia; the role of the Ca2+ -sensor protein, S100B, as an intracellular factor and an extracellular signal regulating cell functions; and the functional role of S100B in muscle tissue. Lastly, building on recent results pointing to S100B as to a molecular determinant of myoblast-brown adipocyte transition, we propose S100B as a transducer of the deleterious effects of accumulation of reactive oxygen species in myoblasts and, potentially, myofibers concurring to the pathophysiology of sarcopenia.
Collapse
Affiliation(s)
- Francesca Riuzzi
- Department of Experimental Medicine, University of Perugia, Perugia, 06132, Italy.,Interuniversity Institute of Myology
| | - Guglielmo Sorci
- Department of Experimental Medicine, University of Perugia, Perugia, 06132, Italy.,Interuniversity Institute of Myology
| | - Cataldo Arcuri
- Department of Experimental Medicine, University of Perugia, Perugia, 06132, Italy.,Interuniversity Institute of Myology
| | - Ileana Giambanco
- Department of Experimental Medicine, University of Perugia, Perugia, 06132, Italy.,Interuniversity Institute of Myology
| | - Ilaria Bellezza
- Department of Experimental Medicine, University of Perugia, Perugia, 06132, Italy
| | - Alba Minelli
- Department of Experimental Medicine, University of Perugia, Perugia, 06132, Italy
| | - Rosario Donato
- Department of Experimental Medicine, University of Perugia, Perugia, 06132, Italy.,Interuniversity Institute of Myology.,Centro Universitario di Ricerca sulla Genomica Funzionale, University of Perugia, Perugia, 06132, Italy
| |
Collapse
|
241
|
Wang L, Wang Y, Wang Z, Qi Y, Zong B, Liu M, Li X, Zhang X, Liu C, Cao R, Ma Y. Growth differentiation factor 11 ameliorates experimental colitis by inhibiting NLRP3 inflammasome activation. Am J Physiol Gastrointest Liver Physiol 2018; 315:G909-G920. [PMID: 30188752 DOI: 10.1152/ajpgi.00159.2018] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Growth differentiation factor 11 (GDF11) has an anti-inflammatory effect in the mouse model of atherosclerosis and Alzheimer's disease, but how GDF11 regulates intestinal inflammation during ulcerative colitis (UC) is poorly defined. The Nod-like receptor family pyrin domain-1 containing 3 (NLRP3) inflammasome is closely associated with intestinal inflammation because of its ability to increase IL-1β secretion. Our aim is to determine whether GDF11 has an effect on attenuating experimental colitis in mice. In this study, using a dextran sodium sulfate (DSS)-induced acute colitis mouse model, we reported that GDF11 treatment attenuated loss of body weight, the severity of the disease activity index, shortening of the colon, and histological changes in the colon. GDF11 remarkably suppressed IL-1β secretion and NLRP3 inflammasome activation in colon samples and RAW 264.7 cells, such as the levels of NLRP3 and activated caspase-1. Furthermore, we found that GDF11 inhibited NLRP3 inflammasome activation by downregulating the Toll-like receptor 4/NF-κB p65 pathway and reactive oxygen species production via the typical Smad2/3 pathway. Thus, our research shows that GDF11 alleviates DSS-induced colitis by inhibiting NLRP3 inflammasome activation, providing some basis for its potential use in the treatment of UC. NEW & NOTEWORTHY Here, we identify a new role for growth differentiation factor 11 (GDF11), which ameliorates dextran sodium sulfate-induced acute colitis. Meanwhile, we discover a new phenomenon of GDF11 inhibiting IL-1β secretion and Nod-like receptor family pyrin domain-1 containing 3 (NLRP3) inflammasome activation. These findings reveal that GDF11 is a new potential candidate for the treatment of ulcerative colitis patients with a hyperactive NLRP3 inflammasome.
Collapse
Affiliation(s)
- Lanju Wang
- School of Basic Medical Sciences, Zhengzhou University , Zhengzhou, Henan , China
| | - Yaohui Wang
- Joint National Laboratory for Antibody Drug Engineering, School of Basic Medical Sciences, Henan University , Kaifeng, Henan , China
| | - Zhenfeng Wang
- Joint National Laboratory for Antibody Drug Engineering, School of Basic Medical Sciences, Henan University , Kaifeng, Henan , China
| | - Yu Qi
- Joint National Laboratory for Antibody Drug Engineering, School of Basic Medical Sciences, Henan University , Kaifeng, Henan , China
| | - Beibei Zong
- Joint National Laboratory for Antibody Drug Engineering, School of Basic Medical Sciences, Henan University , Kaifeng, Henan , China
| | - Meichen Liu
- Joint National Laboratory for Antibody Drug Engineering, School of Basic Medical Sciences, Henan University , Kaifeng, Henan , China
| | - Xuefang Li
- Joint National Laboratory for Antibody Drug Engineering, School of Basic Medical Sciences, Henan University , Kaifeng, Henan , China
| | - Xingkun Zhang
- Joint National Laboratory for Antibody Drug Engineering, School of Basic Medical Sciences, Henan University , Kaifeng, Henan , China
| | - Chengguo Liu
- Joint National Laboratory for Antibody Drug Engineering, School of Basic Medical Sciences, Henan University , Kaifeng, Henan , China
| | - Run Cao
- Joint National Laboratory for Antibody Drug Engineering, School of Basic Medical Sciences, Henan University , Kaifeng, Henan , China
| | - Yuanfang Ma
- School of Basic Medical Sciences, Zhengzhou University , Zhengzhou, Henan , China.,Joint National Laboratory for Antibody Drug Engineering, School of Basic Medical Sciences, Henan University , Kaifeng, Henan , China
| |
Collapse
|
242
|
Wallner C, Wagner JM, Dittfeld S, Drysch M, Lehnhardt M, Behr B. Myostatin serum concentration as an indicator for deviated muscle metabolism in severe burn injuries. Scand J Surg 2018; 108:297-304. [PMID: 30474468 DOI: 10.1177/1457496918812230] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
INTRODUCTION Patients experiencing thermal injuries with an extent of over 20% of total body surface area suffer from systemic catabolic disease. The thermal trauma-induced loss of muscle mass causes a higher incidence for comorbidities and subsequently a higher mortality. In this study, we aimed to investigate the role of myostatin in the interplay with follistatin during muscle cachexia. METHODS Patients with burn injuries (>10% total body surface area) between the ages of 18 and 75 were prospectively included within the first 48 h after trauma to determine deviations of parameters connected to muscle catabolism. In the chronic state of burn injury (9-12 months after trauma), we re-evaluated myostatin and follistatin concentrations as well as muscle strength of the non-dominant forearm. RESULTS We were able to show a time-dependent alteration (9-12 months after burn injury) of myostatin with an initial decrease (p < 0.001) and long-term increase (p < 0.001) after thermal injury in blood serum. For follistatin, a reciprocal correlation was observed (r = -0.707, p = 0.001). Accordingly, muscle strength of the non-dominant hand and forearm was significantly decreased 9-12 months after injury in post-burn patients compared with healthy patients with a significant correlation to myostatin levels (r = -0.899, p < 0.001). In addition, initial myostatin serum concentration was predictive for long-term muscle strength impairment. CONCLUSION With regard to the muscle metabolism after thermal trauma, our data suggest an acute anabolic response, presumably to spare muscle mass, which is converted to catabolic conditions accompanied by muscle strength reduction in the chronic phase. Myostatin plays a crucial role in this orchestration and initial myostatin concentration may predict the long-term muscle strength.
Collapse
Affiliation(s)
- C Wallner
- Department of Plastic Surgery, BG University Hospital Bergmannsheil, Ruhr University Bochum, Bochum, Germany
| | - J M Wagner
- Department of Plastic Surgery, BG University Hospital Bergmannsheil, Ruhr University Bochum, Bochum, Germany
| | - S Dittfeld
- Department of Plastic Surgery, BG University Hospital Bergmannsheil, Ruhr University Bochum, Bochum, Germany
| | - M Drysch
- Department of Plastic Surgery, BG University Hospital Bergmannsheil, Ruhr University Bochum, Bochum, Germany
| | - M Lehnhardt
- Department of Plastic Surgery, BG University Hospital Bergmannsheil, Ruhr University Bochum, Bochum, Germany
| | - B Behr
- Department of Plastic Surgery, BG University Hospital Bergmannsheil, Ruhr University Bochum, Bochum, Germany
| |
Collapse
|
243
|
Ozek C, Krolewski RC, Buchanan SM, Rubin LL. Growth Differentiation Factor 11 treatment leads to neuronal and vascular improvements in the hippocampus of aged mice. Sci Rep 2018; 8:17293. [PMID: 30470794 PMCID: PMC6251885 DOI: 10.1038/s41598-018-35716-6] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Accepted: 11/06/2018] [Indexed: 01/09/2023] Open
Abstract
Aging is the biggest risk factor for several neurodegenerative diseases. Parabiosis experiments have established that old mouse brains are improved by exposure to young mouse blood. Previously, our lab showed that delivery of Growth Differentiation Factor 11 (GDF11) to the bloodstream increases the number of neural stem cells and positively affects vasculature in the subventricular zone of old mice. Our new study demonstrates that GDF11 enhances hippocampal neurogenesis, improves vasculature and increases markers of neuronal activity and plasticity in the hippocampus and cortex of old mice. Our experiments also demonstrate that systemically delivered GDF11, rather than crossing the blood brain barrier, exerts at least some of its effects by acting on brain endothelial cells. Thus, by targeting the cerebral vasculature, GDF11 has a very different mechanism from that of previously studied circulating factors acting to improve central nervous system (CNS) function without entering the CNS.
Collapse
Affiliation(s)
- Ceren Ozek
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, 02138, USA. .,Harvard Stem Cell Institute, Harvard University, Cambridge, MA, 02138, USA.
| | - Richard C Krolewski
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, 02138, USA.,Harvard Stem Cell Institute, Harvard University, Cambridge, MA, 02138, USA.,Department of Neurology, Brigham and Women's Hospital, Massachusetts General Hospital, Boston, MA, 02115, USA
| | - Sean M Buchanan
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, 02138, USA.,Harvard Stem Cell Institute, Harvard University, Cambridge, MA, 02138, USA
| | - Lee L Rubin
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, 02138, USA. .,Harvard Stem Cell Institute, Harvard University, Cambridge, MA, 02138, USA.
| |
Collapse
|
244
|
Amico F, Briggs G, Balogh ZJ. Transfused trauma patients have better outcomes when transfused with blood components from young donors. Med Hypotheses 2018; 122:141-146. [PMID: 30593399 DOI: 10.1016/j.mehy.2018.11.016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Revised: 11/14/2018] [Accepted: 11/21/2018] [Indexed: 12/20/2022]
Abstract
The physiology of tissue healing and aging share common pathways. Both patient age and tissue healing are crucial factors predicting outcomes in trauma patients. The presented hypothesis focuses on the concept that transfused trauma patients have better outcomes when transfused with blood components from young donors. The age of the donor of a blood transfusion could affect recovery following a major traumatic insult and help avoid postinjury immune paralysis and its associated complications. The frequent transfusion of blood components to the severely injured trauma patient provides an opportunity for the recipient to benefit from the potentially favourable effect of blood originating from young donors. Different types of evidence support the presented hypothesis including work on soluble circulating factors, research on animal parabiontic models and epidemiological studies. Theories on the role of transfusion of cells, on bone marrow and on senolytics also represent grounds to elaborate pathways to test this hypothesis. The precise molecular mechanism underlying this hypothesis is uncertain. A beneficial effect on trauma patients following transfusion of blood could be due to a positive effect of blood donated from younger donors or instead to the lack of a negative effect possibly occurring when transfusing blood from older donors. Either way, identifying this mechanism would provide a powerful tool enhance long and short term recovery after trauma.
Collapse
Affiliation(s)
- Francesco Amico
- Department of Traumatology, John Hunter Hospital and University of Newcastle, Australia
| | - Gabrielle Briggs
- Department of Traumatology, John Hunter Hospital and University of Newcastle, Australia
| | - Zsolt J Balogh
- Department of Traumatology, John Hunter Hospital and University of Newcastle, Australia.
| |
Collapse
|
245
|
Patsalos A, Simandi Z, Hays TT, Peloquin M, Hajian M, Restrepo I, Coen PM, Russell AJ, Nagy L. In vivo GDF3 administration abrogates aging related muscle regeneration delay following acute sterile injury. Aging Cell 2018; 17:e12815. [PMID: 30003692 PMCID: PMC6156497 DOI: 10.1111/acel.12815] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2018] [Revised: 05/17/2018] [Accepted: 06/15/2018] [Indexed: 12/22/2022] Open
Abstract
Tissue regeneration is a highly coordinated process with sequential events including immune cell infiltration, clearance of damaged tissues, and immune‐supported regrowth of the tissue. Aging has a well‐documented negative impact on this process globally; however, whether changes in immune cells per se are contributing to the decline in the body’s ability to regenerate tissues with aging is not clearly understood. Here, we set out to characterize the dynamics of macrophage infiltration and their functional contribution to muscle regeneration by comparing young and aged animals upon acute sterile injury. Injured muscle of old mice showed markedly elevated number of macrophages, with a predominance for Ly6Chigh pro‐inflammatory macrophages and a lower ratio of the Ly6Clow repair macrophages. Of interest, a recently identified repair macrophage‐derived cytokine, growth differentiation factor 3 (GDF3), was markedly downregulated in injured muscle of old relative to young mice. Supplementation of recombinant GDF3 in aged mice ameliorated the inefficient regenerative response. Together, these results uncover a deficiency in the quantity and quality of infiltrating macrophages during aging and suggest that in vivo administration of GDF3 could be an effective therapeutic approach.
Collapse
Affiliation(s)
- Andreas Patsalos
- Sanford-Burnham-Prebys Medical Discovery Institute at Lake Nona; Orlando Florida
- Department of Biochemistry and Molecular Biology, Faculty of Medicine; University of Debrecen; Debrecen Hungary
| | - Zoltan Simandi
- Sanford-Burnham-Prebys Medical Discovery Institute at Lake Nona; Orlando Florida
| | - Tristan T. Hays
- Sanford-Burnham-Prebys Medical Discovery Institute at Lake Nona; Orlando Florida
| | - Matthew Peloquin
- Sanford-Burnham-Prebys Medical Discovery Institute at Lake Nona; Orlando Florida
| | - Matine Hajian
- Sanford-Burnham-Prebys Medical Discovery Institute at Lake Nona; Orlando Florida
| | - Isabella Restrepo
- Sanford-Burnham-Prebys Medical Discovery Institute at Lake Nona; Orlando Florida
| | - Paul M. Coen
- Sanford-Burnham-Prebys Medical Discovery Institute at Lake Nona; Orlando Florida
- Florida Hospital; Translational Research Institute for Metabolism and Diabetes; Orlando Florida
| | - Alan J. Russell
- Muscle Metabolism Discovery Performance Unit; GlaxoSmithKline; King of Prussia Pennsylvania
| | - Laszlo Nagy
- Sanford-Burnham-Prebys Medical Discovery Institute at Lake Nona; Orlando Florida
- Department of Biochemistry and Molecular Biology, Faculty of Medicine; University of Debrecen; Debrecen Hungary
| |
Collapse
|
246
|
Zhang J, Li Y, Li H, Zhu B, Wang L, Guo B, Xiang L, Dong J, Liu M, Xiang G. GDF11 Improves Angiogenic Function of EPCs in Diabetic Limb Ischemia. Diabetes 2018; 67:2084-2095. [PMID: 30026260 DOI: 10.2337/db17-1583] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2017] [Accepted: 06/29/2018] [Indexed: 11/13/2022]
Abstract
Growth differentiation factor 11 (GDF11) has been shown to promote stem cell activity and rejuvenate the function of multiple organs in old mice, but little is known about the functions of GDF11 in the diabetic rat model of hindlimb ischemia. In this study, we found that systematic replenishment of GDF11 rescues angiogenic function of endothelial progenitor cells (EPCs) and subsequently improves vascularization and increases blood flow in diabetic rats with hindlimb ischemia. Conversely, anti-GDF11 monoclonal antibody treatment caused impairment of vascularization and thus, decreased blood flow. In vitro treatment of EPCs with recombinant GDF11 attenuated EPC dysfunction and apoptosis. Mechanistically, the GDF11-mediated positive effects could be attributed to the activation of the transforming growth factor-β/Smad2/3 and protein kinase B/hypoxia-inducible factor 1α pathways. These findings suggest that GDF11 repletion may enhance EPC resistance to diabetes-induced damage, improve angiogenesis, and thus, increase blood flow. This benefit of GDF11 may lead to a new therapeutic approach for diabetic hindlimb ischemia.
Collapse
Affiliation(s)
- Jiajia Zhang
- Department of Endocrinology, Wuhan General Hospital of Guangzhou Command, Wuhan, Hubei, China
| | - Yixiang Li
- Department of Hematology and Medical Oncology, School of Medicine, Emory University, Atlanta, GA
| | - Huan Li
- Department of Endocrinology, Wuhan General Hospital of Guangzhou Command, Wuhan, Hubei, China
| | - Biao Zhu
- Department of Endocrinology, Wuhan General Hospital of Guangzhou Command, Wuhan, Hubei, China
| | - Li Wang
- Department of Endocrinology, Wuhan General Hospital of Guangzhou Command, Wuhan, Hubei, China
| | - Bei Guo
- Department of Endocrinology, Wuhan General Hospital of Guangzhou Command, Wuhan, Hubei, China
| | - Lin Xiang
- Department of Endocrinology, Wuhan General Hospital of Guangzhou Command, Wuhan, Hubei, China
| | - Jing Dong
- Department of Endocrinology, Wuhan General Hospital of Guangzhou Command, Wuhan, Hubei, China
| | - Min Liu
- Department of Endocrinology, Wuhan General Hospital of Guangzhou Command, Wuhan, Hubei, China
| | - Guangda Xiang
- Department of Endocrinology, Wuhan General Hospital of Guangzhou Command, Wuhan, Hubei, China
| |
Collapse
|
247
|
Renzini A, Marroncelli N, Noviello C, Moresi V, Adamo S. HDAC4 Regulates Skeletal Muscle Regeneration via Soluble Factors. Front Physiol 2018; 9:1387. [PMID: 30319457 PMCID: PMC6171007 DOI: 10.3389/fphys.2018.01387] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Accepted: 09/12/2018] [Indexed: 12/11/2022] Open
Abstract
Skeletal muscle possesses a high ability to regenerate after an insult or in pathological conditions, relying on satellite cells, the skeletal muscle stem cells. Satellite cell behavior is tightly regulated by the surrounding microenvironment, which provides multiple signals derived from local cells and systemic factors. Among epigenetic mechanisms, histone deacetylation has been proved to affect muscle regeneration. Indeed, pan-histone deacetylase inhibitors were found to improve muscle regeneration, while deletion of histone deacetylase 4 (HDAC4) in satellite cells inhibits their proliferation and differentiation, leading to compromised muscle regeneration. In this study, we delineated the HDAC4 function in adult skeletal muscle, following injury, by using a tissue-specific null mouse line. We showed that HDAC4 is crucial for skeletal muscle regeneration by mediating soluble factors that influence muscle-derived cell proliferation and differentiation. These findings add new biological functions to HDAC4 in skeletal muscle that need considering when administering histone deacetylase inhibitors.
Collapse
Affiliation(s)
- Alessandra Renzini
- DAHFMO Unit of Histology and Medical Embryology, Interuniversity Institute of Myology, Sapienza University of Rome, Rome, Italy
| | - Nicoletta Marroncelli
- DAHFMO Unit of Histology and Medical Embryology, Interuniversity Institute of Myology, Sapienza University of Rome, Rome, Italy
| | - Chiara Noviello
- DAHFMO Unit of Histology and Medical Embryology, Interuniversity Institute of Myology, Sapienza University of Rome, Rome, Italy
| | - Viviana Moresi
- DAHFMO Unit of Histology and Medical Embryology, Interuniversity Institute of Myology, Sapienza University of Rome, Rome, Italy.,Laboratory of Cardiovascular Endocrinology, IRCCS San Raffaele Pisana, Rome, Italy
| | - Sergio Adamo
- DAHFMO Unit of Histology and Medical Embryology, Interuniversity Institute of Myology, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
248
|
Potential therapeutic role of Co-Q10 in alleviating intervertebral disc degeneration and suppressing IL-1β-mediated inflammatory reaction in NP cells. Int Immunopharmacol 2018; 64:424-431. [PMID: 30261465 DOI: 10.1016/j.intimp.2018.09.029] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Revised: 08/13/2018] [Accepted: 09/19/2018] [Indexed: 12/24/2022]
Abstract
Coenzyme Q10 (Co-Q10) is extraordinarily popular and has been used in abundant interventions as an antioxidant reagent that participates in numerous oxidation reactions. According to substantial evidence previously reported, interleukin-1β (IL-1β) is deemed to be one of the chief orchestrator molecules in the degeneration of intervertebral disc (IVD). However, it is unknown whether Co-Q10 is able to protect against IVD degeneration. In the current study, mouse-derived IVDs as well as primary human nucleus pulposus (NP) cells were isolated and cultured. NP cells were stimulated with IL-1β, with or without selective addition of Co-Q10 to investigate the therapeutic effect of Co-Q10 on IVD degeneration. Levels of IL-1β-induced inflammatory biomarkers including TNF-α, COX-2, IL-6 and iNOS were reduced by Co-Q10, which was possibly associated with inhibition of NF-κB signaling activation. Furthermore, Co-Q10 maintained the production of anabolic biomarkers in NP cells such as collagen 2, aggrecan and Sox-9 and altered the enhanced catabolism induced by IL-1β. Moreover, the therapeutic role of Co-Q10 in sustaining IVD tissue-enhanced anabolism is potentially dependent on activation of the Akt signaling pathway. In summary, Co-Q10 may potentially represent an available molecular target that may shed light on approaches to the prevention and treatment of IVD degeneration in the future.
Collapse
|
249
|
Shen GS, Zhou HB, Zhang H, Chen B, Liu ZP, Yuan Y, Zhou XZ, Xu YJ. The GDF11-FTO-PPARγ axis controls the shift of osteoporotic MSC fate to adipocyte and inhibits bone formation during osteoporosis. Biochim Biophys Acta Mol Basis Dis 2018; 1864:3644-3654. [PMID: 30279140 DOI: 10.1016/j.bbadis.2018.09.015] [Citation(s) in RCA: 84] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Revised: 08/23/2018] [Accepted: 09/13/2018] [Indexed: 12/20/2022]
Abstract
During osteoporosis, the shift of bone mesenchymal stem cell (BMSC) lineage commitment to adipocyte leads to the imbalance between bone mass and fat, which increases the risk of fracture. The mechanism underlying this process is not fully understood. Fat mass and obesity-associated protein (FTO) is an RNA demethylase that demethylates various methylated nucleic acids and participates in various physiological and pathological processes. Here we identified FTO as a regulator for BMSC fate determination during osteoporosis. FTO was up-regulated in bone marrow during aging or osteoporosis in human and mice in a GDF11(growth differentiation factor 11)-C/EBPα-dependent mechanism. The expression of FTO was also up-regulated during adipocyte differentiation of BMSCs whereas its expression was down-regulated during osteoblast differentiation. Gain-of-function and loss-of-function experiments showed that FTO favored the BMSCs to differentiate to adipocytes rather than osteoblasts. Further mechanism study demonstrated that FTO bound and demethylated the mRNA of the Peroxisome proliferator-activated receptor gamma (Pparg), leading to the increase in the expression of Pparg mRNA. Reversely, Pparg knockdown blocked the function of GDF11-FTO during osteoblast differentiation of BMSCs. Furthermore, conditionally genetic knockout of Fto in osteoblasts inhibited the development of osteopenia in mice. Collectively, our findings demonstrated that GDF11-FTO-Pparg axis promoted the shift of osteoporotic BMSC fate to adipocyte and inhibited bone formation during osteoporosis.
Collapse
Affiliation(s)
- Guang-Si Shen
- Department of Orthopaedics, The Second Affiliated Hospital of Soochow University, 1055 Sanxiang Road, 215004 Suzhou, China; Osteoporosis Institute of Soochow University, 1055 Sanxiang Road, 215004 Suzhou, China
| | - Hai-Bin Zhou
- Department of Orthopaedics, The Second Affiliated Hospital of Soochow University, 1055 Sanxiang Road, 215004 Suzhou, China
| | - Hong Zhang
- Department of Obstetrics and Gynaecology, The Second Affiliated Hospital of Soochow University, 1055 Sanxiang Road, 215004 Suzhou, China
| | - Bin Chen
- Department of Orthopaedics, The Second Affiliated Hospital of Soochow University, 1055 Sanxiang Road, 215004 Suzhou, China; Osteoporosis Institute of Soochow University, 1055 Sanxiang Road, 215004 Suzhou, China
| | - Zhi-Peng Liu
- Department of Orthopaedics, The Second Affiliated Hospital of Soochow University, 1055 Sanxiang Road, 215004 Suzhou, China
| | - Ye Yuan
- Department of Orthopaedics, The Second Affiliated Hospital of Soochow University, 1055 Sanxiang Road, 215004 Suzhou, China
| | - Xiao-Zhong Zhou
- Department of Orthopaedics, The Second Affiliated Hospital of Soochow University, 1055 Sanxiang Road, 215004 Suzhou, China.
| | - You-Jia Xu
- Department of Orthopaedics, The Second Affiliated Hospital of Soochow University, 1055 Sanxiang Road, 215004 Suzhou, China; Osteoporosis Institute of Soochow University, 1055 Sanxiang Road, 215004 Suzhou, China.
| |
Collapse
|
250
|
Castellano JM. Blood-Based Therapies to Combat Aging. Gerontology 2018; 65:84-89. [PMID: 30196300 DOI: 10.1159/000492573] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Accepted: 07/31/2018] [Indexed: 12/23/2022] Open
Abstract
Dysfunction associated with the aging process positions aging as a leading culprit for development of devastating diseases and mounting health-care costs. Many age-associated conditions for which aging increases risk are neurological disorders with no effective treatments, including Alzhei-mer's disease. As the proportion of aged individuals continues to rise in the coming decades, aging-related costs are expected to increase dramatically. Diverse approaches have emerged to meet the clinical need to treat aging and its associated conditions, including those aimed at increasing longevity, slowing the aging process itself, and improving healthspan. An emerging approach takes advantage of molecules circulating in the blood to limit or reverse aspects of aging in various organs throughout the body. Efforts are underway to translate these findings into novel therapeutics that harness the activity of youth-associated molecules present within blood. Here, we discuss the current state of blood-based approaches in this arena. Despite the apparent ease with which blood products might conceivably be applied as treatment paradigms, we propose that challenges nonetheless exist, which may be overcome with mechanistic studies that identify common pathways for targeted therapeutics.
Collapse
|