1
|
Kim J, Lee MC, Jeon J, Rodríguez-delaRosa A, Endo Y, Kim DS, Madrigal-Salazar AD, Seo JW, Lee H, Kim KT, Moon JI, Park SG, Lopez-Pacheco MC, Alkhateeb AF, Sobahi N, Bassous N, Liu W, Lee JS, Kim S, Aykut DY, Nasr ML, Hussain MA, Lee SH, Kim WJ, Pourquié O, Sinha I, Shin SR. Combinational regenerative inductive effect of bio-adhesive hybrid hydrogels conjugated with hiPSC-derived myofibers and its derived EVs for volumetric muscle regeneration. Bioact Mater 2025; 43:579-602. [PMID: 40115877 PMCID: PMC11923440 DOI: 10.1016/j.bioactmat.2024.09.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 08/28/2024] [Accepted: 09/09/2024] [Indexed: 03/23/2025] Open
Abstract
In regenerative medicine, extracellular vesicles (EVs) possess the potential to repair injured cells by delivering modulatory factors. However, the therapeutic effect of EVs in large-scale tissue defects, which are subject to prolonged timelines for tissue architecture and functional restoration, remains poorly understood. In this study, we introduce EVs and cell-tethering hybrid hydrogels composed of tyramine-conjugated gelatin (GelTA) that can be in-situ crosslinked with EVs derived from human induced pluripotent stem cell-derived myofibers (hiPSC-myofibers) and hiPSC-muscle precursor cells. This hybrid hydrogel sustains the release of EVs and provides a beneficial nano-topography and mechanical properties for creating a favorable extracellular matrix. Secreted EVs from the hiPSC-myofibers contain specific microRNAs, potentially improving myogenesis and angiogenesis. Herein, we demonstrate increased myogenic markers and fusion/differentiation indexes through the combinatory effects of EVs and integrin-mediated adhesions in the 3D matrix. Furthermore, we observe a unique impact of EVs, which aid in maintaining the viability and phenotype of myofibers under harsh environments. The hybrid hydrogel in-situ crosslinked with hiPSCs and EVs is facilely used to fabricate large-scale muscle constructs by the stacking of micro-patterned hydrogel domains. Later, we confirmed a combinational effect, whereby muscle tissue regeneration and functional restoration were improved, via an in vivo murine volumetric muscle loss model.
Collapse
Affiliation(s)
- Jiseong Kim
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, 02139, USA
- Department of Medical Biotechnology, Dongguk University, 32 Dongguk-ro, Goyang, 10326, Republic of Korea
| | - Myung Chul Lee
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, 02139, USA
- Medicinal Materials Research Center, Biomedical Research Division, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Jieun Jeon
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, 02139, USA
- Department of Medical Biotechnology, Dongguk University, 32 Dongguk-ro, Goyang, 10326, Republic of Korea
| | - Alejandra Rodríguez-delaRosa
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, 02115, USA
- Department of Genetics, Harvard Medical School, Boston, MA, 02115, USA
- Harvard Stem Cell Institute, Harvard University, Boston, MA, 02138, USA
| | - Yori Endo
- Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Da-Seul Kim
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, 02139, USA
| | - Andrea Donaxi Madrigal-Salazar
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, 02139, USA
- Tecnológico de Monterrey, Escuela de Ingeniería y Ciencias, Campus Guadalajara, Av. Gral. Ramón Corona No 2514, Colonia Nuevo México, Zapopan, Jalisco, 45121, Mexico
| | - Jeong Wook Seo
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, 02139, USA
| | - Hyeseon Lee
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, 02139, USA
- Department of Biomaterials Science (BK21 Four Program), Life and Industry Convergence Institute, Pusan National University, Miryang, 50463, Republic of Korea
| | - Ki-Tae Kim
- Department of Molecular Genetics & Dental Pharmacology, School of Dentistry and Dental Research Institute, Dental Multi-omics Center, Seoul National University, Seoul, 03080, Republic of Korea
- Epigenetic Regulation of Aged Skeleto-Muscular System Laboratory, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, 08826, Republic of Korea
| | - Jae-I Moon
- Department of Molecular Genetics & Dental Pharmacology, School of Dentistry and Dental Research Institute, Dental Multi-omics Center, Seoul National University, Seoul, 03080, Republic of Korea
- Epigenetic Regulation of Aged Skeleto-Muscular System Laboratory, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, 08826, Republic of Korea
| | - Seung Gwa Park
- Department of Molecular Genetics & Dental Pharmacology, School of Dentistry and Dental Research Institute, Dental Multi-omics Center, Seoul National University, Seoul, 03080, Republic of Korea
- Epigenetic Regulation of Aged Skeleto-Muscular System Laboratory, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, 08826, Republic of Korea
| | - Mariana Carolina Lopez-Pacheco
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, 02139, USA
- Tecnológico de Monterrey, Escuela de Ingeniería y Ciencias, Campus Guadalajara, Av. Gral. Ramón Corona No 2514, Colonia Nuevo México, Zapopan, Jalisco, 45121, Mexico
| | - Abdulhameed F Alkhateeb
- Department of Electrical and Computer Engineering, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
| | - Nebras Sobahi
- Department of Electrical and Computer Engineering, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
| | - Nicole Bassous
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, 02139, USA
| | - Wenpeng Liu
- Division of Engineering in Medicine and Division of Renal Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Jae Seo Lee
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, 02139, USA
- Harvard Medical School and Wellman Center for Photomedicine, Massachusetts General Hospital, Cambridge, MA, 02139, USA
| | - Seongsoo Kim
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, 02139, USA
| | - Dilara Yilmaz Aykut
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, 02139, USA
| | - Mahmoud Lotfi Nasr
- Division of Engineering in Medicine and Division of Renal Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
- College of Medicine, Mohamed Bin Rashid University of Medicine and Health Sciences, Dubai, UAE
| | - Mohammad Asif Hussain
- Department of Electrical and Computer Engineering, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
| | - Soo-Hong Lee
- Department of Medical Biotechnology, Dongguk University, 32 Dongguk-ro, Goyang, 10326, Republic of Korea
| | - Woo-Jin Kim
- Department of Molecular Genetics & Dental Pharmacology, School of Dentistry and Dental Research Institute, Dental Multi-omics Center, Seoul National University, Seoul, 03080, Republic of Korea
- Epigenetic Regulation of Aged Skeleto-Muscular System Laboratory, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, 08826, Republic of Korea
| | - Olivier Pourquié
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, 02115, USA
- Department of Genetics, Harvard Medical School, Boston, MA, 02115, USA
- Harvard Stem Cell Institute, Harvard University, Boston, MA, 02138, USA
| | - Indranil Sinha
- Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Su Ryon Shin
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, 02139, USA
| |
Collapse
|
2
|
Aslam MA, Ma EB, Huh JY. Pathophysiology of sarcopenia: Genetic factors and their interplay with environmental factors. Metabolism 2023; 149:155711. [PMID: 37871831 DOI: 10.1016/j.metabol.2023.155711] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 10/05/2023] [Accepted: 10/17/2023] [Indexed: 10/25/2023]
Abstract
Sarcopenia is a geriatric disorder characterized by a progressive decline in muscle mass and function. This disorder has been associated with a range of adverse health outcomes, including fractures, functional deterioration, and increased mortality. The pathophysiology of sarcopenia is highly complex and multifactorial, involving both genetic and environmental factors as key contributors. This review consolidates current knowledge on the genetic factors influencing the pathogenesis of sarcopenia, particularly focusing on the altered gene expression of structural and metabolic proteins, growth factors, hormones, and inflammatory cytokines. While the influence of environmental factors such as physical inactivity, chronic diseases, smoking, alcohol consumption, and sleep disturbances on sarcopenia is relatively well understood, there is a dearth of studies examining their mechanistic roles. Therefore, this review emphasizes the interplay between genetic and environmental factors, elucidating their cumulative role in exacerbating the progression of sarcopenia beyond their individual effects. The unique contribution of this review lies in synthesizing the latest evidence on the genetic factors and their interaction with environmental factors, aiming to inform the development of novel therapeutic or preventive interventions for sarcopenia.
Collapse
Affiliation(s)
- Muhammad Arif Aslam
- College of Pharmacy, Chonnam National University, Gwangju, Republic of Korea
| | - Eun Bi Ma
- College of Pharmacy, Chonnam National University, Gwangju, Republic of Korea
| | - Joo Young Huh
- College of Pharmacy, Chonnam National University, Gwangju, Republic of Korea.
| |
Collapse
|
3
|
Anastasilakis AD, Polyzos SA, Rodopaios NE, Makras P, Kumar A, Kalra B, Mantzoros CS. Activins, follistatins and inhibins in postmenopausal osteoporosis: A proof of concept, case-control study. Metabolism 2023; 141:155397. [PMID: 36587801 DOI: 10.1016/j.metabol.2022.155397] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 12/24/2022] [Accepted: 12/26/2022] [Indexed: 12/31/2022]
Abstract
BACKGROUND Bone metabolism has been proposed to be affected by the activins-follistatins-inhibins (AFI) hormonal system. We aimed to evaluate AFI in patients with osteoporosis and osteopenia compared with postmenopausal and premenopausal controls. METHODS In this case-control study, circulating levels of the AFI system were evaluated, individually and jointly, between postmenopausal women with osteoporosis (BMD T-score ≤-2.5; n = 25) or osteopenia (BMD T-score >-2.5 and ≤-1; n = 25) and postmenopausal women with normal BMD (T-score >-1.0; n = 25) or premenopausal women with normal BMD (Z-score >-1.0; n = 25), with and without adjustment for potential confounders. RESULTS In the sum of participants, AFI molecules and their ratios followed an opposite pattern of correlations for age and BMI vs. BMD. In unadjusted models, FSTL3 concentrations were higher, whereas activin B, inhibin A and inhibin B and the ratios of activin B/follistatin and activin B/FSTL3 were lower in the three postmenopausal groups compared with the premenopausal group. Activin A/follistatin and activin AB/follistatin ratios were lower in the osteoporosis group than the other three groups. After adjustment for BMI and age, inhibin B (p = 0.005), and the ratios of activin A/follistatin (p = 0.009), activin B/follistatin (p = 0.040) and activin AB/follistatin (p = 0.003) were lower in the osteoporotic group compared with the other groups. In fully adjusted logistic regression analysis log(inhibin B) (p = 0.041), log(activinA/follistatin) (p = 0.014), log(activinB/follistatin) (p = 0.025) and log(activinAB/follistatin) (p = 0.021), but not FSTL3, remained independently associated with the presence of osteoporosis. CONCLUSIONS Lower inhibin B and higher ratios of activins A, B, and AB to follistatin are associated with lumbar spine BMD and the presence of osteoporosis independently from age or BMI.
Collapse
Affiliation(s)
| | - Stergios A Polyzos
- First Laboratory of Pharmacology, Medical School, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Nikolaos E Rodopaios
- Department of Social Medicine, Preventive Medicine and Nutrition Clinic, School of Medicine, University of Crete, Voutes, 71003 Iraklion, Greece
| | - Polyzois Makras
- Department of Endocrinology and Diabetes, 251 Hellenic Air Force & VA General Hospital, Athens, Greece; Department of Medical Research, 251 Hellenic Air Force & VA General Hospital, Athens, Greece
| | | | | | - Christos S Mantzoros
- Division of Endocrinology, Diabetes and Metabolism, Department of Internal Medicine, Boston VA Healthcare System and Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
4
|
Polyzos SA, Hill MA, Fuleihan GEH, Gnudi L, Kim YB, Larsson SC, Masuzaki H, Matarese G, Sanoudou D, Tena-Sempere M, Mantzoros CS. Metabolism, Clinical and Experimental: seventy years young and growing. Metabolism 2022; 137:155333. [PMID: 36244415 DOI: 10.1016/j.metabol.2022.155333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Accepted: 10/10/2022] [Indexed: 11/17/2022]
Affiliation(s)
- Stergios A Polyzos
- First Laboratory of Pharmacology, Medical School, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Michael A Hill
- Dalton Cardiovascular Research Center, Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, MO, USA
| | - Ghada El-Hajj Fuleihan
- Division of Endocrinology, Calcium Metabolism and Osteoporosis Program, World Health Organization Collaborating Center for Metabolic Bone Disorders, Department of Internal Medicine, American University of Beirut, Beirut, Lebanon
| | - Luigi Gnudi
- School of Cardiovascular and Metabolic Medicine & Sciences, King's College, London, UK
| | - Young-Bum Kim
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Susanna C Larsson
- Unit of Medical Epidemiology, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden; Unit of Cardiovascular and Nutritional Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Hiroaki Masuzaki
- Endocrinology, Diabetes and Metabolism, Hematology, Rheumatology, Second Department of Medicine, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan
| | - Giuseppe Matarese
- Treg Cell Lab, Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli "Federico II", Naples, Italy; Laboratorio di Immunogenetica dei Trapianti & Registro Regionale dei Trapianti di Midollo, AOU "Federico II", Naples, Italy; Laboratorio di Immunologia, Istituto per l'Endocrinologia e l'Oncologia Sperimentale Consiglio Nazionale delle Ricerche, Naples, Italy
| | - Despina Sanoudou
- Clinical Genomics and Pharmacogenomics Unit, 4th Department of Internal Medicine, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece; Biomedical Research Foundation of the Academy of Athens, Athens, Greece; Center for New Biotechnologies and Precision Medicine, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Manuel Tena-Sempere
- Instituto Maimónides de Investigación Biomédica de Cordoba (IMIBIC), Cordoba, Spain; Department of Cell Biology, Physiology and Immunology, University of Cordoba, Cordoba, Spain; CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Cordoba, Spain
| | - Christos S Mantzoros
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA; Section of Endocrinology, Boston VA Healthcare System, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
5
|
Valanti EK, Dalakoura-Karagkouni K, Fotakis P, Vafiadaki E, Mantzoros CS, Chroni A, Zannis V, Kardassis D, Sanoudou D. Reconstituted HDL-apoE3 promotes endothelial cell migration through ID1 and its downstream kinases ERK1/2, AKT and p38 MAPK. Metabolism 2022; 127:154954. [PMID: 34875308 DOI: 10.1016/j.metabol.2021.154954] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 11/15/2021] [Accepted: 11/30/2021] [Indexed: 12/16/2022]
Abstract
INTRODUCTION Atherosclerotic Coronary Artery Disease (ASCAD) is the leading cause of mortality worldwide. Novel therapeutic approaches aiming to improve the atheroprotective functions of High Density Lipoprotein (HDL) include the use of reconstituted HDL forms containing human apolipoprotein A-I (rHDL-apoA-I). Given the strong atheroprotective properties of apolipoprotein E3 (apoE3), rHDL-apoE3 may represent an attractive yet largely unexplored therapeutic agent. OBJECTIVE To evaluate the atheroprotective potential of rHDL-apoE3 starting with the unbiased assessment of global transcriptome effects and focusing on endothelial cell (EC) migration as a critical process in re-endothelialization and atherosclerosis prevention. The cellular, molecular and functional effects of rHDL-apoE3 on EC migration-associated pathways were assessed, as well as the potential translatability of these findings in vivo. METHODS Human Aortic ECs (HAEC) were treated with rHDL-apoE3 and total RNA was analyzed by whole genome microarrays. Expression and phosphorylation changes of key EC migration-associated molecules were validated by qRT-PCR and Western blot analysis in primary HAEC, Human Coronary Artery ECs (HCAEC) and the human EA.hy926 EC line. The capacity of rHDL-apoE3 to stimulate EC migration was assessed by wound healing and transwell migration assays. The contribution of MEK1/2, PI3K and the transcription factor ID1 in rHDL-apoE3-induced EC migration and activation of EC migration-related effectors was assessed using specific inhibitors (PD98059: MEK1/2, LY294002: PI3K) and siRNA-mediated gene silencing, respectively. The capacity of rHDL-apoE3 to improve vascular permeability and hypercholesterolemia in vivo was tested in a mouse model of hypercholesterolemia (apoE KO mice) using Evans Blue assays and lipid/lipoprotein analysis in the serum, respectively. RESULTS rHDL-apoE3 induced significant expression changes in 198 genes of HAEC mainly involved in re-endothelialization and atherosclerosis-associated functions. The most pronounced effect was observed for EC migration, with 42/198 genes being involved in the following EC migration-related pathways: 1) MEK/ERK, 2) PI3K/AKT/eNOS-MMP2/9, 3) RHO-GTPases, 4) integrin. rHDL-apoE3 induced changes in 24 representative transcripts of these pathways in HAEC, increasing the expression of their key proteins PIK3CG, EFNB2, ID1 and FLT1 in HCAEC and EA.hy926 cells. In addition, rHDL-apoE3 stimulated migration of HCAEC and EA.hy926 cells, and the migration was markedly attenuated in the presence of PD98059 or LY294002. rHDL-apoE3 also increased the phosphorylation of ERK1/2, AKT, eNOS and p38 MAPK in these cells, while PD98059 and LY294002 inhibited rHDL-apoE3-induced phosphorylation of ERK1/2, AKT and p38 MAPK, respectively. LY had no effect on rHDL-apoE3-mediated eNOS phosphorylation. ID1 siRNA markedly decreased EA.hy926 cell migration by inhibiting rHDL-apoE3-triggered ERK1/2 and AKT phosphorylation. Finally, administration of a single dose of rHDL-apoE3 in apoE KO mice markedly improved vascular permeability as demonstrated by the reduced concentration of Evans Blue dye in tissues such as the stomach, the tongue and the urinary bladder and ameliorated hypercholesterolemia. CONCLUSIONS rHDL-apoE3 significantly enhanced EC migration in vitro, predominantly via overexpression of ID1 and subsequent activation of MEK1/2 and PI3K, and their downstream targets ERK1/2, AKT and p38 MAPK, respectively, and improved vascular permeability in vivo. These novel insights into the rHDL-apoE3 functions suggest a potential clinical use to promote re-endothelialization and retard development of atherosclerosis.
Collapse
Affiliation(s)
- Eftaxia-Konstantina Valanti
- 4th Department of Internal Medicine, Clinical Genomics and Pharmacogenomics Unit, 'Attikon' Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece; Molecular Biology Division, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Katerina Dalakoura-Karagkouni
- Laboratory of Biochemistry, University of Crete Medical School, Heraklion, Greece; Division of Gene Regulation and Genomics, Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology of Hellas, Heraklion, Greece
| | | | - Elizabeth Vafiadaki
- Molecular Biology Division, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | | | - Angeliki Chroni
- Institute of Biosciences and Applications, National Center for Scientific Research "Demokritos", Athens, Greece
| | - Vassilis Zannis
- Molecular Genetics, Boston University Medical School, Boston, USA
| | - Dimitris Kardassis
- Laboratory of Biochemistry, University of Crete Medical School, Heraklion, Greece; Division of Gene Regulation and Genomics, Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology of Hellas, Heraklion, Greece
| | - Despina Sanoudou
- 4th Department of Internal Medicine, Clinical Genomics and Pharmacogenomics Unit, 'Attikon' Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece; Molecular Biology Division, Biomedical Research Foundation of the Academy of Athens, Athens, Greece; Center for New Biotechnologies and Precision Medicine, Medical School, National and Kapodistrian University of Athens, Athens, Greece.
| |
Collapse
|
6
|
Abstract
Age-associated changes in gene expression in skeletal muscle of healthy individuals reflect accumulation of damage and compensatory adaptations to preserve tissue integrity. To characterize these changes, RNA was extracted and sequenced from muscle biopsies collected from 53 healthy individuals (22-83 years old) of the GESTALT study of the National Institute on Aging-NIH. Expression levels of 57,205 protein-coding and non-coding RNAs were studied as a function of aging by linear and negative binomial regression models. From both models, 1134 RNAs changed significantly with age. The most differentially abundant mRNAs encoded proteins implicated in several age-related processes, including cellular senescence, insulin signaling, and myogenesis. Specific mRNA isoforms that changed significantly with age in skeletal muscle were enriched for proteins involved in oxidative phosphorylation and adipogenesis. Our study establishes a detailed framework of the global transcriptome and mRNA isoforms that govern muscle damage and homeostasis with age.
Collapse
|
7
|
Khanal P, He L, Herbert AJ, Stebbings GK, Onambele-Pearson GL, Degens H, Morse CI, Thomis M, Williams AG. The Association of Multiple Gene Variants with Ageing Skeletal Muscle Phenotypes in Elderly Women. Genes (Basel) 2020; 11:genes11121459. [PMID: 33291384 PMCID: PMC7762041 DOI: 10.3390/genes11121459] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 11/24/2020] [Accepted: 12/03/2020] [Indexed: 12/20/2022] Open
Abstract
There is a scarcity of studies that have investigated the role of multiple single nucleotide polymorphisms (SNPs) on a range of muscle phenotypes in an elderly population. The present study investigated the possible association of 24 SNPs with skeletal muscle phenotypes in 307 elderly Caucasian women (aged 60–91 years, 66.3 ± 11.3 kg). Skeletal muscle phenotypes included biceps brachii thickness, vastus lateralis cross-sectional areas, maximal hand grip strength, isometric knee extension and elbow flexion torque. Genotyping for 24 SNPs, chosen on their skeletal muscle structural or functional links, was conducted on DNA extracted from blood or saliva. Of the 24 SNPs, 10 were associated with at least one skeletal muscle phenotype. HIF1A rs11549465 was associated with three skeletal muscle phenotypes and PTK2 rs7460 and ACVR1B rs10783485 were each associated with two phenotypes. PTK2 rs7843014, COL1A1 rs1800012, CNTF rs1800169, NOS3 rs1799983, MSTN rs1805086, TRHR rs7832552 and FTO rs9939609 were each associated with one. Elderly women possessing favourable genotypes were 3.6–13.2% stronger and had 4.6–14.7% larger muscle than those with less favourable genotypes. These associations, together with future work involving a broader range of SNPs, may help identify individuals at particular risk of an age-associated loss of independence.
Collapse
Affiliation(s)
- Praval Khanal
- Musculoskeletal Science and Sports Medicine Research Centre, Department of Sport and Exercise Sciences, Manchester Metropolitan University, Manchester M15 6BH, UK; (L.H.); (G.K.S.); (G.L.O.-P.); (C.I.M.); (A.G.W.)
- Department of Movement Sciences, Physical Activity, Sports & Health Research Group, KU Leuven, 3001 Leuven, Belgium;
- Correspondence: ; Tel.: +977-9841528705
| | - Lingxiao He
- Musculoskeletal Science and Sports Medicine Research Centre, Department of Sport and Exercise Sciences, Manchester Metropolitan University, Manchester M15 6BH, UK; (L.H.); (G.K.S.); (G.L.O.-P.); (C.I.M.); (A.G.W.)
- Department of Movement Sciences, Physical Activity, Sports & Health Research Group, KU Leuven, 3001 Leuven, Belgium;
| | - Adam J. Herbert
- Department of Sport and Exercise, Birmingham City University, Birmingham B5 5JU, UK;
| | - Georgina K. Stebbings
- Musculoskeletal Science and Sports Medicine Research Centre, Department of Sport and Exercise Sciences, Manchester Metropolitan University, Manchester M15 6BH, UK; (L.H.); (G.K.S.); (G.L.O.-P.); (C.I.M.); (A.G.W.)
| | - Gladys L. Onambele-Pearson
- Musculoskeletal Science and Sports Medicine Research Centre, Department of Sport and Exercise Sciences, Manchester Metropolitan University, Manchester M15 6BH, UK; (L.H.); (G.K.S.); (G.L.O.-P.); (C.I.M.); (A.G.W.)
| | - Hans Degens
- Department of Life Sciences, Manchester Metropolitan University, Manchester M15 6BH, UK;
- Institute of Sport Science and Innovations, Lithuanian Sports University, LT-44221 Kaunsas, Lithuania
- Pharmacy of Targu Mures, University of Medicine, 540142 Targu Mures, Romania
| | - Christopher I. Morse
- Musculoskeletal Science and Sports Medicine Research Centre, Department of Sport and Exercise Sciences, Manchester Metropolitan University, Manchester M15 6BH, UK; (L.H.); (G.K.S.); (G.L.O.-P.); (C.I.M.); (A.G.W.)
| | - Martine Thomis
- Department of Movement Sciences, Physical Activity, Sports & Health Research Group, KU Leuven, 3001 Leuven, Belgium;
| | - Alun G. Williams
- Musculoskeletal Science and Sports Medicine Research Centre, Department of Sport and Exercise Sciences, Manchester Metropolitan University, Manchester M15 6BH, UK; (L.H.); (G.K.S.); (G.L.O.-P.); (C.I.M.); (A.G.W.)
- Institute of Sport, Exercise and Health, University College London, London W1T 7HA, UK
| |
Collapse
|
8
|
Perugini J, Di Mercurio E, Tossetta G, Severi I, Monaco F, Reguzzoni M, Tomasetti M, Dani C, Cinti S, Giordano A. Biological Effects of Ciliary Neurotrophic Factor on hMADS Adipocytes. Front Endocrinol (Lausanne) 2019; 10:768. [PMID: 31781039 PMCID: PMC6861295 DOI: 10.3389/fendo.2019.00768] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 10/22/2019] [Indexed: 12/15/2022] Open
Abstract
Administration of ciliary neurotrophic factor (CNTF) to experimental animals exerts anti-obesity effects by acting on multiple targets. In white adipose tissue CNTF reduces lipid content, promotes fatty acid (FA) oxidation and improves insulin sensitivity. This study was performed to establish whether CNTF exerts similar effects on human white adipocytes. To this end, adipose differentiation was induced in vitro in human multipotent adipose-derived stem (hMADS) cells. CNTF receptor α (CNTFRα) expression was assessed in hMADS cells and adipocytes by qRT-PCR, Western blotting, and immunocytochemistry. After administration of human recombinant CNTF, signaling pathways and gene expression were evaluated by Western blotting and qRT-PCR. Glucose uptake was assessed by measuring 2-nitrobenzodeoxyglucose uptake with a fluorescence plate reader. Lastly, CNTF-induced anti-inflammatory responses were evaluated in hMADS adipocytes stressed with tumor necrosis factor α (TNFα) for 24 h. Results showed that CNTFRα protein expression was higher in undifferentiated hMADS cells than in hMADS adipocytes, where it was however clearly detectable. In hMADS adipocytes, 1 nM CNTF strongly activated the JAK-STAT3 (Janus kinase-signaling transducer and activator of transcription 3) pathway and acutely and transiently activated the AMPK (AMP-activated protein kinase) and AKT (protein kinase B) pathways. Acute CNTF treatment for 20 min significantly increased basal glucose uptake and was associated with increased AKT phosphorylation. Longer-term (24 and 48 h) treatment reduced the expression of lipogenic markers (FA synthase and sterol regulatory element-binding protein-1) and increased the expression of lipolytic [hormone-sensitive lipase (HSL) and adipose triglyceride lipase (ATGL)] and mitochondrial (peroxisome proliferator-activated receptor γ coactivator-1α and carnitine palmitoyltransferase 1) markers. In TNFα-treated hMADS adipocytes, CNTF significantly reduced the expression of monocyte chemoattractant protein 1 and TNFα-induced AKT inhibition. Collectively, these findings demonstrate for the first time that CNTF plays a role also in human adipocytes, driving their metabolism toward a less lipid-storing and more energy-consuming phenotype.
Collapse
Affiliation(s)
- Jessica Perugini
- Department of Experimental and Clinical Medicine, Marche Polytechnic University, Ancona, Italy
| | - Eleonora Di Mercurio
- Department of Experimental and Clinical Medicine, Marche Polytechnic University, Ancona, Italy
| | - Giovanni Tossetta
- Department of Experimental and Clinical Medicine, Marche Polytechnic University, Ancona, Italy
| | - Ilenia Severi
- Department of Experimental and Clinical Medicine, Marche Polytechnic University, Ancona, Italy
| | - Federica Monaco
- Department of Clinical and Molecular Sciences, Marche Polytechnic University, Ancona, Italy
| | - Marcella Reguzzoni
- Department of Surgical and Morphological Sciences, University of Insubria, Varese, Italy
| | - Marco Tomasetti
- Department of Clinical and Molecular Sciences, Marche Polytechnic University, Ancona, Italy
| | - Christian Dani
- Université Côte d'Azur, CNRS, INSERM, iBV, Faculté de Médecine, Nice, France
| | - Saverio Cinti
- Department of Experimental and Clinical Medicine, Marche Polytechnic University, Ancona, Italy
- Center of Obesity, United Hospitals, Marche Polytechnic University, Ancona, Italy
| | - Antonio Giordano
- Department of Experimental and Clinical Medicine, Marche Polytechnic University, Ancona, Italy
| |
Collapse
|
9
|
Cui W, Liu CX, Wang J, Zhang YC, Shen Q, Feng ZH, Wu J, Li JX. An oleanolic acid derivative reduces denervation-induced muscle atrophy via activation of CNTF-mediated JAK2/STAT3 signaling pathway. Eur J Pharmacol 2019; 861:172612. [PMID: 31421088 DOI: 10.1016/j.ejphar.2019.172612] [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: 04/17/2019] [Revised: 08/10/2019] [Accepted: 08/14/2019] [Indexed: 02/07/2023]
Abstract
Denervation caused by sciatic nerve injury has brought great harm to the patients, especially denervation-induced muscle atrophy. The body stress produces a large number of Schwann cells when the sciatic nerve is injured, and the cells secrete some cytokines including ciliary neurotrophic factor (CNTF) that not only play a role in promoting the repair of sciatic nerve, but also maintain the normal physiological function of the muscles surrounding the damaged nerves. CNTF upregulates janus kinase 2 (JAK2) and signal transducers and activators of transcription 3 (STAT3) signals in myoblasts, and consequently accelerates the proliferation and differentiation of myoblasts. This effect on myoblasts is the most effective way to relieve muscle atrophy. Therefore, increasing CNTF is a promising direction to improve muscle atrophy. In the present study, an oleanolic acid derivative, HA-19, increased the proliferation of Schwann cells, and elevated CNTF production of the cells. HA-19 up-regulated the phosphorylation of JAK2 and STAT3 not only by directly acting on myoblasts, but also by increasing the secretion of CNTF of Schwann cells; and consequently, promoted the proliferation and differentiation of myoblasts. In denervation-induced muscle atrophy mice model, treatment with HA-19 significantly increased the weights of tibialis anterior (TA), gastrocnemius (Gastroc.), extensor digitorum longus (EDL), soleus and quadriceps (Quad.) under atrophied state. And, very interestingly, these muscles under normal condition were also strengthened by HA-19. Our finding demonstrated that HA-19 has a great potential as a lead compound for the drug discovery of anti-denervation-induced muscle atrophy.
Collapse
Affiliation(s)
- Wei Cui
- State Key Laboratory of Analytical Chemistry for Life Science and Collaborative Innovation Center of Chemistry for Life Sciences, Jiangsu Key Laboratory of Advanced Organic Materials, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| | - Chen-Xi Liu
- State Key Laboratory of Analytical Chemistry for Life Science and Collaborative Innovation Center of Chemistry for Life Sciences, Jiangsu Key Laboratory of Advanced Organic Materials, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| | - Jie Wang
- State Key Laboratory of Analytical Chemistry for Life Science and Collaborative Innovation Center of Chemistry for Life Sciences, Jiangsu Key Laboratory of Advanced Organic Materials, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| | - Yu-Chao Zhang
- State Key Laboratory of Analytical Chemistry for Life Science and Collaborative Innovation Center of Chemistry for Life Sciences, Jiangsu Key Laboratory of Advanced Organic Materials, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| | - Qi Shen
- State Key Laboratory of Analytical Chemistry for Life Science and Collaborative Innovation Center of Chemistry for Life Sciences, Jiangsu Key Laboratory of Advanced Organic Materials, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| | - Zhen-Hua Feng
- The Center of Diagnosis and Treatment for Joint Disease, Drum Tower Hospital Affiliated to Medical School of Nanjing University, Nanjing, 210008, China
| | - Jing Wu
- State Key Laboratory of Analytical Chemistry for Life Science and Collaborative Innovation Center of Chemistry for Life Sciences, Jiangsu Key Laboratory of Advanced Organic Materials, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China.
| | - Jian-Xin Li
- State Key Laboratory of Analytical Chemistry for Life Science and Collaborative Innovation Center of Chemistry for Life Sciences, Jiangsu Key Laboratory of Advanced Organic Materials, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China.
| |
Collapse
|
10
|
Kalozoumi G, Kel-Margoulis O, Vafiadaki E, Greenberg D, Bernard H, Soreq H, Depaulis A, Sanoudou D. Glial responses during epileptogenesis in Mus musculus point to potential therapeutic targets. PLoS One 2018; 13:e0201742. [PMID: 30114263 PMCID: PMC6095496 DOI: 10.1371/journal.pone.0201742] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Accepted: 07/21/2018] [Indexed: 01/21/2023] Open
Abstract
The Mesio-Temporal Lobe Epilepsy syndrome is the most common form of intractable epilepsy. It is characterized by recurrence of focal seizures and is often associated with hippocampal sclerosis and drug resistance. We aimed to characterize the molecular changes occurring during the initial stages of epileptogenesis in search of new therapeutic targets for Mesio-Temporal Lobe Epilepsy. We used a mouse model obtained by intra-hippocampal microinjection of kainate and performed hippocampal whole genome expression analysis at 6h, 12h and 24h post-injection, followed by multilevel bioinformatics analysis. We report significant changes in immune and inflammatory responses, neuronal network reorganization processes and glial functions, predominantly initiated during status epilepticus at 12h and persistent after the end of status epilepticus at 24h post-kainate. Upstream regulator analysis highlighted Cyba, Cybb and Vim as central regulators of multiple overexpressed genes implicated in glial responses at 24h. In silico microRNA analysis indicated that miR-9, miR-19b, miR-129, and miR-223 may regulate the expression of glial-associated genes at 24h. Our data support the hypothesis that glial-mediated inflammatory response holds a key role during epileptogenesis, and that microglial cells may participate in the initial process of epileptogenesis through increased ROS production via the NOX complex.
Collapse
Affiliation(s)
- Georgia Kalozoumi
- Clinical Genomics and Pharmacogenomics Unit, 4 Department of Internal Medicine, Attikon Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | | | - Elizabeth Vafiadaki
- Molecular Biology Division, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - David Greenberg
- The Edmond and Lily Safra Center for Brain Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
| | | | - Hermona Soreq
- The Edmond and Lily Safra Center for Brain Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Antoine Depaulis
- INSERM, Grenoble, France
- Univ. Grenoble Alpes, Grenoble Institut des Neurosciences, Grenoble, France
- CHU de Grenoble, Hôpital Michallon, Grenoble, France
| | - Despina Sanoudou
- Clinical Genomics and Pharmacogenomics Unit, 4 Department of Internal Medicine, Attikon Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
- Molecular Biology Division, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
- * E-mail:
| |
Collapse
|
11
|
Liu H, Jiang W, Chen X, Chang G, Zhao L, Li X, Zhang H. Skeletal muscle-specific Sidt2 knockout in mice induced muscular dystrophy-like phenotype. Metabolism 2018; 85:259-270. [PMID: 29752955 DOI: 10.1016/j.metabol.2018.05.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2018] [Revised: 04/21/2018] [Accepted: 05/07/2018] [Indexed: 02/06/2023]
Abstract
BACKGROUND Sidt2 is an integral lysosomal membrane protein. Previously, we generated a Sidt2 global knockout mouse and found impaired insulin secretion, along with skeletal muscle pathology. METHODS A mouse model with a muscle-specific knockout of the Sidt2 gene (Sidt2f/fCre) had been generated, to which extensive morphologic study as well as functional study was applied to investigate the direct role of Sidt2 on skeletal muscle tissue in vivo. Secondly, the autophagy-lysosomal pathway was examined by Western blot and immunostaining. Additionally, RNA expression changes in Sidt2f/fCre mice were analyzed by genechip. RESULTS Sidt2 deficiency in skeletal muscle results in pathognomonic hallmarks of muscular dystrophy, including muscle mass decrease, muscle weakness, fibrosis, central nucleation, fiber regeneration, mildly elevated serum creatine kinase, and dramatically elevated sarcolipin mRNA. Along with accumulation of autophagolysomes, LC3-II, adaptor protein p62, ubiquitinated aggregates, and Lamp2-positive vacuoles were increased significantly in Sidt2f/fCre skeletal muscle fibers. However, only lysosomal-related genes were upregulated, while the genes upstream of the autophagy pathway were unchanged. Simultaneously, the proteasome chymotryptic activity and the lysosomal soluble enzyme activity were unimpaired, which largely excluded the possibility of proteasome chymotryptic activity defect and the lysosomal soluble enzyme defect leading to ubiquitinated aggregates accumulation. CONCLUSION We concluded that Sidt2 deficiency leads to muscular dystrophy-like phenotype in mice and Sidt2 plays a critical role in the late stage of autophagy.
Collapse
Affiliation(s)
- Huan Liu
- Pediatric Endocrinology and Genetic Metabolism, Xinhua Hospital, Shanghai Institute for Pediatric Research, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wenjun Jiang
- Pediatric Endocrinology and Genetic Metabolism, Xinhua Hospital, Shanghai Institute for Pediatric Research, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xueru Chen
- Pediatric Endocrinology and Genetic Metabolism, Xinhua Hospital, Shanghai Institute for Pediatric Research, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Guoying Chang
- Pediatric Endocrinology and Genetic Metabolism, Xinhua Hospital, Shanghai Institute for Pediatric Research, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lei Zhao
- Department of Neurology, Children's Hospital of Fudan University, Shanghai, China
| | - Xihua Li
- Department of Neurology, Children's Hospital of Fudan University, Shanghai, China.
| | - Huiwen Zhang
- Pediatric Endocrinology and Genetic Metabolism, Xinhua Hospital, Shanghai Institute for Pediatric Research, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
12
|
Cerquone Perpetuini A, Re Cecconi AD, Chiappa M, Martinelli GB, Fuoco C, Desiderio G, Castagnoli L, Gargioli C, Piccirillo R, Cesareni G. Group I Paks support muscle regeneration and counteract cancer-associated muscle atrophy. J Cachexia Sarcopenia Muscle 2018; 9:727-746. [PMID: 29781585 PMCID: PMC6104114 DOI: 10.1002/jcsm.12303] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Revised: 03/02/2018] [Accepted: 03/06/2018] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Skeletal muscle is characterized by an efficient regeneration potential that is often impaired during myopathies. Understanding the molecular players involved in muscle homeostasis and regeneration could help to find new therapies against muscle degenerative disorders. Previous studies revealed that the Ser/Thr kinase p21 protein-activated kinase 1 (Pak1) was specifically down-regulated in the atrophying gastrocnemius of Yoshida hepatoma-bearing rats. In this study, we evaluated the role of group I Paks during cancer-related atrophy and muscle regeneration. METHODS We examined Pak1 expression levels in the mouse Tibialis Anterior muscles during cancer cachexia induced by grafting colon adenocarcinoma C26 cells and in vitro by dexamethasone treatment. We investigated whether the overexpression of Pak1 counteracts muscle wasting in C26-bearing mice and in vitro also during interleukin-6 (IL6)-induced or dexamethasone-induced C2C12 atrophy. Moreover, we analysed the involvement of group I Paks on myogenic differentiation in vivo and in vitro using the group I chemical inhibitor IPA-3. RESULTS We found that Pak1 expression levels are reduced during cancer-induced cachexia in the Tibialis Anterior muscles of colon adenocarcinoma C26-bearing mice and in vitro during dexamethasone-induced myotube atrophy. Electroporation of muscles of C26-bearing mice with plasmids directing the synthesis of PAK1 preserves fiber size in cachectic muscles by restraining the expression of atrogin-1 and MuRF1 and possibly by inducing myogenin expression. Consistently, the overexpression of PAK1 reduces the dexamethasone-induced expression of MuRF1 in myotubes and increases the phospho-FOXO3/FOXO3 ratio. Interestingly, the ectopic expression of PAK1 counteracts atrophy in vitro by restraining the IL6-Stat3 signalling pathway measured in luciferase-based assays and by reducing rates of protein degradation in atrophying myotubes exposed to IL6. On the other hand, we observed that the inhibition of group I Paks has no effect on myotube atrophy in vitro and is associated with impaired muscle regeneration in vivo and in vitro. In fact, we found that mice treated with the group I inhibitor IPA-3 display a delayed recovery from cardiotoxin-induced muscle injury. This is consistent with in vitro experiments showing that IPA-3 impairs myogenin expression and myotube formation in vessel-associated myogenic progenitors, C2C12 myoblasts, and satellite cells. Finally, we observed that IPA-3 reduces p38α/β phosphorylation that is required to proceed through various stages of satellite cells differentiation: activation, asymmetric division, and ultimately myotube formation. CONCLUSIONS Our data provide novel evidence that is consistent with group I Paks playing a central role in the regulation of muscle homeostasis, atrophy and myogenesis.
Collapse
Affiliation(s)
| | - Andrea David Re Cecconi
- Department of NeurosciencesIRCCS‐Mario Negri Institute for Pharmacological ResearchVia Giuseppe La Masa20156MilanItaly
| | - Michela Chiappa
- Department of NeurosciencesIRCCS‐Mario Negri Institute for Pharmacological ResearchVia Giuseppe La Masa20156MilanItaly
| | - Giulia Benedetta Martinelli
- Department of NeurosciencesIRCCS‐Mario Negri Institute for Pharmacological ResearchVia Giuseppe La Masa20156MilanItaly
| | - Claudia Fuoco
- Department of BiologyUniversity of Rome Tor VergataVia della ricerca scientifica00133RomeItaly
| | - Giovanni Desiderio
- Department of BiologyUniversity of Rome Tor VergataVia della ricerca scientifica00133RomeItaly
| | - Luisa Castagnoli
- Department of BiologyUniversity of Rome Tor VergataVia della ricerca scientifica00133RomeItaly
| | - Cesare Gargioli
- Department of BiologyUniversity of Rome Tor VergataVia della ricerca scientifica00133RomeItaly
| | - Rosanna Piccirillo
- Department of NeurosciencesIRCCS‐Mario Negri Institute for Pharmacological ResearchVia Giuseppe La Masa20156MilanItaly
| | - Gianni Cesareni
- Department of BiologyUniversity of Rome Tor VergataVia della ricerca scientifica00133RomeItaly
| |
Collapse
|
13
|
Perakakis N, Farr OM, Tuccinardi D, Upadhyay J, Mantzoros CS. Research advances in metabolism 2016. Metabolism 2017; 67:41-53. [PMID: 28081777 PMCID: PMC5871911 DOI: 10.1016/j.metabol.2016.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Revised: 10/30/2016] [Accepted: 11/02/2016] [Indexed: 11/17/2022]
Affiliation(s)
- Nikolaos Perakakis
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.
| | - Olivia M Farr
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Dario Tuccinardi
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Jagriti Upadhyay
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA; Section of Endocrinology, Boston VA Healthcare System, Harvard Medical School, Boston, MA 02130, USA
| | - Christos S Mantzoros
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA; Section of Endocrinology, Boston VA Healthcare System, Harvard Medical School, Boston, MA 02130, USA
| |
Collapse
|
14
|
Anastasilakis AD, Polyzos SA, Skouvaklidou EC, Kynigopoulos G, Saridakis ZG, Apostolou A, Triantafyllou GA, Karagiozoglou-Lampoudi T, Mantzoros CS. Circulating follistatin displays a day-night rhythm and is associated with muscle mass and circulating leptin levels in healthy, young humans. Metabolism 2016; 65:1459-65. [PMID: 27621181 PMCID: PMC5022793 DOI: 10.1016/j.metabol.2016.07.002] [Citation(s) in RCA: 21] [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: 05/13/2016] [Revised: 07/01/2016] [Accepted: 07/04/2016] [Indexed: 12/22/2022]
Abstract
PURPOSE Follistatin may affect lean and fat mass and be implicated in metabolic diseases. We aimed to elucidate physiological predictors of circulating follistatin variation in healthy young humans. PROCEDURES This was an observational, cross-sectional study with two additional prospective observational arms (circadian, seasonal sub-studies) and one prospective interventional arm (mixed meal sub-study). Healthy, young individuals of both sexes (n=122) were subjected to anthropometric and body composition measurements and their eating and exercise behavior profiles were assessed by validated questionnaires. Sub-groups were subjected to standardized meal ingestion (n=36), day-night rhythm (n=20) and seasonal variation (n=20) studies. Main outcome of the study were circulating follistatin levels. RESULTS At baseline follistatin levels were correlated with creatinine (r=0.24; p=0.01), creatine phosphokinase (rs=0.22; p=0.02), and with lean body mass (rs=0.19; p=0.04) and were higher in males than females (p=0.004) after adjustment for leptin, which was its major predictor. Follistatin levels showed a circadian (p<0.001), but not a seasonal, variation, and were also affected by the phase of menstrual cycle in females (p=0.034). Follistatin levels were not affected by dietary or exercise habits but levels increased after a standardized meal ingestion (250kcal) (p=0.002). CONCLUSIONS In healthy young individuals circulating follistatin levels are correlated with muscle mass. Follistatin levels are associated with circulating leptin levels and display a day-night rhythm and a menstrual cycle, but not a seasonal, variation.
Collapse
Affiliation(s)
| | - Stergios A Polyzos
- Department of Medicine, Aristotle University of Thessaloniki, Ippokration General Hospital, Thessaloniki, Greece
| | | | | | | | - Aggeliki Apostolou
- Nutrition-Dietetics Department, Alexander Technological Education Institute of Thessaloniki, Central Macedonia, Greece
| | - Georgios A Triantafyllou
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Thomai Karagiozoglou-Lampoudi
- Nutrition-Dietetics Department, Alexander Technological Education Institute of Thessaloniki, Central Macedonia, Greece
| | - Christos S Mantzoros
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| |
Collapse
|