1
|
Niu J, Han B, Wang S, Wei Y, Liu Z, Yang H. Hyperglycemia-induced α-Klotho downregulation impairs mitophagy and promotes placental dysfunction in T2DM pregnancies. Placenta 2025; 167:95-103. [PMID: 40349475 DOI: 10.1016/j.placenta.2025.04.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Revised: 04/16/2025] [Accepted: 04/26/2025] [Indexed: 05/14/2025]
Abstract
INTRODUCTION Placental dysfunction in pregnancies complicated by type 2 diabetes mellitus (T2DM) is associated with adverse maternal and fetal outcomes. α-Klotho, a multifunctional anti-aging protein, plays a critical role in maintaining cellular homeostasis, but its role in T2DM-induced placental dysfunction remains poorly understood. METHODS Placental tissues from T2DM pregnancies and normoglycemic controls were analyzed for α-Klotho expression using qRT-PCR, Western blot, and immunohistochemistry. BeWo trophoblast cells were cultured under normoglycemic and hyperglycemic conditions, with α-Klotho knockdown and overexpression to explore its regulatory effects. Transcriptomic analysis was conducted to identify affected pathways, and markers of mitophagy and reactive oxygen species (ROS) were analyzed. RESULTS α-Klotho expression was significantly reduced in the placentas of T2DM pregnancies and in trophoblast cells under hyperglycemic conditions. Transcriptomic analysis identified pathways related to mitochondrial dysfunction and impaired mitophagy as key processes regulated by α-Klotho. Hyperglycemia and α-Klotho knockdown suppressed mitophagy, while ROS production was increased, further exacerbating oxidative stress. Overexpression of α-Klotho restored mitophagy and mitigated ROS activation. DISCUSSION This study reveals that α-Klotho downregulation contributes to T2DM-induced placental dysfunction by impairing mitophagy and increasing oxidative stress. These findings provide new insights into the molecular mechanisms underlying placental abnormalities in diabetic pregnancies and highlight α-Klotho as a potential therapeutic target.
Collapse
Affiliation(s)
- Jianhua Niu
- Department of Obstetrics and Gynaecology, Peking University First Hospital, Beijing, 100034, China; Beijing Key Laboratory of Maternal Foetal Medicine of Gestational Diabetes Mellitus, Beijing, 100034, China
| | - Bing Han
- Department of Obstetrics and Gynaecology, Peking University First Hospital, Beijing, 100034, China; Beijing Key Laboratory of Maternal Foetal Medicine of Gestational Diabetes Mellitus, Beijing, 100034, China; Peking University, Beijing, 100034, China
| | - Shuxian Wang
- Department of Obstetrics and Gynaecology, Peking University First Hospital, Beijing, 100034, China; Beijing Key Laboratory of Maternal Foetal Medicine of Gestational Diabetes Mellitus, Beijing, 100034, China
| | - Yumei Wei
- Department of Obstetrics and Gynaecology, Peking University First Hospital, Beijing, 100034, China; Beijing Key Laboratory of Maternal Foetal Medicine of Gestational Diabetes Mellitus, Beijing, 100034, China; Peking University, Beijing, 100034, China
| | - Zhe Liu
- Department of Obstetrics and Gynaecology, Peking University First Hospital, Beijing, 100034, China; Beijing Key Laboratory of Maternal Foetal Medicine of Gestational Diabetes Mellitus, Beijing, 100034, China.
| | - Huixia Yang
- Department of Obstetrics and Gynaecology, Peking University First Hospital, Beijing, 100034, China; Beijing Key Laboratory of Maternal Foetal Medicine of Gestational Diabetes Mellitus, Beijing, 100034, China; Peking University, Beijing, 100034, China.
| |
Collapse
|
2
|
Roig-Soriano J, Edo Á, Verdés S, Martín-Alonso C, Sánchez-de-Diego C, Rodriguez-Estevez L, Serrano AL, Abraham CR, Bosch A, Ventura F, Jordan BA, Muñoz-Cánoves P, Chillón M. Long-term effects of s-KL treatment in wild-type mice: Enhancing longevity, physical well-being, and neurological resilience. Mol Ther 2025; 33:1449-1465. [PMID: 39988871 PMCID: PMC11997498 DOI: 10.1016/j.ymthe.2025.02.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 09/30/2024] [Accepted: 02/19/2025] [Indexed: 02/25/2025] Open
Abstract
Aging is a major risk factor for pathologies including sarcopenia, osteoporosis, and cognitive decline, which bring suffering, disability, and elevated economic and social costs. Therefore, new therapies are needed to achieve healthy aging. The protein Klotho (KL) has emerged as a promising anti-aging molecule due to its pleiotropic actions modulating insulin, insulin-like growth factor-1, and Wnt signaling pathways and reducing inflammatory and oxidative stress. Here, we explored the anti-aging potential of the secreted isoform of this protein on the non-pathological aging progression of wild-type mice. The delivery of an adeno-associated virus serotype 9 (AAV9) coding for secreted KL (s-KL) efficiently increased the concentration of s-KL in serum, resulting in a 20% increase in lifespan. Notably, KL treatment improved physical fitness, related to a reduction in muscle fibrosis and an increase in muscular regenerative capacity. KL treatment also improved bone microstructural parameters associated with osteoporosis. Finally, s-KL-treated mice exhibited increased cellular markers of adult neurogenesis and immune response, with transcriptomic analysis revealing induced phagocytosis and immune cell activity in the aged hippocampus. These results show the potential of elevating s-KL expression to simultaneously reduce the age-associated degeneration in multiple organs, increasing both life and health span.
Collapse
Affiliation(s)
- Joan Roig-Soriano
- Institut de Neurociènces (INc), Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain; Vall d'Hebron Institut de Recerca (VHIR), 08035 Barcelona, Spain
| | - Ángel Edo
- Institut de Neurociènces (INc), Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain; Vall d'Hebron Institut de Recerca (VHIR), 08035 Barcelona, Spain
| | - Sergi Verdés
- Institut de Neurociènces (INc), Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain; Vall d'Hebron Institut de Recerca (VHIR), 08035 Barcelona, Spain
| | - Carlos Martín-Alonso
- Institut de Neurociènces (INc), Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
| | | | - Laura Rodriguez-Estevez
- Institut de Neurociènces (INc), Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain; Vall d'Hebron Institut de Recerca (VHIR), 08035 Barcelona, Spain
| | - Antonio L Serrano
- Department of Medicine and Life Sciences, Pompeu Fabra University, 08003 Barcelona, Spain; Altos Labs, San Diego Institute of Science, San Diego, CA 92122, USA
| | | | - Assumpció Bosch
- Institut de Neurociènces (INc), Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain; Vall d'Hebron Institut de Recerca (VHIR), 08035 Barcelona, Spain; Ciberned, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | | | - Bryen A Jordan
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, New York, NY 10461, USA
| | - Pura Muñoz-Cánoves
- Department of Medicine and Life Sciences, Pompeu Fabra University, 08003 Barcelona, Spain; ICREA, 08010 Barcelona, Spain; Altos Labs, San Diego Institute of Science, San Diego, CA 92122, USA
| | - Miguel Chillón
- Institut de Neurociènces (INc), Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain; Vall d'Hebron Institut de Recerca (VHIR), 08035 Barcelona, Spain; ICREA, 08010 Barcelona, Spain; Unitat de Producció de Vectors (UPV), Universitat Autònoma de Barcelona, 08193 Barcelona, Spain.
| |
Collapse
|
3
|
Zavvari Oskuye Z, Mehri K, Khalilpour J, Nemati S, Hosseini L, Bafadam S, Abdollahzade N, Badalzadeh R. Klotho in age-related cardiovascular diseases: Insights into mitochondrial dysfunction and cell death. IJC HEART & VASCULATURE 2025; 57:101629. [PMID: 40129656 PMCID: PMC11930703 DOI: 10.1016/j.ijcha.2025.101629] [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: 10/19/2024] [Revised: 01/26/2025] [Accepted: 02/08/2025] [Indexed: 03/26/2025]
Abstract
Aging is a major risk factor for the development of cardiovascular diseases (CVD), leading to specific alterations in the heart and vasculature. Besides, the mechanisms and intracellular pathways of aging and the factors affecting it are still not completely clear. Age-related complications such as oxidative stress, decreased autophagy, mitochondrial dysfunction, inflammatory responses, and cardiac dysfunction are associated with relative Klotho deficiency. Klotho, an anti-aging protein, with anti-oxidative and anti-inflammatory properties, has been shown to modulate calcium regulation and autophagy. It also protects against endothelial dysfunction by increasing nitric oxide production. Furthermore, emerging research has revealed that klotho significantly impacts vascular smooth muscle cells (VSMC) energetics and survival. This article has focused on recent advances in using Klotho in age-related CVD and summarizes the pre-clinical evidence supporting this approach. Based on the research, Klotho could provide more therapeutic options for ameliorating aging-related CVD.
Collapse
Affiliation(s)
- Zohreh Zavvari Oskuye
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Keyvan Mehri
- Department of Basic Medical Sciences, Khoy University of Medical Sciences, Khoy, Iran
| | - Jamal Khalilpour
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Samira Nemati
- Student Research Committee, Semnan University of Medical Sciences, Semnan, Iran
| | - Leila Hosseini
- Research Center of Psychiatry and Behavioral Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Soleyman Bafadam
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Naseh Abdollahzade
- Neurophysiology Research Center, Urmia University of Medical Sciences, Urmia, Iran
| | - Reza Badalzadeh
- Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Physiology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
4
|
Xu Y, You J, Yao J, Hou B, Wang W, Hao Z. Klotho alleviates oxidative stress and mitochondrial dysfunction through the Nrf2/HO-1 pathway, thereby reducing renal senescence induced by calcium oxalate crystals. Urolithiasis 2025; 53:61. [PMID: 40156629 DOI: 10.1007/s00240-025-01734-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Accepted: 03/15/2025] [Indexed: 04/01/2025]
Abstract
Klotho is an antiaging protein that is primarily secreted by the kidneys. This study aimed to explore the protective effects of Klotho against calcium oxalate (CaOx) crystal-induced renal aging and the underlying mechanisms involved. We established a mouse model of CaOx crystal deposition via the intraperitoneal injection of glyoxylate (Gly) and constructed an in vitro model by stimulating HK2 cells with calcium oxalate monohydrate (COM). Renal aging levels were assessed through β-galactosidase (SA-β-gal) staining and the detection of senescence-associated markers. By overexpressing Klotho both in vitro and in vivo, we examined oxidative stress, mitochondrial function, and renal aging levels. We then evaluated the role of Nrf2/HO-1 signalling pathway-mediated oxidative stress in CaOx crystal-induced renal aging by applying the oxidative stress scavenger N-acetylcysteine (NAC) and overexpressing or inhibiting Nrf2 in HK2 cells. We subsequently overexpressed Klotho while inhibiting Nrf2 to confirm that Klotho exerts its protective effects through the Nrf2/HO-1 pathway. Finally, we measured the methylation levels of the Klotho promoter and assessed the degree of renal aging induced by CaOx crystals after the inhibition of Klotho DNA methylation. We found that the overexpression of Klotho alleviated CaOx crystal-induced oxidative stress and mitochondrial dysfunction, thereby reducing renal aging. NAC mitigated CaOx crystal-induced renal aging. The overexpression of Nrf2 alleviated CaOx crystal-induced oxidative stress and mitochondrial dysfunction, thus reducing renal aging, whereas the knockdown of Nrf2 exacerbated CaOx crystal-induced oxidative stress and mitochondrial dysfunction, leading to more severe renal aging. The combination of Klotho overexpression and Nrf2 knockdown reversed the protective effects of Klotho. CaOx crystals induced an increase in the DNA methylation levels of Klotho in the kidneys, and the inhibition of DNA methylation alleviated CaOx-induced renal aging. This study revealed that Klotho plays a crucial role in calcium oxalate crystal-induced kidney senescence by influencing kidney oxidative stress and mitochondrial function through the Nrf2/HO-1 pathway.
Collapse
Affiliation(s)
- Yuexian Xu
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Institute of Urology, Anhui Medical University, Hefei, China
- Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, Anhui Medical University, Hefei, China
| | - Jianmin You
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Institute of Urology, Anhui Medical University, Hefei, China
- Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, Anhui Medical University, Hefei, China
| | - Junfeng Yao
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Institute of Urology, Anhui Medical University, Hefei, China
- Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, Anhui Medical University, Hefei, China
| | - Bingbing Hou
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, China.
- Institute of Urology, Anhui Medical University, Hefei, China.
- Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, Anhui Medical University, Hefei, China.
| | - Wei Wang
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, China.
- Institute of Urology, Anhui Medical University, Hefei, China.
- Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, Anhui Medical University, Hefei, China.
- Department of Urology, Fuyang Hospital of Anhui Medical University, Fuyang, 236000, China.
| | - Zongyao Hao
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, China.
- Institute of Urology, Anhui Medical University, Hefei, China.
- Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, Anhui Medical University, Hefei, China.
| |
Collapse
|
5
|
Wang Z, Ni Y, Lou YR, Prud’homme GJ, Wang Q. Klotho protects INS-1 pancreatic β-cells from senescence and enhances mitochondrial function. FRONTIERS IN AGING 2025; 6:1512322. [PMID: 40018267 PMCID: PMC11865844 DOI: 10.3389/fragi.2025.1512322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 01/24/2025] [Indexed: 03/01/2025]
Abstract
Aging is an important contributing factor for β-cell failure which could lead to the development of type 2 diabetes (T2D). Aging β-cell exhibits signs of senescence and develops senescence-associated secretory phenotype (SASP), causing the senescence and dysfunction of neighboring cells through paracrine action. Klotho is recognized as an anti-aging gene, and the corresponding protein is α-Klotho (KL). KL exerts potent anti-aging effects on multiple cell types, but its role in β-cell aging remains unclear. Here we showed that pancreatic INS-1 cell (a rat insulinoma cell line commonly used to study pancreatic β-cell function) developed the typical hallmarks of senescent cells when treated with doxorubicin in vitro, and this was accompanied by downregulation of endogenous KL expression. Supplementation with exogenous KL protein protected pancreatic INS-1 cell against senescence, as indicated by downregulation of senescent markers and SA-β-gal staining. Notably, these effects were associated with improved mitochondrial ATP production and mitochondrial dynamic balance, as well as reduced ROS production. Our study further revealed that INS-1 cell treated with doxorubicin exhibited a reduced insulin secretion response to glucose stimulation, while supplementation with KL could reverse this effect. Our results indicate the important role of KL in regulating β-cell senescence and provide new mechanistic insights into its role in β-cell aging.
Collapse
Affiliation(s)
- Zhihong Wang
- Department of Endocrinology and Metabolism, Huashan Hospital, Fudan University, Shanghai, China
| | - Yunzhi Ni
- Department of Endocrinology and Metabolism, Huashan Hospital, Fudan University, Shanghai, China
| | - Yan-Ru Lou
- Department of Clinical Pharmacy and Drug Administration, School of Pharmacy, Fudan University, Shanghai, China
| | - Gerald J. Prud’homme
- Department of Laboratory Medicine and Pathobiology, Keenan Research Centre for Biomedical Science of St. Michael’s Hospital, Toronto, ON, Canada
| | - Qinghua Wang
- Department of Endocrinology and Metabolism, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
6
|
Parr MK, Keiler AM. Oligonucleotide therapeutics in sports? An antidoping perspective. Arch Pharm (Weinheim) 2025; 358:e2400404. [PMID: 39449227 PMCID: PMC11704058 DOI: 10.1002/ardp.202400404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 09/26/2024] [Accepted: 09/30/2024] [Indexed: 10/26/2024]
Abstract
Within the last two decades, the European Medicines Agency and the US Food and Drug Administration have approved several gene therapies. One category is oligonucleotide therapeutics, which allow for the regulation of the expression of target genes. Besides already approved therapeutics, there are several preclinical and clinical trials ongoing. The World Anti-Doping Agency prohibits the use of "nucleic acids or nucleic acid analogs that may alter genome sequences and/or alter gene expression by any mechanism" as a nonspecified method at all times. Hence, the administration of nucleic acids or analogs by athletes would cause an Anti-Doping Rule Violation. Herein, we discuss types of oligonucleotide therapeutics, their potential to be misused in sports, and considerations to sample preparation and mass spectrometric approaches with regard to antidoping analysis.
Collapse
Affiliation(s)
- Maria K. Parr
- Institute of Pharmacy, Pharmaceutical and Medicinal ChemistryFreie Universität BerlinBerlinGermany
| | - Annekathrin M. Keiler
- Institute of Doping Analysis & Sports BiochemistryKreischaGermany
- Environmental Monitoring & Endocrinology, Faculty of BiologyTechnische Universität DresdenDresdenGermany
| |
Collapse
|
7
|
Alakhdar AA, Sivakumar S, Kopchak RM, Hunter AN, Ambrosio F, Washburn NR. Age-Related ECM Stiffness Mediates TRAIL Activation in Muscle Stem Cell Differentiation. Adv Biol (Weinh) 2024; 8:e2400334. [PMID: 39601528 PMCID: PMC11889993 DOI: 10.1002/adbi.202400334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 10/01/2024] [Indexed: 11/29/2024]
Abstract
The stiffening of the extracellular matrix (ECM) with age hinders muscle regeneration by causing intrinsic muscle stem cell (MuSC) dysfunction through a poorly understood mechanism. Here, the study aims to study those age-related molecular changes in the differentiation of MuSCs due to age and/or stiffness. Hence, young and aged MuSCs are seeded onto substrates engineered to mimic a soft and stiff ECM microenvironment to study those molecular changes using single-cell RNA sequencing (scRNA). The trajectory of scRNA data of the MuSCs under four different conditions undergoing differentiation is analyzed as well as the active molecular pathways and transcription factors driving those differentiation fates. Data revealed the presence of a branching point within the trajectory leading to the emergence of an age-related fibroblastic population characterized by activation of the TNF-related apoptosis-inducing ligand (TRAIL) pathway, which is significantly activated in aged cells cultured on stiff substrates. Next, using the collagen cross-linking inhibitor β-aminopropionitrile (BAPN) in vivo, the study elucidates stiffness changes on TRAIL downstream apoptotic targets (caspase 8 and caspase 3) using immunostaining. TRAIL activity is significantly inhibited by BAPN in aged animals, indicating a complex mechanism of age-related declines in muscle function through inflammatory and apoptotic mediators.
Collapse
Affiliation(s)
- Amira A. Alakhdar
- Department of Chemistry, Carnegie Mellon University, Pittsburgh, PA, 15213, USA
| | | | - Rylee M. Kopchak
- Discovery Center for Musculoskeletal Recovery, Schoen Adams Research Institute at Spaulding, Boston, MA, USA
| | - Allison N. Hunter
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA, 15213, USA
| | - Fabrisia Ambrosio
- Discovery Center for Musculoskeletal Recovery, Schoen Adams Research Institute at Spaulding, Boston, MA, USA
- Department of Physical Medicine & Rehabilitation, Spaulding Rehabilitation Hospital, Harvard Medical School, Boston, MA, USA
| | - Newell R. Washburn
- Department of Chemistry, Carnegie Mellon University, Pittsburgh, PA, 15213, USA
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA, 15213, USA
| |
Collapse
|
8
|
Neto IVDS, Pinto AP, de Andrade RV, de Souza FHV, de Souza PEN, Assis V, Tibana RA, Neves RVP, Rosa TS, Prestes J, da Silva ASR, Marqueti RDC. Paternal exercise induces antioxidant defenses by α-Klotho/Keap1 pathways in the skeletal muscle of offspring exposed to a high fat-diet without changing telomere length. J Nutr Biochem 2024; 134:109747. [PMID: 39197728 DOI: 10.1016/j.jnutbio.2024.109747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 08/01/2024] [Accepted: 08/19/2024] [Indexed: 09/01/2024]
Abstract
Although previous studies demonstrated that the ancestral lifestyle can enhance the metabolic health of offspring exposed to an obesogenic diet, the specific connections between these positive effects in redox state and telomere length are unknown. We investigated the impact of paternal resistance training (RT) on stress-responsive signaling and the pathways involved in telomere homeostasis in skeletal muscle. This investigation encompassed both the fathers and first-generation litter exposed to a long-term standard diet (24 weeks) and high fat diet (HFD). Wistar rats were randomized into sedentary or trained fathers (8 weeks of resistance training). The offspring were obtained by mating with sedentary females. Upon weaning, male offspring were divided into four groups: offspring of sedentary or trained fathers exposed to either a control diet or HFD. The gastrocnemius was prepared for reverse transcription-quantitative polymerase chain reaction, immunoblotting, ELISA, and electron paramagnetic resonance spectroscopy. RT upregulated shelterin mRNA levels and antioxidant protein, preserving muscle telomere in fathers. Conversely, HFD induced a disturbance in the redox balance, which may have contributed to the offspring telomere shortening from sedentary fathers. Preconceptional paternal RT downregulates Kelch-like ECH-associated protein 1 (Keap1) mRNA levels in the skeletal muscle of progeny exposed to HFD, driving an increase in Glutathione reductase mRNA levels, Sod1 and Catalase protein levels to mitigate ROS production. Also, paternal exercise upregulates α-Klotho protein levels, mediating antioxidative responses without altering shelterin mRNA levels and telomere length. We provide the first in-depth analysis that the offspring's redox state seems to be directly associated with the beneficial effects of paternal exercise.
Collapse
Affiliation(s)
- Ivo Vieira de Sousa Neto
- School of Physical Education and Sport of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, São Paulo, Brazil.
| | - Ana Paula Pinto
- School of Physical Education and Sport of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, São Paulo, Brazil
| | - Rosangela Vieira de Andrade
- Graduate Program in Genomic Science and Biotechnology, Catholic University of Brasília, Taguatinga, Distrito Federal, Brazil
| | | | - Paulo Eduardo Narcizo de Souza
- Laboratory of Electron Paramagnetic Resonance, Institute of Physics, Universidade de Brasília, Brasília, Distrito Federal, Brazil
| | - Victória Assis
- Molecular of Analysis Laboratory, Faculty of Ceilândia, Universidade de Brasília (UNB), Brasília, Distrito Federal, Brazil
| | - Ramires Alsamir Tibana
- Graduate Program in Health Sciences, Faculdade de Medicine, Universidade Federal do Mato Grosso (UFMT), Cuiabá, Mato Grosso, Brazil
| | | | - Thiago Santos Rosa
- Graduate Program in Genomic Science and Biotechnology, Catholic University of Brasília, Taguatinga, Distrito Federal, Brazil; Graduate Program in Physical Education, Universidade Católica de Brasilia, Brasília, Distrito Federal, Brazil
| | - Jonato Prestes
- Graduate Program in Physical Education, Universidade Católica de Brasilia, Brasília, Distrito Federal, Brazil
| | - Adelino Sanchez Ramos da Silva
- School of Physical Education and Sport of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, São Paulo, Brazil; Postgraduate Program in Rehabilitation and Functional Performance, Ribeirão Preto Medical School, University of São Paulo (USP), Ribeirão Preto, São Paulo, Brazil
| | - Rita de Cassia Marqueti
- Molecular of Analysis Laboratory, Faculty of Ceilândia, Universidade de Brasília (UNB), Brasília, Distrito Federal, Brazil
| |
Collapse
|
9
|
Tang X, Song Y, Liang H, He W, Chen Z, Yang L, Yang C, Du R. The relationship between serum alpha-klotho levels and urinary incontinence in middle-aged and older females: insights from NHANES. Sci Rep 2024; 14:28667. [PMID: 39562625 PMCID: PMC11577013 DOI: 10.1038/s41598-024-80231-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 11/18/2024] [Indexed: 11/21/2024] Open
Abstract
Urinary incontinence (UI) in females is a prevailing condition that affects individuals across various age groups and is not limited to older females. The presence of serum alpha-Klotho (α-klotho) serves as a reliable biomarker to indicate the effects of antiaging. Nevertheless, the scientific research on the association between α-klotho and UI remains limited. Therefore, the purpose of this study is to investigate and evaluate the connection between α-klotho levels and the UI among females in the US. We utilized data from the National Health and Nutrition Examination Survey 2007-2016 to investigate the potential connection between α-klotho levels and the UI among females aged 40 to 79. Weighted linear regression models and sensitivity tests were conducted to explore the correlation. 2628 females were involved in this study, representing 22,492,348 non-institutionalized residents in the US. The mean age was 53.9 ± 0.2 years and the mean level of α-klotho was 873.0 ± 8.9 pg/mL. After adjusting for relevant covariables, weighted linear regression models revealed that individuals with severe UI exhibited significantly lower serum α-klotho levels (β = - 100.66; 95% CI: - 156.31, - 45.01; P < 0.001) than those without UI. Furthermore, in stratified analyses, the correlation was not significant in individuals with normal weight, cardiovascular disease, or chronic kidney disease. We did not find a significant association between the type of UI and α-klotho levels. In the NHANES data from 2007 to 2016, a noteworthy inverse relationship was noted between α-klotho levels and the severity of UI among females aged 40 to 79.
Collapse
Affiliation(s)
- Xiaoyan Tang
- Department of Cardiology, General Hospital of Central Theater Command, No.627, Wuluo Road, Wuhan, 430070, Hubei, China
| | - Yanhua Song
- Department of Obstetrics and Gynaecology, WuHan TongJi Aerospace City Hospital, No.80, Shuangliu Street, Wuhan, 430416, Hubei, China
| | - Hao Liang
- Department of Ultrasound, General Hospital of Central Theater Command, No.627, Wuluo Road, Wuhan, 430070, Hubei, China
| | - Wenjin He
- Department of Radiology, General Hospital of Central Theater Command, No.627, Wuluo Road, Wuhan, 430070, Hubei, China
| | - Zili Chen
- Department of Ultrasound, General Hospital of Central Theater Command, No.627, Wuluo Road, Wuhan, 430070, Hubei, China
| | - Li Yang
- Department of Ultrasound, General Hospital of Central Theater Command, No.627, Wuluo Road, Wuhan, 430070, Hubei, China
| | - Cheng Yang
- Department of Radiology, General Hospital of Central Theater Command, No.627, Wuluo Road, Wuhan, 430070, Hubei, China.
| | - Rui Du
- Department of Ultrasound, General Hospital of Central Theater Command, No.627, Wuluo Road, Wuhan, 430070, Hubei, China.
| |
Collapse
|
10
|
Prud’homme GJ, Wang Q. Anti-Inflammatory Role of the Klotho Protein and Relevance to Aging. Cells 2024; 13:1413. [PMID: 39272986 PMCID: PMC11394293 DOI: 10.3390/cells13171413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 08/17/2024] [Accepted: 08/23/2024] [Indexed: 09/15/2024] Open
Abstract
The α-Klotho protein (hereafter Klotho) is an obligate coreceptor for fibroblast growth factor 23 (FGF23). It is produced in the kidneys, brain and other sites. Klotho insufficiency causes hyperphosphatemia and other anomalies. Importantly, it is associated with chronic pathologies (often age-related) that have an inflammatory component. This includes atherosclerosis, diabetes and Alzheimer's disease. Its mode of action in these diseases is not well understood, but it inhibits or regulates multiple major pathways. Klotho has a membrane form and a soluble form (s-Klotho). Cytosolic Klotho is postulated but not well characterized. s-Klotho has endocrine properties that are incompletely elucidated. It binds to the FGF receptor 1c (FGFR1c) that is widely expressed (including endothelial cells). It also attaches to soluble FGF23, and FGF23/Klotho binds to FGFRs. Thus, s-Klotho might be a roaming FGF23 coreceptor, but it has other functions. Notably, Klotho (cell-bound or soluble) counteracts inflammation and appears to mitigate related aging (inflammaging). It inhibits NF-κB and the NLRP3 inflammasome. This inflammasome requires priming by NF-κB and produces active IL-1β, membrane pores and cell death (pyroptosis). In accord, Klotho countered inflammation and cell injury induced by toxins, damage-associated molecular patterns (DAMPs), cytokines, and reactive oxygen species (ROS). s-Klotho also blocks the TGF-β receptor and Wnt ligands, which lessens fibrotic disease. Low Klotho is associated with loss of muscle mass (sarcopenia), as occurs in aging and chronic diseases. s-Klotho counters the inhibitory effects of myostatin and TGF-β on muscle, reduces inflammation, and improves muscle repair following injury. The inhibition of TGF-β and other factors may also be protective in diabetic retinopathy and age-related macular degeneration (AMD). This review examines Klotho functions especially as related to inflammation and potential applications.
Collapse
Affiliation(s)
- Gérald J. Prud’homme
- Department of Laboratory Medicine and Pathobiology, University of Toronto, 220 Walmer Rd, Toronto, ON M5R 3R7, Canada
- Department of Laboratory Medicine, Keenan Research Centre for Biomedical Science, Unity Health Toronto, Toronto, ON M5B 1W8, Canada
| | - Qinghua Wang
- Department of Endocrinology and Metabolism, Huashan Hospital, Shanghai Medical School, Fudan University, Shanghai 200030, China
- Shanghai Innogen Pharmaceutical Co., Ltd., Shanghai 201318, China
| |
Collapse
|
11
|
Mohanty SK, Mohanty AK, Kumar MS, Suchiang K. Triiodothyronine enhances various forms of kidney-specific Klotho protein and suppresses the Wnt/β-catenin pathway: Insights from in-vitro, in-vivo and in-silico investigations. Cell Signal 2024; 120:111214. [PMID: 38729322 DOI: 10.1016/j.cellsig.2024.111214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 04/21/2024] [Accepted: 05/06/2024] [Indexed: 05/12/2024]
Abstract
Age-related diseases are intricately linked to the molecular processes underlying aging, with the decline of the antiaging protein Klotho being a key factor. Investigating these processes is crucial for developing therapeutic strategies. The age-associated reduction in Klotho expression, coupled with a decline in the endocrine hormone triiodothyronine (T3), prompted a detailed exploration of their potential interplay. Our research, conducted through both in-vitro and in-vivo studies on BALB/c mice, unveiled a significant capacity of T3 to upregulate various forms of Klotho via ATF-3/p-c-Jun transcription factor. This effect was particularly noteworthy in aged individuals, where Klotho expression had waned compared to their younger counterparts. Importantly, T3 demonstrated a promising therapeutic impact in rejuvenating Klotho expression in this context. Further investigations elucidated the molecular mechanisms underlying T3's impact on aging-related pathways. In-vitro and in-vivo experiments established T3's ability to downregulate the Wnt/β-Catenin pathway by enhancing Klotho expression. In-silico analyses provided insights into Klotho's intricate role, showing its capacity to inhibit Wnt ligands such as Wnt3 and Wnt8a, consequently disrupting their interaction with the Wnt receptor. Additionally, T3 was found to downregulate kidney-specific GSK-3β expression through the augmentation of Klotho expression. The study also highlighted T3's role in maintaining calcium and phosphate homeostasis via Klotho. This comprehensive investigation not only sheds light on the intricate mechanisms governing aging processes but also presents promising avenues for therapeutic interventions targeting the Wnt/β-Catenin pathway implicated in various age-associated diseases.
Collapse
Affiliation(s)
- Saswat Kumar Mohanty
- Department of Biochemistry and Molecular Biology, Pondicherry University, Pondicherry 605 014, India.
| | | | | | - Kitlangki Suchiang
- Department of Biochemistry, North Eastern Hill University, Shillong, Meghalaya 793022, India.
| |
Collapse
|
12
|
Lemche E, Killick R, Mitchell J, Caton PW, Choudhary P, Howard JK. Molecular mechanisms linking type 2 diabetes mellitus and late-onset Alzheimer's disease: A systematic review and qualitative meta-analysis. Neurobiol Dis 2024; 196:106485. [PMID: 38643861 DOI: 10.1016/j.nbd.2024.106485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 03/18/2024] [Accepted: 03/23/2024] [Indexed: 04/23/2024] Open
Abstract
Research evidence indicating common metabolic mechanisms through which type 2 diabetes mellitus (T2DM) increases risk of late-onset Alzheimer's dementia (LOAD) has accumulated over recent decades. The aim of this systematic review is to provide a comprehensive review of common mechanisms, which have hitherto been discussed in separate perspectives, and to assemble and evaluate candidate loci and epigenetic modifications contributing to polygenic risk linkages between T2DM and LOAD. For the systematic review on pathophysiological mechanisms, both human and animal studies up to December 2023 are included. For the qualitative meta-analysis of genomic bases, human association studies were examined; for epigenetic mechanisms, data from human studies and animal models were accepted. Papers describing pathophysiological studies were identified in databases, and further literature gathered from cited work. For genomic and epigenomic studies, literature mining was conducted by formalised search codes using Boolean operators in search engines, and augmented by GeneRif citations in Entrez Gene, and other sources (WikiGenes, etc.). For the systematic review of pathophysiological mechanisms, 923 publications were evaluated, and 138 gene loci extracted for testing candidate risk linkages. 3 57 publications were evaluated for genomic association and descriptions of epigenomic modifications. Overall accumulated results highlight insulin signalling, inflammation and inflammasome pathways, proteolysis, gluconeogenesis and glycolysis, glycosylation, lipoprotein metabolism and oxidation, cell cycle regulation or survival, autophagic-lysosomal pathways, and energy. Documented findings suggest interplay between brain insulin resistance, neuroinflammation, insult compensatory mechanisms, and peripheral metabolic dysregulation in T2DM and LOAD linkage. The results allow for more streamlined longitudinal studies of T2DM-LOAD risk linkages.
Collapse
Affiliation(s)
- Erwin Lemche
- Section of Cognitive Neuropsychiatry and Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology & Neuroscience, King's College London, De Crespigny Park, London SE5 8AF, United Kingdom.
| | - Richard Killick
- Section of Old Age Psychiatry, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology & Neuroscience, King's College London, De Crespigny Park, London SE5 8AF, United Kingdom
| | - Jackie Mitchell
- Department of Basic and Clinical Neurosciences, Maurice Wohl CIinical Neurosciences Institute, Institute of Psychiatry, Psychology & Neuroscience, King's College London, 125 Coldharbour Lane, London SE5 9NU, United Kingdom
| | - Paul W Caton
- Diabetes Research Group, School of Life Course Sciences, King's College London, Hodgkin Building, Guy's Campus, London SE1 1UL, United Kingdom
| | - Pratik Choudhary
- Diabetes Research Group, Weston Education Centre, King's College London, 10 Cutcombe Road, London SE5 9RJ, United Kingdom
| | - Jane K Howard
- School of Cardiovascular and Metabolic Medicine & Sciences, Hodgkin Building, Guy's Campus, King's College London, Great Maze Pond, London SE1 1UL, United Kingdom
| |
Collapse
|
13
|
Jasper AA, Shah KH, Karim H, Gujral S, Miljkovic I, Rosano C, Barchowsky A, Sahu A. Regenerative rehabilitation measures to restore tissue function after arsenic exposure. CURRENT OPINION IN BIOMEDICAL ENGINEERING 2024; 30:100529. [PMID: 40191583 PMCID: PMC11970924 DOI: 10.1016/j.cobme.2024.100529] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Environmental exposure of arsenic impairs the cardiometabolic profile, skeletal muscle health, and neurological function. Such declining tissue health is observed as early as in one's childhood, where the exposure is prevalent, thereby accelerating the effect of time's arrow. Despite the known deleterious effects of arsenic exposure, there is a paucity of specific treatment plans for restoring tissue function in exposed individuals. In this review, we propose to harness the untapped potential of existing regenerative rehabilitation programs, such as stem cell therapeutics with rehabilitation, acellular therapeutics, and artificial intelligence/robotics technologies, to address this critical gap in environmental toxicology. With regenerative rehabilitation techniques showing promise in other injury paradigms, fostering collaboration between these scientific realms offers an effective means of mitigating the detrimental effects of arsenic on tissue function.
Collapse
Affiliation(s)
- Adam A Jasper
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, USA
- Department of Environmental and Occupational Health, University of Pittsburgh, USA
| | - Kush H Shah
- The Lake Erie College of Osteopathic Medicine (LECOM), Erie, PA, USA
| | - Helmet Karim
- Department of Psychiatry, University of Pittsburgh, USA
- Department of Bioengineering, University of Pittsburgh, USA
| | - Swathi Gujral
- Department of Psychiatry, University of Pittsburgh, USA
| | - Iva Miljkovic
- Department of Epidemiology, University of Pittsburgh, USA
| | | | - Aaron Barchowsky
- Department of Environmental and Occupational Health, University of Pittsburgh, USA
| | - Amrita Sahu
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, USA
- Department of Environmental and Occupational Health, University of Pittsburgh, USA
| |
Collapse
|
14
|
Xie Y, Huang K, Li H, Kong W, Ye J. High serum klotho levels are inversely associated with the risk of low muscle mass in middle-aged adults: results from a cross-sectional study. Front Nutr 2024; 11:1390517. [PMID: 38854159 PMCID: PMC11157077 DOI: 10.3389/fnut.2024.1390517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 05/15/2024] [Indexed: 06/11/2024] Open
Abstract
Objective Muscle mass gradually declines with advancing age, and as an anti-aging protein, klotho may be associated with muscle mass. This study aims to explore the relationship between klotho levels and muscle mass in the middle-aged population. Methods Utilizing data from the National Health and Nutrition Examination Survey (NHANES) spanning 2011 to 2018, we conducted a cross-sectional analysis on a cohort of individuals aged 40-59. Weighted multivariable analysis was employed to assess the correlation between klotho and low muscle mass, with stratified and Restricted Cubic Spline (RCS) analyses. Results The cross-sectional investigation revealed a significant negative correlation between klotho levels and the risk of low muscle mass (Model 3: OR = 0.807, 95% CI: 0.712-0.915). A notable interaction between klotho and sex was observed, with a significant interaction effect (P for interaction = 0.01). The risk association was notably higher in females. The risk association was notably higher in females. Additionally, RCS analysis unveiled a significant linear relationship between klotho and low muscle mass (P for nonlinear = 0.9495, P for overall<0.0001). Conclusion Our observational analysis revealed a noteworthy inverse relationship between klotho and low muscle mass, particularly prominent among female participants. This discovery provides crucial insights for the development of more effective intervention strategies and offers a new direction for enhancing muscle quality in the middle-aged population.
Collapse
Affiliation(s)
- Yilian Xie
- Department of Infectious Diseases, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, China
- Department of Hepatology, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Kai Huang
- Department of General Medicine, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Hui Li
- Department of Infectious Diseases, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, China
- Department of Hepatology, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, China
- Health Science Center, Ningbo University, Ningbo, Zhejiang, China
| | - Weiliang Kong
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Jiayuan Ye
- Department of Infectious Diseases, Shangyu People's Hospital of Shaoxing, Shaoxing, Zhejiang, China
| |
Collapse
|
15
|
Heitman K, Alexander MS, Faul C. Skeletal Muscle Injury in Chronic Kidney Disease-From Histologic Changes to Molecular Mechanisms and to Novel Therapies. Int J Mol Sci 2024; 25:5117. [PMID: 38791164 PMCID: PMC11121428 DOI: 10.3390/ijms25105117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 05/03/2024] [Accepted: 05/06/2024] [Indexed: 05/26/2024] Open
Abstract
Chronic kidney disease (CKD) is associated with significant reductions in lean body mass and in the mass of various tissues, including skeletal muscle, which causes fatigue and contributes to high mortality rates. In CKD, the cellular protein turnover is imbalanced, with protein degradation outweighing protein synthesis, leading to a loss of protein and cell mass, which impairs tissue function. As CKD itself, skeletal muscle wasting, or sarcopenia, can have various origins and causes, and both CKD and sarcopenia share common risk factors, such as diabetes, obesity, and age. While these pathologies together with reduced physical performance and malnutrition contribute to muscle loss, they cannot explain all features of CKD-associated sarcopenia. Metabolic acidosis, systemic inflammation, insulin resistance and the accumulation of uremic toxins have been identified as additional factors that occur in CKD and that can contribute to sarcopenia. Here, we discuss the elevation of systemic phosphate levels, also called hyperphosphatemia, and the imbalance in the endocrine regulators of phosphate metabolism as another CKD-associated pathology that can directly and indirectly harm skeletal muscle tissue. To identify causes, affected cell types, and the mechanisms of sarcopenia and thereby novel targets for therapeutic interventions, it is important to first characterize the precise pathologic changes on molecular, cellular, and histologic levels, and to do so in CKD patients as well as in animal models of CKD, which we describe here in detail. We also discuss the currently known pathomechanisms and therapeutic approaches of CKD-associated sarcopenia, as well as the effects of hyperphosphatemia and the novel drug targets it could provide to protect skeletal muscle in CKD.
Collapse
Affiliation(s)
- Kylie Heitman
- Division of Nephrology and Section of Mineral Metabolism, Department of Medicine, Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, AL 35294, USA;
| | - Matthew S. Alexander
- Division of Neurology, Department of Pediatrics, The University of Alabama at Birmingham and Children’s of Alabama, Birmingham, AL 35294, USA
- Center for Exercise Medicine, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
- Department of Genetics, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
- Civitan International Research Center, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
- Center for Neurodegeneration and Experimental Therapeutics, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Christian Faul
- Division of Nephrology and Section of Mineral Metabolism, Department of Medicine, Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, AL 35294, USA;
| |
Collapse
|
16
|
Koike M, Sato T, Shiozaki Y, Komiya A, Miura M, Higashi A, Ishikawa A, Takayanagi K, Uga M, Miyamoto KI, Segawa H. Involvement of α-klotho in growth hormone (GH) signaling. J Clin Biochem Nutr 2024; 74:221-229. [PMID: 38799134 PMCID: PMC11111466 DOI: 10.3164/jcbn.23-127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Accepted: 01/10/2024] [Indexed: 05/29/2024] Open
Abstract
Growth hormone (GH) exerts multiple effects on different organs directly or via its main mediator, insulin-like growth factor1 (IGF1). In this study, we focused on the novel relationship between GH action and the antiaging hormone α-klotho. Immunofluorescent staining of α-klotho was observed in the renal distal tubules and pituitary glands of somatostatin- and GH-positive cells in wild-type (WT) mice. Treatment of 4-week-old WT mice with GH increased IGF1 mRNA expression in the pituitary gland, liver, heart, kidney, and bone but increased α-klotho mRNA expression only in the pituitary gland, kidney, and bone. Increased α-klotho protein levels were observed in the kidney but not in the pituitary gland. No induction of α-klotho RNA expression by GH was observed in juvenile mice with kidney disease, indicating GH resistance. Furthermore, GH and α-klotho supplementation in HEK293 cells transfected with GHR increased Janus kinase 2 mRNA (a GH downstream signal) expression compared to supplementation with GH alone. In conclusion, we suggest that 1) the kidney is the main source of secreted α-klotho, which is detected in blood by the downstream action of GH, 2) α-klotho induction by GH is resistant in kidney disease, and 3) α-klotho might be an enhanced regulator of GH signaling.
Collapse
Affiliation(s)
- Megumi Koike
- Department of Applied Nutrition, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan
| | - Tetsuhiko Sato
- General Medicine, Nagoya Daini Red Cross Hospital, 2-9 Myoken-cho, Showa-ku, Nagoya 466-8650, Japan
| | - Yuji Shiozaki
- Department of Applied Nutrition, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan
| | - Aoi Komiya
- Department of Applied Nutrition, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan
| | - Mizuki Miura
- Department of Applied Nutrition, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan
| | - Ayami Higashi
- Department of Applied Nutrition, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan
| | - Akane Ishikawa
- Department of Applied Nutrition, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan
| | - Kaori Takayanagi
- Department of Applied Nutrition, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan
| | - Minori Uga
- Department of Applied Nutrition, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan
| | - Ken-ichi Miyamoto
- Department of Applied Nutrition, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan
- Graduate School of Agriculture, Ryukoku University, 1-5 Yokotani, Seta Oe-cho, Otsu, Shiga 520-2194, Japan
| | - Hiroko Segawa
- Department of Applied Nutrition, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan
| |
Collapse
|
17
|
Mylonas KS, Karakitsos P, Tajik A, Pagliuso D, Emadzadeh H, Soukouli I, Hemmati P, Avgerinos DV, Stavridis GT, Boletis JN. Klotho as an Early Marker of Acute Kidney Injury Following Cardiac Surgery: A Systematic Review. J Cardiovasc Dev Dis 2024; 11:135. [PMID: 38786957 PMCID: PMC11122318 DOI: 10.3390/jcdd11050135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Revised: 04/13/2024] [Accepted: 04/23/2024] [Indexed: 05/25/2024] Open
Abstract
Acute kidney injury is a common complication following cardiac surgery (CSA-AKI). Serum creatinine levels require a minimum of 24-48 h to indicate renal injury. Nevertheless, early diagnosis remains critical for improving patient outcomes. A PRISMA-compliant systematic review of the PubMed and CENTRAL databases was performed to assess the role of Klotho as a predictive biomarker for CSA-AKI (end-of-search date: 17 February 2024). An evidence quality assessment of the four included studies was performed with the Newcastle-Ottawa scale. Among the 234 patients studied, 119 (50.8%) developed CSA-AKI postoperatively. Serum Klotho levels above 120 U/L immediately postoperatively correlated with an area under the curve (AUC) of 0.806 and 90% sensitivity. Additionally, a postoperative serum creatinine to Klotho ratio above 0.695 showed 94.7% sensitivity and 87.5% specificity, with an AUC of 92.4%, maintaining its prognostic validity for up to three days. Urinary Klotho immunoreactivity was better maintained in samples obtained via direct catheterization rather than indwelling catheter collection bags. Storage at -80 °C was necessary for delayed testing. Optimal timing for both serum and urine Klotho measurements was from the end of cardiopulmonary bypass to the time of the first ICU lab tests. In conclusion, Klotho could be a promising biomarker for the early diagnosis of CSA-AKI. Standardization of measurement protocols and larger studies are needed to validate these findings.
Collapse
Affiliation(s)
- Konstantinos S. Mylonas
- Department of Cardiac Surgery, Onassis Cardiac Surgery Center, 17674 Athens, Greece; (D.V.A.); (G.T.S.)
| | | | - Alireza Tajik
- School of Medicine, St. George’s University, True Blue, Grenada; (A.T.); (H.E.)
| | | | - Hamidreza Emadzadeh
- School of Medicine, St. George’s University, True Blue, Grenada; (A.T.); (H.E.)
| | - Ioanna Soukouli
- Department of Nephrology and Renal Transplantation, Laiko General Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece; (I.S.); (J.N.B.)
| | - Pouya Hemmati
- Department of Cardiothoracic Surgery, Baptist Health Medical Center, Little Rock, AR 72205, USA
| | - Dimitrios V. Avgerinos
- Department of Cardiac Surgery, Onassis Cardiac Surgery Center, 17674 Athens, Greece; (D.V.A.); (G.T.S.)
| | - George T. Stavridis
- Department of Cardiac Surgery, Onassis Cardiac Surgery Center, 17674 Athens, Greece; (D.V.A.); (G.T.S.)
| | - John N. Boletis
- Department of Nephrology and Renal Transplantation, Laiko General Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece; (I.S.); (J.N.B.)
| |
Collapse
|
18
|
Sun Y, Sheng R, Cao Z, Liu C, Li J, Zhang P, Du Y, Mo Q, Yao Q, Chen J, Zhang W. Bioactive fiber-reinforced hydrogel to tailor cell microenvironment for structural and functional regeneration of myotendinous junction. SCIENCE ADVANCES 2024; 10:eadm7164. [PMID: 38657071 PMCID: PMC11042749 DOI: 10.1126/sciadv.adm7164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Accepted: 03/18/2024] [Indexed: 04/26/2024]
Abstract
Myotendinous junction (MTJ) injuries are prevalent in clinical practice, yet the treatment approaches are limited to surgical suturing and conservative therapy, exhibiting a high recurrence rate. Current research on MTJ tissue engineering is scarce and lacks in vivo evaluation of repair efficacy. Here, we developed a three-dimensional-printed bioactive fiber-reinforced hydrogel containing mesenchymal stem cells (MSCs) and Klotho for structural and functional MTJ regeneration. In a rat MTJ defect model, the bioactive fiber-reinforced hydrogel promoted the structural restoration of muscle, tendon, and muscle-tendon interface and enhanced the functional recovery of injured MTJ. In vivo proteomics and in vitro cell cultures elucidated the regenerative mechanisms of the bioactive fiber-reinforced hydrogel by modulating oxidative stress and inflammation, thus engineering an optimized microenvironment to support the survival and differentiation of transplanted MSCs and maintain the functional phenotype of resident cells within MTJ tissues, including tendon/muscle cells and macrophages. This strategy provides a promising treatment for MTJ injuries.
Collapse
Affiliation(s)
- Yuzhi Sun
- Department of Orthopaedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, 210006 Nanjing, China
| | - Renwang Sheng
- School of Medicine, Southeast University, 210009 Nanjing, China
| | - Zhicheng Cao
- Department of Orthopaedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, 210006 Nanjing, China
| | - Chuanquan Liu
- School of Medicine, Southeast University, 210009 Nanjing, China
| | - Jiaxiang Li
- Department of Orthopaedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, 210006 Nanjing, China
| | - Po Zhang
- Department of Orthopaedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, 210006 Nanjing, China
| | - Yan Du
- School of Medicine, Southeast University, 210009 Nanjing, China
| | - Qingyun Mo
- School of Medicine, Southeast University, 210009 Nanjing, China
| | - Qingqiang Yao
- Department of Orthopaedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, 210006 Nanjing, China
- China Orthopedic Regenerative Medicine Group (CORMed), 310000 Hangzhou, China
| | - Jialin Chen
- School of Medicine, Southeast University, 210009 Nanjing, China
- China Orthopedic Regenerative Medicine Group (CORMed), 310000 Hangzhou, China
- Jiangsu Key Laboratory for Biomaterials and Devices, Southeast University, 210096 Nanjing, China
| | - Wei Zhang
- School of Medicine, Southeast University, 210009 Nanjing, China
- China Orthopedic Regenerative Medicine Group (CORMed), 310000 Hangzhou, China
- Jiangsu Key Laboratory for Biomaterials and Devices, Southeast University, 210096 Nanjing, China
| |
Collapse
|
19
|
Wu R, Xie Y, Peng Y, Wu X, Ma Y, Lyu FJ, Zheng Q, Deng Z. Young human plasma-derived extracellular vesicles rescue and reactivate IL-1β and TNF-α treated chondrocytes. Exp Cell Res 2024; 437:114009. [PMID: 38537745 DOI: 10.1016/j.yexcr.2024.114009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 03/06/2024] [Accepted: 03/16/2024] [Indexed: 04/19/2024]
Abstract
Osteoarthritis (OA) is a degenerative disease that affects millions of individuals worldwide. Despite its prevalence, the exact causes and mechanisms behind OA are still not fully understood, resulting in a lack of effective treatments to slow down or halt disease progression. Recent research has discovered that extracellular vesicles (EVs) present in the circulation of young mice have a remarkable ability to activate musculoskeletal stem cells in elderly mice. Conversely, EVs derived from elderly mice do not exhibit the same potential, indicating that EVs obtained from young individuals may hold promise to activate aging cells in degenerative tissue. However, it remains unknown whether EVs derived from young individuals can also address cartilage degeneration caused by aging. In this study, we first evaluated EVs derived from young human plasma (YEVs) and EVs derived from old human plasma (OEVs) in an in vitro experiment using chondrocytes. The results revealed that YEVs effectively stimulated chondrocyte proliferation and migration, while OEVs from old plasma did not exhibit a similar effect. Given that OA represents a more complex inflammatory microenvironment, we further determine whether the benefits of YEVs on chondrocytes can be maintained in this context. Our findings indicate that YEVs have the ability to positively regulate chondrocyte function and protect them against apoptosis induced by IL-1β and TNF-α in an in vitro OA model. Furthermore, we discovered that lyophilized EVs could be stored under mild conditions without any alterations in their physical characteristics. Considering the exceptional therapeutic effects and the wide availability of EVs from young plasma, they hold significant promise as a potential approach to activate chondrocytes and promote cartilage regeneration in early-stage OA.
Collapse
Affiliation(s)
- Rongjie Wu
- Department of Orthopedics, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510000, PR China; Shantou University Medical College, Shantou, Guangdong Province, 515000, PR China
| | - Yu Xie
- Department of Orthopedics, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510000, PR China; Shantou University Medical College, Shantou, Guangdong Province, 515000, PR China
| | - Yujie Peng
- Department of Orthopedics, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510000, PR China; Shantou University Medical College, Shantou, Guangdong Province, 515000, PR China
| | - Xiaohu Wu
- Department of Orthopedics, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510000, PR China
| | - Yuanchen Ma
- Department of Orthopedics, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510000, PR China
| | - Feng-Juan Lyu
- The Sixth Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, 510000, PR China; Joint Center for Regenerative Medicine Research of South China University of Technology and the University of Western Australia, School of Medicine, South China University of Technology, Guangzhou, 515000, PR China.
| | - Qiujian Zheng
- Department of Orthopedics, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510000, PR China.
| | - Zhantao Deng
- Department of Orthopedics, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510000, PR China.
| |
Collapse
|
20
|
Giacona JM, Afridi A, Bezan Petric U, Johnson T, Pastor J, Ren J, Sandon L, Malloy C, Pandey A, Shah A, Berry JD, Moe OW, Vongpatanasin W. Association between dietary phosphate intake and skeletal muscle energetics in adults without cardiovascular disease. J Appl Physiol (1985) 2024; 136:1007-1014. [PMID: 38482570 PMCID: PMC11575913 DOI: 10.1152/japplphysiol.00818.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 02/05/2024] [Accepted: 02/24/2024] [Indexed: 04/17/2024] Open
Abstract
Highly bioavailable inorganic phosphate (Pi) is present in large quantities in the typical Western diet and represents a large fraction of total phosphate intake. Dietary Pi excess induces exercise intolerance and skeletal muscle mitochondrial dysfunction in normal mice. However, the relevance of this to humans remains unknown. The study was conducted on 13 individuals without a history of cardiopulmonary disease (46% female, 15% Black participants) enrolled in the pilot-phase of the Dallas Heart and Mind Study. Total dietary phosphate was estimated from 24-h dietary recall (ASA24). Muscle ATP synthesis was measured at rest, and phosphocreatinine (PCr) dynamics was measured during plantar flexion exercise using 7-T 31P magnetic resonance (MR) spectroscopy in the calf muscle. Correlation was assessed between dietary phosphate intake normalized to total caloric intake, resting ATP synthesis, and PCr depletion during exercise. Higher dietary phosphate intake was associated with lower resting ATP synthesis (r = -0.62, P = 0.03), and with higher levels of PCr depletion during plantar flexion exercise relative to the resting period (r = -0.72; P = 0.004). These associations remain significant after adjustment for age and estimated glomerular filtration rate (both P < 0.05). High dietary phosphate intake was also associated with lower serum Klotho levels, and Klotho levels are in turn associated with PCr depletion and higher ADP accumulation post exercise. Our study suggests that higher dietary phosphate is associated with reduced skeletal muscle mitochondrial function at rest and exercise in humans providing new insight into potential mechanisms linking the Western diet to impaired energy metabolism.NEW & NOTEWORTHY This is the first translational research study directly demonstrating the adverse effects of dietary phosphate on muscle energy metabolism in humans. Importantly, our data show that dietary phosphate is associated with impaired muscle ATP synthesis at rest and during exercise, independent of age and renal function. This is a new biologic paradigm with significant clinical dietary implications.
Collapse
Affiliation(s)
- John M Giacona
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, United States
- Department of Applied Clinical Research, University of Texas Southwestern Medical Center, Dallas, Texas, United States
| | - Areeb Afridi
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, United States
| | - Ursa Bezan Petric
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, United States
| | - Talon Johnson
- Advanced Imaging Research Center, University of Texas Southwestern Medical Center, Dallas, Texas, United States
| | - Johanne Pastor
- Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, Texas, United States
| | - Jimin Ren
- Advanced Imaging Research Center, University of Texas Southwestern Medical Center, Dallas, Texas, United States
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, Texas, United States
| | - Lona Sandon
- Department of Clinical Nutrition, University of Texas Southwestern Medical Center, Dallas, Texas, United States
| | - Craig Malloy
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, United States
- Advanced Imaging Research Center, University of Texas Southwestern Medical Center, Dallas, Texas, United States
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, Texas, United States
| | - Ambarish Pandey
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, United States
| | - Amil Shah
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, United States
| | - Jarett D Berry
- Department of Internal Medicine, University of Texas Health Science Center at Tyler, Tyler, Texas, United States
| | - Orson W Moe
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, United States
- Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, Texas, United States
- Department of Physiology, University of Texas Southwestern Medical Center, Dallas, Texas, United States
| | - Wanpen Vongpatanasin
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, United States
- Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, Texas, United States
| |
Collapse
|
21
|
Park MJ, Lee J, Bagon BB, Matienzo ME, Lee CM, Kim K, Kim DI. Therapeutic potential of AAV-FL-Klotho in obesity: Impact on weight loss and lipid metabolism in mice. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167055. [PMID: 38325589 DOI: 10.1016/j.bbadis.2024.167055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 01/04/2024] [Accepted: 02/02/2024] [Indexed: 02/09/2024]
Abstract
Klotho, an anti-aging protein, has gained attention for its protective effects against various diseases, including metabolic disorders, through recombinant Klotho administration. However, the potential of Klotho as a target for gene therapy requires further exploration, as it remains relatively understudied in the context of metabolic disorders. In this study, we demonstrate that AAV-full length(FL)-Klotho administration induces weight loss in mice and provides protection against high-fat diet (HFD)-induced obesity and hepatic steatosis, concurrently reducing the weights of white adipose tissue and liver. AAV-FL-Klotho administration also enhanced thermogenic gene expression in brown adipose tissue (BAT) and improved the morphology of interscapular BAT. The weight loss effect of AAV-FL-Klotho was found to be, at least in part, mediated by UCP1-dependent thermogenesis in brown adipocytes, potentially influenced by hepatokines secreted from AAV-FL-Klotho-transduced hepatocytes. These findings suggest that AAV-FL-Klotho is an attractive candidate for gene therapy to combat obesity. Nevertheless, unbiased experiments have also revealed disturbances in lipid metabolism due to AAV-FL-Klotho, as evidenced by the emergence of lipomas and increased expression of hepatic lipogenic proteins.
Collapse
Affiliation(s)
- Min-Jung Park
- Department of Veterinary Physiology, College of Veterinary Medicine, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Junhyeong Lee
- Department of Veterinary Physiology, College of Veterinary Medicine, Chonnam National University, Gwangju 61186, Republic of Korea; College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Bernadette B Bagon
- Department of Veterinary Physiology, College of Veterinary Medicine, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Merc Emil Matienzo
- Department of Veterinary Physiology, College of Veterinary Medicine, Chonnam National University, Gwangju 61186, Republic of Korea; College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Chang-Min Lee
- College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju 61186, Republic of Korea; Department of Veterinary Internal Medicine, College of Veterinary Medicine, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Keon Kim
- College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju 61186, Republic of Korea; Department of Veterinary Internal Medicine, College of Veterinary Medicine, Chonnam National University, Gwangju 61186, Republic of Korea.
| | - Dong-Il Kim
- Department of Veterinary Physiology, College of Veterinary Medicine, Chonnam National University, Gwangju 61186, Republic of Korea; College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju 61186, Republic of Korea.
| |
Collapse
|
22
|
Cisterna B, Malatesta M. Molecular and Structural Alterations of Skeletal Muscle Tissue Nuclei during Aging. Int J Mol Sci 2024; 25:1833. [PMID: 38339110 PMCID: PMC10855217 DOI: 10.3390/ijms25031833] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 01/30/2024] [Accepted: 01/31/2024] [Indexed: 02/12/2024] Open
Abstract
Aging is accompanied by a progressive loss of skeletal muscle mass and strength. The mechanisms underlying this phenomenon are certainly multifactorial and still remain to be fully elucidated. Changes in the cell nucleus structure and function have been considered among the possible contributing causes. This review offers an overview of the current knowledge on skeletal muscle nuclei in aging, focusing on the impairment of nuclear pathways potentially involved in age-related muscle decline. In skeletal muscle two types of cells are present: fiber cells, constituting the contractile muscle mass and containing hundreds of myonuclei, and the satellite cells, i.e., the myogenic mononuclear stem cells occurring at the periphery of the fibers and responsible for muscle growth and repair. Research conducted on different experimental models and with different methodological approaches demonstrated that both the myonuclei and satellite cell nuclei of aged skeletal muscles undergo several structural and molecular alterations, affecting chromatin organization, gene expression, and transcriptional and post-transcriptional activities. These alterations play a key role in the impairment of muscle fiber homeostasis and regeneration, thus contributing to the age-related decrease in skeletal muscle mass and function.
Collapse
Affiliation(s)
| | - Manuela Malatesta
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Strada Le Grazie 8, 37134 Verona, Italy;
| |
Collapse
|
23
|
Bartolacci JG, Behun MN, Warunek JP, Li T, Sahu A, Dwyer GK, Lucas A, Rong J, Ambrosio F, Turnquist HR, Badylak SF. Matrix-bound nanovesicle-associated IL-33 supports functional recovery after skeletal muscle injury by initiating a pro-regenerative macrophage phenotypic transition. NPJ Regen Med 2024; 9:7. [PMID: 38280914 PMCID: PMC10821913 DOI: 10.1038/s41536-024-00346-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Accepted: 01/04/2024] [Indexed: 01/29/2024] Open
Abstract
Injuries to skeletal muscle are among the most common injuries in civilian and military populations, accounting for nearly 60% of extremity injuries. The standard of care for severe extremity injury has been focused upon limb salvage procedures and the utilization of tissue grafts or orthotics in conjunction with rehabilitation to avoid amputation. Nonetheless, many patients have persistent strength and functional deficits that permanently impact their quality of life. Preclinical and clinical studies have shown that partial restoration of functional skeletal muscle tissue following injury can be achieved by the implantation of a biologic scaffold composed of extracellular matrix (ECM). These favorable outcomes are mediated, at least in part, through local immunomodulation. The mechanisms underlying this immunomodulatory effect, however, are poorly understood. The present study investigates a potential mechanistic driver of the immunomodulatory effects; specifically, the effect of selected ECM components upon inflammation resolution and repair. Results show that the host response to skeletal muscle injury is profoundly altered and functional recovery decreased in il33-/- mice compared to age- and sex-matched wildtype counterparts by 14 days post-injury. Results also show that IL-33, contained within matrix-bound nanovesicles (MBV), supports skeletal muscle regeneration by regulating local macrophage activation toward a pro-remodeling phenotype via canonical and non-canonical pathways to improve functional recovery from injury compared to untreated il33-/- counterparts. Taken together, these data suggest that MBV and their associated IL-33 cargo represent a novel homeostatic signaling mechanism that contributes to skeletal muscle repair.
Collapse
Affiliation(s)
- J G Bartolacci
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA
| | - M N Behun
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA
| | - J P Warunek
- Departments of Surgery and Immunology, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - T Li
- Departments of Surgery and Immunology, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - A Sahu
- Department of Physical Medicine and Rehabilitation Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| | - G K Dwyer
- Departments of Surgery and Immunology, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - A Lucas
- Departments of Surgery and Immunology, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - J Rong
- McGowan Institute for Regenerative Medicine, Pittsburgh, USA
| | - F Ambrosio
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Physical Medicine and Rehabilitation Sciences, University of Pittsburgh, Pittsburgh, PA, USA
- McGowan Institute for Regenerative Medicine, Pittsburgh, USA
| | - H R Turnquist
- Departments of Surgery and Immunology, University of Pittsburgh Medical Center, Pittsburgh, PA, USA.
- McGowan Institute for Regenerative Medicine, Pittsburgh, USA.
| | - S F Badylak
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA.
- Departments of Surgery and Immunology, University of Pittsburgh Medical Center, Pittsburgh, PA, USA.
- McGowan Institute for Regenerative Medicine, Pittsburgh, USA.
| |
Collapse
|
24
|
Ochi E, Barrington A, Wehling‐Henricks M, Avila M, Kuro‐o M, Tidball JG. Klotho regulates the myogenic response of muscle to mechanical loading and exercise. Exp Physiol 2023; 108:1531-1547. [PMID: 37864311 PMCID: PMC10841225 DOI: 10.1113/ep091263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 08/16/2023] [Indexed: 10/22/2023]
Abstract
NEW FINDINGS What is the central question of this study? Does the hormone Klotho affect the myogenic response of muscle cells to mechanical loading or exercise? What is the main finding and its importance? Klotho prevents direct, mechanical activation of genes that regulate muscle differentiation, including genes that encode the myogenic regulatory factor myogenin and proteins in the canonical Wnt signalling pathway. Similarly, elevated levels of klotho expression in vivo prevent the exercise-induced increase in myogenin-expressing cells and reduce exercise-induced activation of the Wnt pathway. These findings demonstrate a new mechanism through which the responses of muscle to the mechanical environment are regulated. ABSTRACT Muscle growth is influenced by changes in the mechanical environment that affect the expression of genes that regulate myogenesis. We tested whether the hormone Klotho could influence the response of muscle to mechanical loading. Applying mechanical loads to myoblasts in vitro increased RNA encoding transcription factors that are expressed in activated myoblasts (Myod) and in myogenic cells that have initiated terminal differentiation (Myog). However, application of Klotho to myoblasts prevented the loading-induced activation of Myog without affecting loading-induced activation of Myod. This indicates that elevated Klotho inhibits mechanically-induced differentiation of myogenic cells. Elevated Klotho also reduced the transcription of genes encoding proteins involved in the canonical Wnt pathway or their target genes (Wnt9a, Wnt10a, Ccnd1). Because the canonical Wnt pathway promotes differentiation of myogenic cells, these findings indicate that Klotho inhibits the differentiation of myogenic cells experiencing mechanical loading. We then tested whether these effects of Klotho occurred in muscles of mice experiencing high-intensity interval training (HIIT) by comparing wild-type mice and klotho transgenic mice. The expression of a klotho transgene combined with HIIT synergized to tremendously elevate numbers of Pax7+ satellite cells and activated MyoD+ cells. However, transgene expression prevented the increase in myogenin+ cells caused by HIIT in wild-type mice. Furthermore, transgene expression diminished the HIIT-induced activation of the canonical Wnt pathway in Pax7+ satellite cells. Collectively, these findings show that Klotho inhibits loading- or exercise-induced activation of muscle differentiation and indicate a new mechanism through which the responses of muscle to the mechanical environment are regulated.
Collapse
Affiliation(s)
- Eisuke Ochi
- Faculty of Bioscience and Applied ChemistryHosei UniversityTokyoJapan
- Department of Integrative Biology and PhysiologyUniversity of CaliforniaLos AngelesCAUSA
| | - Alice Barrington
- Department of Integrative Biology and PhysiologyUniversity of CaliforniaLos AngelesCAUSA
| | | | - Marcus Avila
- Department of Integrative Biology and PhysiologyUniversity of CaliforniaLos AngelesCAUSA
| | - Makoto Kuro‐o
- Division of Anti‐Aging MedicineCenter for Molecular MedicineJichi Medical UniversityTochigiJapan
| | - James G. Tidball
- Department of Integrative Biology and PhysiologyUniversity of CaliforniaLos AngelesCAUSA
- Molecular, Cellular & Integrative Physiology ProgramUniversity of CaliforniaLos AngelesCAUSA
- Department of BioengineeringUniversity of CaliforniaLos AngelesCAUSA
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLAUniversity of CaliforniaLos AngelesCAUSA
| |
Collapse
|
25
|
Arroyo E, Leber CA, Burney HN, Narayanan G, Moorthi R, Avin KG, Warden SJ, Moe SM, Lim K. Relationship between klotho and physical function in healthy aging. Sci Rep 2023; 13:21158. [PMID: 38036596 PMCID: PMC10689840 DOI: 10.1038/s41598-023-47791-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 11/18/2023] [Indexed: 12/02/2023] Open
Abstract
Epidemiological studies have reported a strong association between circulating Klotho and physical function; however, the cohorts were comprised of older adults with multiple comorbidities. Herein, we examined the relationship between Klotho and physical function in a community-based cohort of healthy adults. In this cross-sectional study, serum Klotho was measured in 80 adults who visited the Musculoskeletal Function, Imaging, and Tissue Resource Core of the Indiana Center for Musculoskeletal Health. Participants (n = 20, 10 [50%] men per group) were chosen into four age groups: 20-34, 35-49, 50-64, and ≥ 65 years, and were further grouped based on performance (low vs. high) on grip strength and chair stand tests. Klotho levels were lower in the ≥ 65 years group (703.0 [189.3] pg/mL; p = 0.022) and the 50-64 years group (722.6 [190.5] pg/mL; p = 0.045) compared to 20-34 years (916.1 [284.8] pg/mL). No differences were observed in Klotho between the low and high performers. The ≥ 65 years group walked a shorter distance during the 6-min walk test (6MWT) compared to 20-34 years (p = 0.005). Klotho was correlated with age (p < 0.001), body fat (p = 0.037), and 6MWT distance (p = 0.022). Klotho levels decline as early as the fifth decade of life, potentially before the onset of age-related impairment in exercise capacity.
Collapse
Affiliation(s)
- Eliott Arroyo
- Division of Nephrology & Hypertension, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Health and Exercise Science, Wake Forest University, Winston-Salem, NC, USA
| | - Cecilia A Leber
- Division of Nephrology & Hypertension, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN, USA
| | - Heather N Burney
- Department of Biostatistics and Health Data Science, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Gayatri Narayanan
- Division of Nephrology & Hypertension, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Ranjani Moorthi
- Division of Nephrology & Hypertension, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Keith G Avin
- Division of Nephrology & Hypertension, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Physical Therapy, Indiana University School of Health and Human Sciences, Indianapolis, IN, USA
| | - Stuart J Warden
- Department of Physical Therapy, Indiana University School of Health and Human Sciences, Indianapolis, IN, USA
| | - Sharon M Moe
- Division of Nephrology & Hypertension, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Kenneth Lim
- Division of Nephrology & Hypertension, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA.
| |
Collapse
|
26
|
Wu Y, Huang X, Tan Z, Zang J, Peng M, He N, Zhang T, Mai H, Xu A, Lu D. FUS-mediated HypEVs: Neuroprotective effects against ischemic stroke. Bioact Mater 2023; 29:196-213. [PMID: 37621770 PMCID: PMC10444975 DOI: 10.1016/j.bioactmat.2023.07.009] [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: 09/26/2022] [Revised: 06/02/2023] [Accepted: 07/05/2023] [Indexed: 08/26/2023] Open
Abstract
Few studies have investigated the properties and protein composition of small extracellular vesicles (sEVs) derived from neurons under hypoxic conditions. Presently, the extent of the involvement of these plentiful sEVs in the onset and progression of ischemic stroke remains an unresolved question. Our study systematically identified the characteristics of sEVs derived from neurons under hypoxic conditions (HypEVs) by physical characterization, sEV absorption, proteomics and transcriptomics analysis. The effects of HypEVs on neurites, cell survival, and neuron structure were assessed in vitro and in vivo by neural complexity tests, magnetic resonance imaging (MRI), Golgi staining, and Western blotting of synaptic plasticity-related proteins and apoptotic proteins. Knockdown of Fused in Sarcoma (FUS) small interfering RNA (siRNA) was used to validate FUS-mediated HypEV neuroprotection and mitochondrial mRNA release. Hypoxia promoted the secretion of sEVs, and HypEVs were more easily taken up and utilized by recipient cells. The MRI results illustrated that the cerebral infarction volume was reduced by 45% with the application of HypEVs, in comparison to the non- HypEV treatment group. Mechanistically, the FUS protein is necessary for the uptake and neuroprotection of HypEVs against ischemic stroke as well as carrying a large amount of mitochondrial mRNA in HypEVs. However, FUS knockdown attenuated the neuroprotective rescue capabilities of HypEVs. Our comprehensive dataset clearly illustrates that FUS-mediated HypEVs deliver exceptional neuroprotective effects against ischemic stroke, primarily through the maintenance of neurite integrity and the reduction of mitochondria-associated apoptosis.
Collapse
Affiliation(s)
- Yousheng Wu
- Department of Neurology and Stroke Center, The First Affiliated Hospital of Jinan University, Guangzhou, China
- Clinical Neuroscience Institute, The First Affiliated Hospital of Jinan University, Guangzhou, China
- Key Lab of Guangzhou Basic and Translational Research of Pan-vascular Diseases, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Xiaoxiong Huang
- Department of Neurology and Stroke Center, The First Affiliated Hospital of Jinan University, Guangzhou, China
- Clinical Neuroscience Institute, The First Affiliated Hospital of Jinan University, Guangzhou, China
- Department of Neurology and Stroke Center, The Central Hospital of Shaoyang, Hunan, China
| | - Zefeng Tan
- Department of Neurology, The First People's Hospital of Foshan, Guangdong, China
| | - Jiankun Zang
- Department of Neurology and Stroke Center, The First Affiliated Hospital of Jinan University, Guangzhou, China
- Clinical Neuroscience Institute, The First Affiliated Hospital of Jinan University, Guangzhou, China
- Key Lab of Guangzhou Basic and Translational Research of Pan-vascular Diseases, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Min Peng
- Department of Neurology and Stroke Center, The First Affiliated Hospital of Jinan University, Guangzhou, China
- Clinical Neuroscience Institute, The First Affiliated Hospital of Jinan University, Guangzhou, China
- Key Lab of Guangzhou Basic and Translational Research of Pan-vascular Diseases, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Niu He
- Department of Neurology and Stroke Center, The First Affiliated Hospital of Jinan University, Guangzhou, China
- Key Lab of Guangzhou Basic and Translational Research of Pan-vascular Diseases, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Tao Zhang
- Department of Cardiology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Hongcheng Mai
- Department of Neurology and Stroke Center, The First Affiliated Hospital of Jinan University, Guangzhou, China
- Clinical Neuroscience Institute, The First Affiliated Hospital of Jinan University, Guangzhou, China
- Munich Medical Research School (MMRS), Ludwig-Maximilians University Munich, Munich, Germany
- Insititute for Tissue Engineering and Regenerative Medicine (iTERM), Helmholtz Zentrum München, Neuherberg, Germany
| | - Anding Xu
- Department of Neurology and Stroke Center, The First Affiliated Hospital of Jinan University, Guangzhou, China
- Clinical Neuroscience Institute, The First Affiliated Hospital of Jinan University, Guangzhou, China
- Key Lab of Guangzhou Basic and Translational Research of Pan-vascular Diseases, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Dan Lu
- Department of Neurology and Stroke Center, The First Affiliated Hospital of Jinan University, Guangzhou, China
- Clinical Neuroscience Institute, The First Affiliated Hospital of Jinan University, Guangzhou, China
- Key Lab of Guangzhou Basic and Translational Research of Pan-vascular Diseases, The First Affiliated Hospital of Jinan University, Guangzhou, China
| |
Collapse
|
27
|
Jiang W, Gan C, Zhou X, Yang Q, Chen D, Xiao H, Dai L, Chen Y, Wang M, Yang H, Li Q. Klotho inhibits renal ox-LDL deposition via IGF-1R/RAC1/OLR1 signaling to ameliorate podocyte injury in diabetic kidney disease. Cardiovasc Diabetol 2023; 22:293. [PMID: 37891556 PMCID: PMC10612302 DOI: 10.1186/s12933-023-02025-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 10/11/2023] [Indexed: 10/29/2023] Open
Abstract
OBJECTIVE Diabetic kidney disease (DKD) is characterized by the abnormal deposition of oxidized low-density lipoprotein (ox-LDL), which contributes to podocyte damage. Klotho, an aging suppressor that plays a critical role in protecting podocytes in DKD, is mainly expressed in kidney tubular epithelium and secreted in the blood. However, it has not been established whether Klotho can alleviate podocyte injury by inhibiting renal ox-LDL deposition, and the potential molecular mechanisms require further investigation. METHODS We conducted a comprehensive analysis of serum and kidney biopsy samples obtained from patients diagnosed with DKD. Additionally, to explore the underlying mechanism of Klotho in the deposition of ox-LDL in the kidneys, we employed a mouse model of DKD with the Klotho genotype induced by streptozotocin (STZ). Furthermore, we conducted meticulous in vitro experiments on podocytes to gain further insights into the specific role of Klotho in the deposition of ox-LDL within the kidney. RESULTS Our groundbreaking study unveiled the remarkable ability of the soluble form of Klotho to effectively inhibit high glucose-induced ox-LDL deposition in podocytes affected by DKD. Subsequent investigations elucidated that Klotho achieved this inhibition by reducing the expression of the insulin/insulin-like growth factor 1 receptor (IGF-1R), consequently leading to a decrease in the expression of Ras-related C3 botulinum toxin substrate 1 (RAC1) and an enhancement of mitochondrial function. Ultimately, this series of events culminated in a significant reduction in the expression of the oxidized low-density lipoprotein receptor (OLR1), thereby resulting in a notable decrease in renal ox-LDL deposition in DKD. CONCLUSION Our findings suggested that Klotho had the potential to mitigate podocyte injury and reduced high glucose-induced ox-LDL deposition in glomerulus by modulating the IGF-1R/RAC1/OLR1 signaling. These results provided valuable insights that could inform the development of novel strategies for diagnosing and treating DKD.
Collapse
Affiliation(s)
- Wei Jiang
- Chongqing Key Laboratory of Pediatrics, Department of Nephrology, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Pediatric Research Institute, Children's Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Chun Gan
- Chongqing Key Laboratory of Pediatrics, Department of Nephrology, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Pediatric Research Institute, Children's Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Xindi Zhou
- Chongqing Key Laboratory of Pediatrics, Department of Nephrology, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Pediatric Research Institute, Children's Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Qing Yang
- Chongqing Key Laboratory of Pediatrics, Department of Nephrology, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Pediatric Research Institute, Children's Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Dan Chen
- Chongqing Key Laboratory of Pediatrics, Department of Nephrology, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Pediatric Research Institute, Children's Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Han Xiao
- Chongqing Key Laboratory of Pediatrics, Department of Nephrology, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Pediatric Research Institute, Children's Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Lujun Dai
- Department of Pathology, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, People's Republic of China
| | - Yaxi Chen
- Department of Infectious Diseases, Centre for Lipid Research & Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, People's Republic of China
| | - Mo Wang
- Chongqing Key Laboratory of Pediatrics, Department of Nephrology, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Pediatric Research Institute, Children's Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Haiping Yang
- Chongqing Key Laboratory of Pediatrics, Department of Nephrology, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Pediatric Research Institute, Children's Hospital of Chongqing Medical University, Chongqing, People's Republic of China.
| | - Qiu Li
- Chongqing Key Laboratory of Pediatrics, Department of Nephrology, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Pediatric Research Institute, Children's Hospital of Chongqing Medical University, Chongqing, People's Republic of China.
| |
Collapse
|
28
|
Cai Z, Wu X, Song Z, Sun S, Su Y, Wang T, Cheng X, Yu Y, Yu C, Chen E, Chen W, Yu Y, Linkermann A, Min J, Wang F. Metformin potentiates nephrotoxicity by promoting NETosis in response to renal ferroptosis. Cell Discov 2023; 9:104. [PMID: 37848438 PMCID: PMC10582023 DOI: 10.1038/s41421-023-00595-3] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Accepted: 08/16/2023] [Indexed: 10/19/2023] Open
Abstract
Given the rapidly aging population, aging-related diseases are becoming an excessive burden on the global healthcare system. Metformin has been shown to be beneficial to many age-related disorders, as well as increase lifespan in preclinical animal models. During the aging process, kidney function progressively declines. Currently, whether and how metformin protects the kidney remains unclear. In this study, among longevity drugs, including metformin, nicotinamide, resveratrol, rapamycin, and senolytics, we unexpectedly found that metformin, even at low doses, exacerbated experimentally-induced acute kidney injury (AKI) and increased mortality in mice. By single-cell transcriptomics analysis, we found that death of renal parenchymal cells together with an expansion of neutrophils occurs upon metformin treatment after AKI. We identified programmed cell death by ferroptosis in renal parenchymal cells and blocking ferroptosis, or depleting neutrophils protects against metformin-induced nephrotoxicity. Mechanistically, upon induction of AKI, ferroptosis in renal parenchymal cells initiates the migration of neutrophils to the site of injury via the surface receptor CXCR4-bound to metformin-iron-NGAL complex, which results in NETosis aggravated AKI. Finally, we demonstrated that reducing iron showed protective effects on kidney injury, which supports the notion that iron plays an important role in metformin-triggered AKI. Taken together, these findings delineate a novel mechanism underlying metformin-aggravated nephropathy and highlight the mechanistic relationship between iron, ferroptosis, and NETosis in the resulting AKI.
Collapse
Affiliation(s)
- Zhaoxian Cai
- The Second Affiliated Hospital, The First Affiliated Hospital, School of Public Health, Institute of Translational Medicine, Cancer Center, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- The First Affiliated Hospital, Basic Medical Sciences, School of Public Health, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Xiaotian Wu
- The Second Affiliated Hospital, The First Affiliated Hospital, School of Public Health, Institute of Translational Medicine, Cancer Center, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Zijun Song
- The Second Affiliated Hospital, The First Affiliated Hospital, School of Public Health, Institute of Translational Medicine, Cancer Center, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Shumin Sun
- The Second Affiliated Hospital, The First Affiliated Hospital, School of Public Health, Institute of Translational Medicine, Cancer Center, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yunxing Su
- The Second Affiliated Hospital, The First Affiliated Hospital, School of Public Health, Institute of Translational Medicine, Cancer Center, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Tianyi Wang
- The First Affiliated Hospital, Basic Medical Sciences, School of Public Health, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Xihao Cheng
- The Second Affiliated Hospital, The First Affiliated Hospital, School of Public Health, Institute of Translational Medicine, Cancer Center, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yingying Yu
- The Second Affiliated Hospital, The First Affiliated Hospital, School of Public Health, Institute of Translational Medicine, Cancer Center, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Chao Yu
- The Second Affiliated Hospital, The First Affiliated Hospital, School of Public Health, Institute of Translational Medicine, Cancer Center, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - En Chen
- College of Pharmaceutical Science, Zhejiang University, Hangzhou, Zhejiang, China
| | - Wenteng Chen
- College of Pharmaceutical Science, Zhejiang University, Hangzhou, Zhejiang, China
| | - Yongping Yu
- College of Pharmaceutical Science, Zhejiang University, Hangzhou, Zhejiang, China
| | - Andreas Linkermann
- Division of Nephrology, Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany
- Division of Nephrology, Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Junxia Min
- The Second Affiliated Hospital, The First Affiliated Hospital, School of Public Health, Institute of Translational Medicine, Cancer Center, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
| | - Fudi Wang
- The Second Affiliated Hospital, The First Affiliated Hospital, School of Public Health, Institute of Translational Medicine, Cancer Center, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
- The First Affiliated Hospital, Basic Medical Sciences, School of Public Health, Hengyang Medical School, University of South China, Hengyang, Hunan, China.
| |
Collapse
|
29
|
Zhou J, Fan Z, Bi Y, Li D, Chen X, Hou K, Ji S. The significance of serum Klotho to hearing loss: a potential protector under noise pollution. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2023; 30:104464-104476. [PMID: 37700133 DOI: 10.1007/s11356-023-29788-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 09/05/2023] [Indexed: 09/14/2023]
Abstract
The issue of hearing protection in the presence of noise pollution is of great importance in the fields of environmental science and clinical medicine. Currently, the clinical significance of Klotho in relation to hearing has not been revealed. The aim of this study was to examine the correlation between serum Klotho levels and Pure Tone Average (PTA) hearing thresholds among individuals in the U.S.. The analysis involved a sample of 1,781 individuals aged 20 to 69, obtained from the 2007-2012 National Health and Nutrition Examination Survey. Various methods were utilized for the analysis, including univariate and multivariate linear regression, stratified analysis, smooth curve fitting, a two-segment linear regression model, and log-likelihood ratio analysis. The results of the univariate analysis indicated that serum Klotho concentration, age, education level, hypertension, diabetes, and smoking all exhibited a significant influence on PTAs. After adjusting for potential confounding factors, it was observed that a decrease in serum Klotho was significantly associated with PTA thresholds at low frequency (β = -0.002; 95% CI: -0.003, -0.001; P = 0.004), speech frequency (β = -0.002; 95% CI: -0.003, -0.001; P = 0.007), and high frequency (β = -0.002; 95% CI: -0.003, -0.001; P = 0.045). Specifically, for every 1 pg/ml decrease in serum Klotho concentration, the PTAs increased by 0.002 dB. Moreover, age and gender-specific analyses revealed significant associations. For individuals aged 59-69, a significant association was found between serum Klotho concentration and high-frequency PTA (β = -4.153; 95% CI: -7.948, -0.358; P = 0.032). Additionally, among females, significant associations were observed between serum Klotho concentration and speech-frequency PTA (β = -1.648, 95% CI: -3.197, -0.099; P = 0.037) as well as high-frequency PTA (β = -3.046; 95% CI: -5.319, -0.772; P = 0.009). Finally, the results of smooth curve fitting and threshold effect analyses indicated a potential negative linear correlation between serum Klotho concentration and PTA thresholds. In conclusion, a lower level of serum Klotho was found to be associated with increased hearing thresholds, particularly among the elderly population. This finding has significant implications for the prevention and treatment of hearing damage.
Collapse
Affiliation(s)
- Jingcheng Zhou
- Senior Department of Otolaryngology-Head & Neck Surgery, Chinese PLA General Hospital, Beijing, 100083, China
- National Clinical Research Center for Otolaryngologic Diseases, Beijing, 100083, China
- State Key Lab of Hearing Science, Ministry of Education, Beijing, 100083, China
- Beijing Key Lab of Hearing Impairment Prevention and Treatment, Beijing, 100083, China
- Chinese PLA General Hospital and PLA Medical College, Beijing, 100083, China
| | - Zheqi Fan
- Chinese PLA General Hospital and PLA Medical College, Beijing, 100083, China
| | - Yiming Bi
- Chinese PLA General Hospital and PLA Medical College, Beijing, 100083, China
| | - Dingchang Li
- Chinese PLA General Hospital and PLA Medical College, Beijing, 100083, China
| | - Xuemin Chen
- Senior Department of Otolaryngology-Head & Neck Surgery, Chinese PLA General Hospital, Beijing, 100083, China
- National Clinical Research Center for Otolaryngologic Diseases, Beijing, 100083, China
- State Key Lab of Hearing Science, Ministry of Education, Beijing, 100083, China
- Beijing Key Lab of Hearing Impairment Prevention and Treatment, Beijing, 100083, China
- Chinese PLA General Hospital and PLA Medical College, Beijing, 100083, China
| | - Kun Hou
- Senior Department of Otolaryngology-Head & Neck Surgery, Chinese PLA General Hospital, Beijing, 100083, China
- National Clinical Research Center for Otolaryngologic Diseases, Beijing, 100083, China
- State Key Lab of Hearing Science, Ministry of Education, Beijing, 100083, China
- Beijing Key Lab of Hearing Impairment Prevention and Treatment, Beijing, 100083, China
- Chinese PLA General Hospital and PLA Medical College, Beijing, 100083, China
| | - Shuaifei Ji
- Chinese PLA General Hospital and PLA Medical College, Beijing, 100083, China.
- Research Center for Tissue Repair and Regeneration Affiliated To Medical Innovation Research Department, Chinese PLA General Hospital and PLA Medical College, 28 Fu Xing Road, Beijing, 100853, China.
- Medical School of Chinese PLA, Beijing, 100083, China.
| |
Collapse
|
30
|
Mattinzoli D, Molinari P, Romero-González G, Bover J, Cicero E, Pesce F, Abinti M, Conti C, Castellano G, Alfieri C. Is there a role in acute kidney injury for FGF23 and Klotho? Clin Kidney J 2023; 16:1555-1562. [PMID: 37779849 PMCID: PMC10539225 DOI: 10.1093/ckj/sfad093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Indexed: 10/03/2023] Open
Abstract
Cardio-renal syndrome is a clinical condition that has recently been well defined. In acute kidney disease, this interaction might trigger chronic processes determining the onset of cardiovascular events and the progression of chronic kidney disease. Moreover, the high mortality rate of acute kidney injury (AKI) is also linked to the fact that this condition is often complicated by dysfunctions of other organs such as lungs or heart, or is associated with septic episodes. In this context the role and the potential link between bone, heart and kidney is becoming an important topic of research. The aim of this review is to describe the cardiac alterations in the presence of AKI (cardiorenal syndrome type 3) and explore how bone can interact with heart and kidney in determining and influencing the trend of AKI in the short and long term. The main anomalies of mineral metabolism in patients with AKI will be reported, with specific reference to the alterations of fibroblast growth factor 23 and Klotho as a link between the bone-kidney-heart axis.
Collapse
Affiliation(s)
- Deborah Mattinzoli
- Department of Nephrology, Dialysis and Renal Transplantation, Fondazione IRCCS Ca’ Granda Ospedale Policlinico, Milan, Italy
| | - Paolo Molinari
- Department of Nephrology, Dialysis and Renal Transplantation, Fondazione IRCCS Ca’ Granda Ospedale Policlinico, Milan, Italy
- Post-Graduate School of Specialization in Nephrology, University of Milan, Milan, Italy
| | - Gregorio Romero-González
- Department of Nephrology, Germans Trias i Pujol University Hospital, Research Group on Renal Diseases (REMAR), Germans Trias i Pujol Research Institute, Badalona, Spain
| | - Jordi Bover
- Department of Nephrology, Germans Trias i Pujol University Hospital, Research Group on Renal Diseases (REMAR), Germans Trias i Pujol Research Institute, Badalona, Spain
| | - Elisa Cicero
- Department of Nephrology, Dialysis and Renal Transplantation, Fondazione IRCCS Ca’ Granda Ospedale Policlinico, Milan, Italy
- Post-Graduate School of Specialization in Nephrology, University of Milan, Milan, Italy
| | - Francesco Pesce
- Nephrology, Dialysis and Transplantation Unit Department of Precision and Regenerative Medicine and Ionian Area (DiMePre-J) University of Bari “Aldo Moro”
| | - Matteo Abinti
- Department of Nephrology, Dialysis and Renal Transplantation, Fondazione IRCCS Ca’ Granda Ospedale Policlinico, Milan, Italy
- Post-Graduate School of Specialization in Nephrology, University of Milan, Milan, Italy
| | - Costanza Conti
- Department of Nephrology, Dialysis and Renal Transplantation, Fondazione IRCCS Ca’ Granda Ospedale Policlinico, Milan, Italy
- Post-Graduate School of Specialization in Nephrology, University of Milan, Milan, Italy
| | - Giuseppe Castellano
- Department of Nephrology, Dialysis and Renal Transplantation, Fondazione IRCCS Ca’ Granda Ospedale Policlinico, Milan, Italy
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Carlo Alfieri
- Department of Nephrology, Dialysis and Renal Transplantation, Fondazione IRCCS Ca’ Granda Ospedale Policlinico, Milan, Italy
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| |
Collapse
|
31
|
Clemens Z, Wang K, Ambrosio F, Barchowsky A. Arsenic disrupts extracellular vesicle-mediated signaling in regenerating myofibers. Toxicol Sci 2023; 195:231-245. [PMID: 37527016 PMCID: PMC10535782 DOI: 10.1093/toxsci/kfad075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/03/2023] Open
Abstract
Chronic exposure to environmental arsenic is a public health crisis affecting hundreds of millions of individuals worldwide. Though arsenic is known to contribute to many pathologies and diseases, including cancers, cardiovascular and pulmonary diseases, and neurological impairment, the mechanisms for arsenic-promoted disease remain unresolved. This is especially true for arsenic impacts on skeletal muscle function and metabolism, despite the crucial role that skeletal muscle health plays in maintaining cardiovascular health, systemic homeostasis, and cognition. A barrier to researching this area is the challenge of interrogating muscle cell-specific effects in biologically relevant models. Ex vivo studies investigating mechanisms for muscle-specific responses to arsenic or other environmental contaminants primarily utilize traditional 2-dimensional culture models that cannot elucidate effects on muscle physiology or function. Therefore, we developed a contractile 3-dimensional muscle construct model-composed of primary mouse muscle progenitor cells differentiated in a hydrogel matrix-to study arsenic exposure impacts on skeletal muscle regeneration. Muscle constructs exposed to low-dose (50 nM) arsenic exhibited reduced strength and myofiber diameter following recovery from muscle injury. These effects were attributable to dysfunctional paracrine signaling mediated by extracellular vesicles (EVs) released from muscle cells. Specifically, we found that EVs collected from arsenic-exposed muscle constructs recapitulated the inhibitory effects of direct arsenic exposure on myofiber regeneration. In addition, muscle constructs treated with EVs isolated from muscles of arsenic-exposed mice displayed significantly decreased strength. Our findings highlight a novel model for muscle toxicity research and uncover a mechanism of arsenic-induced muscle dysfunction by the disruption of EV-mediated intercellular communication.
Collapse
Affiliation(s)
- Zachary Clemens
- Department of Environmental and Occupational Health, University of Pittsburgh School of Public Health, Pittsburgh, Pennsylvania, USA
| | - Kai Wang
- Discovery Center for Musculoskeletal Recovery, Schoen Adams Research Institute at Spaulding, Boston, Massachusetts, USA
- Department of Physical Medicine and Rehabilitation, Harvard Medical School, Boston, Massachusetts, USA
| | - Fabrisia Ambrosio
- Discovery Center for Musculoskeletal Recovery, Schoen Adams Research Institute at Spaulding, Boston, Massachusetts, USA
- Department of Physical Medicine and Rehabilitation, Harvard Medical School, Boston, Massachusetts, USA
| | - Aaron Barchowsky
- Department of Environmental and Occupational Health, University of Pittsburgh School of Public Health, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
32
|
Lathe R, St Clair D. Programmed ageing: decline of stem cell renewal, immunosenescence, and Alzheimer's disease. Biol Rev Camb Philos Soc 2023; 98:1424-1458. [PMID: 37068798 DOI: 10.1111/brv.12959] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 03/27/2023] [Accepted: 03/30/2023] [Indexed: 04/19/2023]
Abstract
The characteristic maximum lifespan varies enormously across animal species from a few hours to hundreds of years. This argues that maximum lifespan, and the ageing process that itself dictates lifespan, are to a large extent genetically determined. Although controversial, this is supported by firm evidence that semelparous species display evolutionarily programmed ageing in response to reproductive and environmental cues. Parabiosis experiments reveal that ageing is orchestrated systemically through the circulation, accompanied by programmed changes in hormone levels across a lifetime. This implies that, like the circadian and circannual clocks, there is a master 'clock of age' (circavital clock) located in the limbic brain of mammals that modulates systemic changes in growth factor and hormone secretion over the lifespan, as well as systemic alterations in gene expression as revealed by genomic methylation analysis. Studies on accelerated ageing in mice, as well as human longevity genes, converge on evolutionarily conserved fibroblast growth factors (FGFs) and their receptors, including KLOTHO, as well as insulin-like growth factors (IGFs) and steroid hormones, as key players mediating the systemic effects of ageing. Age-related changes in these and multiple other factors are inferred to cause a progressive decline in tissue maintenance through failure of stem cell replenishment. This most severely affects the immune system, which requires constant renewal from bone marrow stem cells. Age-related immune decline increases risk of infection whereas lifespan can be extended in germfree animals. This and other evidence suggests that infection is the major cause of death in higher organisms. Immune decline is also associated with age-related diseases. Taking the example of Alzheimer's disease (AD), we assess the evidence that AD is caused by immunosenescence and infection. The signature protein of AD brain, Aβ, is now known to be an antimicrobial peptide, and Aβ deposits in AD brain may be a response to infection rather than a cause of disease. Because some cognitively normal elderly individuals show extensive neuropathology, we argue that the location of the pathology is crucial - specifically, lesions to limbic brain are likely to accentuate immunosenescence, and could thus underlie a vicious cycle of accelerated immune decline and microbial proliferation that culminates in AD. This general model may extend to other age-related diseases, and we propose a general paradigm of organismal senescence in which declining stem cell proliferation leads to programmed immunosenescence and mortality.
Collapse
Affiliation(s)
- Richard Lathe
- Division of Infection Medicine, Chancellor's Building, University of Edinburgh Medical School, Little France, Edinburgh, EH16 4SB, UK
| | - David St Clair
- Institute of Medical Sciences, School of Medicine, University of Aberdeen, Aberdeen, AB25 2ZD, UK
| |
Collapse
|
33
|
Pai HL, Wu PY, Chen DM, Chen ZJ, Yang YS, Chang HH, Lin DPC. Klotho Null Mutation Involvement in Adenosine A2B Receptor-Related Skeletal Muscle Degeneration. THE AMERICAN JOURNAL OF PATHOLOGY 2023; 193:950-959. [PMID: 37028594 DOI: 10.1016/j.ajpath.2023.03.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 03/09/2023] [Accepted: 03/20/2023] [Indexed: 04/09/2023]
Abstract
Klotho is known for its age-suppressing function and has been implicated in sarcopenia pathology. It has recently been proposed that the adenosine A2B receptor plays a crucial role in skeletal muscle energy expenditure. However, the association between Klotho and A2B remains elusive. In this study, Klotho knockout mice, aged 10 weeks, and wild-type mice, aged 10 and 64 weeks, were used for comparison in indicators of sarcopenia (n = 6 for each group). PCR was performed to confirm the mice genotypes. Skeletal muscle sections were analyzed using hematoxylin and eosin staining as well as immunohistochemistry staining. The skeletal muscle cross-sectional area was significantly reduced in Klotho knockout mice and wild-type mice, aged 64 weeks, when compared with wild-type mice, aged 10 weeks, with a decreased percentage of type IIa and IIb myofibers. Likely impaired regenerative capacity, as reflected by the reduction of paired box 7 (Pax7)- and myogenic differentiation protein 1 (MyoD)-positive cells, was also observed in Klotho knockout mice and aged wild-type mice. 8-Hydroxy-2-deoxyguanosine expression was enhanced with Klotho knockout and aging, indicating higher oxidative stress. Adenosine A2B signaling was impaired, with a lower expression of the A2B receptor and the cAMP-response element binding protein in Klotho knockout and aged mice. The present study provides the novel finding that sarcopenia involves adenosine signaling under the influence of Klotho knockout.
Collapse
Affiliation(s)
- Hung-Liang Pai
- Department of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Pei-Yu Wu
- Department of Nutrition, Chung Shan Medical University, Taichung, Taiwan
| | - De-Ming Chen
- Department of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Zhi-Jia Chen
- Department of Medical Laboratory and Biotechnology, Chung Shan Medical University, Taichung, Taiwan
| | - Yi-Sun Yang
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Chung Shan Medical University Hospital, Taichung, Taiwan; Institute of Medicine, School of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Han-Hsin Chang
- Department of Nutrition, Chung Shan Medical University, Taichung, Taiwan.
| | - David Pei-Cheng Lin
- Department of Medical Laboratory and Biotechnology, Chung Shan Medical University, Taichung, Taiwan; Department of Ophthalmology, Chung Shan Medical University Hospital, Taichung, Taiwan.
| |
Collapse
|
34
|
Bean AC, Sahu A, Piechocki C, Gualerzi A, Picciolini S, Bedoni M, Ambrosio F. Neuromuscular electrical stimulation enhances the ability of serum extracellular vesicles to regenerate aged skeletal muscle after injury. Exp Gerontol 2023; 177:112179. [PMID: 37087025 PMCID: PMC10278579 DOI: 10.1016/j.exger.2023.112179] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 04/09/2023] [Accepted: 04/18/2023] [Indexed: 04/24/2023]
Abstract
Exercise promotes healthy aging of skeletal muscle. This benefit may be mediated by youthful factors in the circulation released in response to an exercise protocol. While numerous studies to date have explored soluble proteins as systemic mediators of rejuvenating effect of exercise on tissue function, here we showed that the beneficial effect of skeletal muscle contractile activity on aged muscle function is mediated, at least in part, by regenerative properties of circulating extracellular vesicles (EVs). Muscle contractile activity elicited by neuromuscular electrical stimulation (NMES) decreased intensity of expression of the tetraspanin surface marker, CD63, on circulating EVs. Moreover, NMES shifted the biochemical Raman fingerprint of circulating EVs in aged animals with significant changes in lipid and sugar content in response to NMES when compared to controls. As a demonstration of the physiological relevance of these EV changes, we showed that intramuscular administration of EVs derived from aged animals subjected to NMES enhanced aged skeletal muscle healing after injury. These studies suggest that repetitive muscle contractile activity enhances the regenerative properties of circulating EVs in aged animals.
Collapse
Affiliation(s)
- Allison C Bean
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, PA, United States of America; McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, United States of America.
| | - Amrita Sahu
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, PA, United States of America.
| | - Camilla Piechocki
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, PA, United States of America.
| | | | | | - Marzia Bedoni
- IRCCS Fondazione Don Carlo Gnocchi ONLUS, Milan, Italy.
| | - Fabrisia Ambrosio
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, PA, United States of America; McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, United States of America.
| |
Collapse
|
35
|
Iijima H, Wang K, D'Amico E, Tang WY, Rogers RJ, Jakicic JM, Ambrosio F. Exercise-primed extracellular vesicles improve cell-matrix adhesion and chondrocyte health. RESEARCH SQUARE 2023:rs.3.rs-2958821. [PMID: 37333349 PMCID: PMC10274961 DOI: 10.21203/rs.3.rs-2958821/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
Extracellular vesicles (EVs) have been suggested to transmit the health-promoting effects of exercise throughout the body. Yet, the mechanisms by which beneficial information is transmitted from extracellular vesicles to recipient cells are poorly understood, precluding a holistic understanding of how exercise promotes cellular and tissue health. In this study, using articular cartilage as a model, we introduced a network medicine paradigm to simulate how exercise facilitates communication between circulating EVs and chondrocytes, the cells resident in articular cartilage. Using the archived small RNA-seq data of EV before and after aerobic exercise, microRNA regulatory network analysis based on network propagation inferred that circulating EVs activated by aerobic exercise perturb chondrocyte-matrix interactions and downstream cellular aging processes. Building on the mechanistic framework identified through computational analyses, follow up experimental studies interrogated the direct influence of exercise on EV-mediated chondrocyte-matrix interactions. We found that pathogenic matrix signaling in chondrocytes was abrogated in the presence of exercise-primed EVs, restoring a more youthful phenotype, as determined by chondrocyte morphological profiling and evaluation of chondrogenicity. Epigenetic reprograming of the gene encoding the longevity protein, α-Klotho, mediated these effects. These studies provide mechanistic evidence that exercise transduces rejuvenation signals to circulating EVs, endowing EVs with the capacity to ameliorate cellular health even in the presence of an unfavorable microenvironmental signals.
Collapse
Affiliation(s)
- Hirotaka Iijima
- Institute for Advanced Research, Nagoya University, Nagoya, Japan
- Biomedical and Health Informatics Unit, Graduate School of Medicine, Nagoya University, Nagoya, Japan
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, PA
| | - Kai Wang
- Discovery Center for Musculoskeletal Recovery, Schoen Adams Research Institute at Spaulding, Boston, MA
- Department of Physical Medicine & Rehabilitation, Harvard Medical School, Boston, MA
| | - Ella D'Amico
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, PA
| | - Wan-Yee Tang
- Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, PA
| | - Renee J Rogers
- Department of Internal Medicine, Division of Physical Activity and Weight Management, University of Kansas Medical Center, Kansas City, KS
| | - John M Jakicic
- Department of Internal Medicine, Division of Physical Activity and Weight Management, University of Kansas Medical Center, Kansas City, KS
| | - Fabrisia Ambrosio
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, PA
- Discovery Center for Musculoskeletal Recovery, Schoen Adams Research Institute at Spaulding, Boston, MA
- Department of Physical Medicine & Rehabilitation, Harvard Medical School, Boston, MA
| |
Collapse
|
36
|
Corsetti G, Romano C, Pasini E, Scarabelli T, Chen-Scarabelli C, Dioguardi FS. Essential Amino Acids-Rich Diet Increases Cardiomyocytes Protection in Doxorubicin-Treated Mice. Nutrients 2023; 15:nu15102287. [PMID: 37242170 DOI: 10.3390/nu15102287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 05/08/2023] [Accepted: 05/10/2023] [Indexed: 05/28/2023] Open
Abstract
BACKGROUND Doxorubicin (Doxo) is a widely prescribed drug against many malignant cancers. Unfortunately, its utility is limited by its toxicity, in particular a progressive induction of congestive heart failure. Doxo acts primarily as a mitochondrial toxin, with consequent increased production of reactive oxygen species (ROS) and attendant oxidative stress, which drives cardiac dysfunction and cell death. A diet containing a special mixture of all essential amino acids (EAAs) has been shown to increase mitochondriogenesis, and reduce oxidative stress both in skeletal muscle and heart. So, we hypothesized that such a diet could play a favorable role in preventing Doxo-induced cardiomyocyte damage. METHODS Using transmission electron microscopy, we evaluated cells' morphology and mitochondria parameters in adult mice. In addition, by immunohistochemistry, we evaluated the expression of pro-survival marker Klotho, as well as markers of necroptosis (RIP1/3), inflammation (TNFα, IL1, NFkB), and defense against oxidative stress (SOD1, glutathione peroxidase, citrate synthase). RESULTS Diets with excess essential amino acids (EAAs) increased the expression of Klotho and enhanced anti-oxidative and anti-inflammatory responses, thereby promoting cell survival. CONCLUSION Our results further extend the current knowledge about the cardioprotective role of EAAs and provide a novel theoretical basis for their preemptive administration to cancer patients undergoing chemotherapy to alleviate the development and severity of Doxo-induced cardiomyopathy.
Collapse
Affiliation(s)
- Giovanni Corsetti
- Division of Human Anatomy and Physiopathology, Department of Clinical and Experimental Sciences, University of Brescia, 25123 Brescia, Italy
| | - Claudia Romano
- Division of Human Anatomy and Physiopathology, Department of Clinical and Experimental Sciences, University of Brescia, 25123 Brescia, Italy
| | - Evasio Pasini
- Italian Association of Functional Medicine, 20855 Lesmo (Milan), Italy
| | - Tiziano Scarabelli
- Center for Heart and Vessel Preclinical Studies, St. John Hospital and Medical Center, Wayne State University, Detroit, MI 48236, USA
| | - Carol Chen-Scarabelli
- Division of Cardiology, Richmond Veterans Affairs Medical Center (VAMC), Richmond, VA 23249, USA
| | - Francesco S Dioguardi
- Department of Internal Medicine, University of Cagliari, 09042 Monserrato (Cagliari), Italy
| |
Collapse
|
37
|
Han ZZ, Fleet A, Larrieu D. Can accelerated ageing models inform us on age-related tauopathies? Aging Cell 2023; 22:e13830. [PMID: 37013265 PMCID: PMC10186612 DOI: 10.1111/acel.13830] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 03/17/2023] [Accepted: 03/20/2023] [Indexed: 04/05/2023] Open
Abstract
Ageing is the greatest risk factor of late-onset neurodegenerative diseases. In the realm of sporadic tauopathies, modelling the process of biological ageing in experimental animals forms the foundation of searching for the molecular origin of pathogenic tau and developing potential therapeutic interventions. Although prior research into transgenic tau models offers valuable lessons for studying how tau mutations and overexpression can drive tau pathologies, the underlying mechanisms by which ageing leads to abnormal tau accumulation remains poorly understood. Mutations associated with human progeroid syndromes have been proposed to be able to mimic an aged environment in animal models. Here, we summarise recent attempts in modelling ageing in relation to tauopathies using animal models that carry mutations associated with human progeroid syndromes, or genetic elements unrelated to human progeroid syndromes, or have exceptional natural lifespans, or a remarkable resistance to ageing-related disorders.
Collapse
Affiliation(s)
- Zhuang Zhuang Han
- Department of PharmacologyUniversity of CambridgeTennis Ct RdCambridgeCB2 1PDUK
| | - Alex Fleet
- Department of PharmacologyUniversity of CambridgeTennis Ct RdCambridgeCB2 1PDUK
| | - Delphine Larrieu
- Department of PharmacologyUniversity of CambridgeTennis Ct RdCambridgeCB2 1PDUK
| |
Collapse
|
38
|
Zhao X, Zhou S, Liu Y, Gong C, Xiang L, Li S, Wang P, Wang Y, Sun L, Zhang Q, Yang Y. Parishin alleviates vascular ageing in mice by upregulation of Klotho. J Cell Mol Med 2023; 27:1398-1409. [PMID: 37032511 PMCID: PMC10183705 DOI: 10.1111/jcmm.17740] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 03/22/2023] [Accepted: 03/24/2023] [Indexed: 04/11/2023] Open
Abstract
Senescence of vascular endothelial cells is the major risk of vascular dysfunction and disease among elderly people. Parishin, which is a phenolic glucoside derived from Gastrodia elata, significantly prolonged yeast lifespan. However, the action of parishin in vascular ageing remains poorly understood. Here, we treated human coronary artery endothelial cells (HCAEC) and naturally aged mice by parishin. Parishin alleviated HCAEC senescence and general age-related features in vascular tissue in naturally aged mice. Network pharmacology approach was applied to determine the compound-target networks of parishin. Our analysis indicated that parishin had a strong binding affinity for Klotho. Expression of Klotho, a protein of age-related declines, was upregulated by parishin in serum and vascular tissue in naturally aged mice. Furthermore, FoxO1, on Klotho/FoxO1 signalling pathway, was increased in the parishin-intervened group, accompanied by the downregulated phosphorylated FoxO1. Taken together, parishin can increase Klotho expression to alleviate vascular endothelial cell senescence and vascular ageing.
Collapse
Affiliation(s)
- Xinxiu Zhao
- Department of Geriatrics, The First Affiliated Hospital, School of MedicineZhejiang UniversityHangzhouChina
- Key Laboratory of Diagnosis and Treatment of Aging and Physic‐chemical Injury Diseases of Zhejiang Province, The First Affiliated Hospital, School of MedicineZhejiang UniversityHangzhouZhejiangChina
| | - Shixian Zhou
- Department of Geriatrics, The First Affiliated Hospital, School of MedicineZhejiang UniversityHangzhouChina
- Key Laboratory of Diagnosis and Treatment of Aging and Physic‐chemical Injury Diseases of Zhejiang Province, The First Affiliated Hospital, School of MedicineZhejiang UniversityHangzhouZhejiangChina
| | - Yang Liu
- Department of Geriatrics, The First Affiliated Hospital, School of MedicineZhejiang UniversityHangzhouChina
- Key Laboratory of Diagnosis and Treatment of Aging and Physic‐chemical Injury Diseases of Zhejiang Province, The First Affiliated Hospital, School of MedicineZhejiang UniversityHangzhouZhejiangChina
| | - Caixia Gong
- Department of Geriatrics, The First Affiliated Hospital, School of MedicineZhejiang UniversityHangzhouChina
- Key Laboratory of Diagnosis and Treatment of Aging and Physic‐chemical Injury Diseases of Zhejiang Province, The First Affiliated Hospital, School of MedicineZhejiang UniversityHangzhouZhejiangChina
| | - Lan Xiang
- College of Pharmaceutical SciencesZhejiang University866 Yu Hang Tang RoadHangzhouChina
| | - Shumin Li
- Department of Geriatrics, The First Affiliated Hospital, School of MedicineZhejiang UniversityHangzhouChina
- Key Laboratory of Diagnosis and Treatment of Aging and Physic‐chemical Injury Diseases of Zhejiang Province, The First Affiliated Hospital, School of MedicineZhejiang UniversityHangzhouZhejiangChina
| | - Peixia Wang
- Department of Geriatrics, The First Affiliated Hospital, School of MedicineZhejiang UniversityHangzhouChina
- Key Laboratory of Diagnosis and Treatment of Aging and Physic‐chemical Injury Diseases of Zhejiang Province, The First Affiliated Hospital, School of MedicineZhejiang UniversityHangzhouZhejiangChina
| | - Yuejun Wang
- Zhejiang Aged Care HospitalHangzhou Normal UniversityHangzhouZhejiangChina
| | - Linlin Sun
- Department of Geriatrics, The First Affiliated Hospital, School of MedicineZhejiang UniversityHangzhouChina
- Key Laboratory of Diagnosis and Treatment of Aging and Physic‐chemical Injury Diseases of Zhejiang Province, The First Affiliated Hospital, School of MedicineZhejiang UniversityHangzhouZhejiangChina
| | - Qin Zhang
- Department of Geriatrics, The First Affiliated Hospital, School of MedicineZhejiang UniversityHangzhouChina
- Key Laboratory of Diagnosis and Treatment of Aging and Physic‐chemical Injury Diseases of Zhejiang Province, The First Affiliated Hospital, School of MedicineZhejiang UniversityHangzhouZhejiangChina
| | - Yunmei Yang
- Department of Geriatrics, The First Affiliated Hospital, School of MedicineZhejiang UniversityHangzhouChina
- Key Laboratory of Diagnosis and Treatment of Aging and Physic‐chemical Injury Diseases of Zhejiang Province, The First Affiliated Hospital, School of MedicineZhejiang UniversityHangzhouZhejiangChina
| |
Collapse
|
39
|
Picciolini S, Mangolini V, Rodà F, Montesano A, Arnaboldi F, Liuzzi P, Mannini A, Bedoni M, Gualerzi A. Multiplexing Biosensor for the Detection of Extracellular Vesicles as Biomarkers of Tissue Damage and Recovery after Ischemic Stroke. Int J Mol Sci 2023; 24:ijms24097937. [PMID: 37175644 PMCID: PMC10177901 DOI: 10.3390/ijms24097937] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 04/19/2023] [Accepted: 04/25/2023] [Indexed: 05/15/2023] Open
Abstract
The inflammatory, reparative and regenerative mechanisms activated in ischemic stroke patients immediately after the event cooperate in the response to injury, in the restoration of functions and in brain remodeling even weeks after the event and can be sustained by the rehabilitation treatment. Nonetheless, patients' response to treatments is difficult to predict because of the lack of specific measurable markers of recovery, which could be complementary to clinical scales in the evaluation of patients. Considering that Extracellular Vesicles (EVs) are carriers of multiple molecules involved in the response to stroke injury, in the present study, we have identified a panel of EV-associated molecules that (i) confirm the crucial involvement of EVs in the processes that follow ischemic stroke, (ii) could possibly profile ischemic stroke patients at the beginning of the rehabilitation program, (iii) could be used in predicting patients' response to treatment. By means of a multiplexing Surface Plasmon Resonance imaging biosensor, subacute ischemic stroke patients were proven to have increased expression of vascular endothelial growth factor receptor 2 (VEGFR2) and translocator protein (TSPO) on the surface of small EVs in blood. Besides, microglia EVs and endothelial EVs were shown to be significantly involved in the intercellular communications that occur more than 10 days after ischemic stroke, thus being potential tools for the profiling of patients in the subacute phase after ischemic stroke and in the prediction of their recovery.
Collapse
Affiliation(s)
| | - Valentina Mangolini
- IRCCS Fondazione Don Carlo Gnocchi ONLUS, 20148 Milano, Italy
- Dipartimento di Medicina Molecolare e Traslazionale, Università degli Studi di Brescia, 25122 Brescia, Italy
| | - Francesca Rodà
- IRCCS Fondazione Don Carlo Gnocchi ONLUS, 20148 Milano, Italy
- Clinical and Experimental Medicine PhD Program, University of Modena and Reggio Emilia, 42100 Modena, Italy
| | | | - Francesca Arnaboldi
- Dipartimento di Scienze Biomediche per la Salute, Università degli Studi di Milano, 20133 Milano, Italy
| | - Piergiuseppe Liuzzi
- IRCCS Fondazione Don Carlo Gnocchi ONLUS, 50143 Firenze, Italy
- Scuola Superiore Sant'Anna, Istituto di BioRobotica, 56025 Pontedera, Italy
| | - Andrea Mannini
- IRCCS Fondazione Don Carlo Gnocchi ONLUS, 50143 Firenze, Italy
| | - Marzia Bedoni
- IRCCS Fondazione Don Carlo Gnocchi ONLUS, 20148 Milano, Italy
| | - Alice Gualerzi
- IRCCS Fondazione Don Carlo Gnocchi ONLUS, 20148 Milano, Italy
| |
Collapse
|
40
|
Wang K, Smith SH, Iijima H, Hettinger ZR, Mallepally A, Shroff SG, Ambrosio F. Bioengineered 3D Skeletal Muscle Model Reveals Complement 4b as a Cell-Autonomous Mechanism of Impaired Regeneration with Aging. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2207443. [PMID: 36650030 DOI: 10.1002/adma.202207443] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 12/11/2022] [Indexed: 05/17/2023]
Abstract
A mechanistic understanding of cell-autonomous skeletal muscle changes after injury can lead to novel interventions to improve functional recovery in an aged population. However, major knowledge gaps persist owing to limitations of traditional biological aging models. 2D cell culture represents an artificial environment, while aging mammalian models are contaminated by influences from non-muscle cells and other organs. Here, a 3D muscle aging system is created to overcome the limitations of these traditional platforms. It is shown that old muscle constructs (OMC) manifest a sarcopenic phenotype, as evidenced by hypotrophic myotubes, reduced contractile function, and decreased regenerative capacity compared to young muscle constructs. OMC also phenocopy the regenerative responses of aged muscle to two interventions, pharmacological and biological. Interrogation of muscle cell-specific mechanisms that contribute to impaired regeneration over time further reveals that an aging-induced increase of complement component 4b (C4b) delays muscle progenitor cell amplification and impairs functional recovery. However, administration of complement factor I, a C4b inactivator, improves muscle regeneration in vitro and in vivo, indicating that C4b inhibition may be a novel approach to enhance aged muscle repair. Collectively, the model herein exhibits capabilities to study cell-autonomous changes in skeletal muscle during aging, regeneration, and intervention.
Collapse
Affiliation(s)
- Kai Wang
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, PA, 15213, USA
- Discovery Center for Musculoskeletal Recovery, Schoen Adams Research Institute at Spaulding, Charlestown, MA, 02129, USA
- Department of Physical Medicine and Rehabilitation, Harvard Medical School, Boston, MA, 02115, USA
| | - Stephen H Smith
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Hirotaka Iijima
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Zachary R Hettinger
- Discovery Center for Musculoskeletal Recovery, Schoen Adams Research Institute at Spaulding, Charlestown, MA, 02129, USA
- Department of Physical Medicine and Rehabilitation, Harvard Medical School, Boston, MA, 02115, USA
- Department of Medicine, Division of Geriatric Medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Adarsh Mallepally
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Sanjeev G Shroff
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, 15213, USA
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Fabrisia Ambrosio
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, PA, 15213, USA
- Discovery Center for Musculoskeletal Recovery, Schoen Adams Research Institute at Spaulding, Charlestown, MA, 02129, USA
- Department of Physical Medicine and Rehabilitation, Harvard Medical School, Boston, MA, 02115, USA
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, 15213, USA
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| |
Collapse
|
41
|
Sousa NS, Brás MF, Antunes IB, Lindholm P, Neves J, Sousa-Victor P. Aging disrupts MANF-mediated immune modulation during skeletal muscle regeneration. NATURE AGING 2023; 3:585-599. [PMID: 37118549 DOI: 10.1038/s43587-023-00382-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 02/16/2023] [Indexed: 04/30/2023]
Abstract
Age-related decline in skeletal muscle regenerative capacity is multifactorial, yet the contribution of immune dysfunction to regenerative failure is unknown. Macrophages are essential for effective debris clearance and muscle stem cell activity during muscle regeneration, but the regulatory mechanisms governing macrophage function during muscle repair are largely unexplored. Here, we uncover a new mechanism of immune modulation operating during skeletal muscle regeneration that is disrupted in aged animals and relies on the regulation of macrophage function. The immune modulator mesencephalic astrocyte-derived neurotrophic factor (MANF) is induced following muscle injury in young mice but not in aged animals, and its expression is essential for regenerative success. Regenerative impairments in aged muscle are associated with defects in the repair-associated myeloid response similar to those found in MANF-deficient models and could be improved through MANF delivery. We propose that restoring MANF levels is a viable strategy to improve myeloid response and regenerative capacity in aged muscle.
Collapse
Affiliation(s)
- Neuza S Sousa
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Margarida F Brás
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Inês B Antunes
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Päivi Lindholm
- Institute of Biotechnology, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Joana Neves
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal.
| | - Pedro Sousa-Victor
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal.
| |
Collapse
|
42
|
Ji S, Xiong M, Chen H, Liu Y, Zhou L, Hong Y, Wang M, Wang C, Fu X, Sun X. Cellular rejuvenation: molecular mechanisms and potential therapeutic interventions for diseases. Signal Transduct Target Ther 2023; 8:116. [PMID: 36918530 PMCID: PMC10015098 DOI: 10.1038/s41392-023-01343-5] [Citation(s) in RCA: 53] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 12/16/2022] [Accepted: 01/19/2023] [Indexed: 03/16/2023] Open
Abstract
The ageing process is a systemic decline from cellular dysfunction to organ degeneration, with more predisposition to deteriorated disorders. Rejuvenation refers to giving aged cells or organisms more youthful characteristics through various techniques, such as cellular reprogramming and epigenetic regulation. The great leaps in cellular rejuvenation prove that ageing is not a one-way street, and many rejuvenative interventions have emerged to delay and even reverse the ageing process. Defining the mechanism by which roadblocks and signaling inputs influence complex ageing programs is essential for understanding and developing rejuvenative strategies. Here, we discuss the intrinsic and extrinsic factors that counteract cell rejuvenation, and the targeted cells and core mechanisms involved in this process. Then, we critically summarize the latest advances in state-of-art strategies of cellular rejuvenation. Various rejuvenation methods also provide insights for treating specific ageing-related diseases, including cellular reprogramming, the removal of senescence cells (SCs) and suppression of senescence-associated secretory phenotype (SASP), metabolic manipulation, stem cells-associated therapy, dietary restriction, immune rejuvenation and heterochronic transplantation, etc. The potential applications of rejuvenation therapy also extend to cancer treatment. Finally, we analyze in detail the therapeutic opportunities and challenges of rejuvenation technology. Deciphering rejuvenation interventions will provide further insights into anti-ageing and ageing-related disease treatment in clinical settings.
Collapse
Affiliation(s)
- Shuaifei Ji
- Research Center for Tissue Repair and Regeneration Affiliated to Medical Innovation Research Department and 4th Medical Center, PLA General Hospital and PLA Medical College; PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration; Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, Beijing, 100048, P. R. China
| | - Mingchen Xiong
- Research Center for Tissue Repair and Regeneration Affiliated to Medical Innovation Research Department and 4th Medical Center, PLA General Hospital and PLA Medical College; PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration; Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, Beijing, 100048, P. R. China
| | - Huating Chen
- Research Center for Tissue Repair and Regeneration Affiliated to Medical Innovation Research Department and 4th Medical Center, PLA General Hospital and PLA Medical College; PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration; Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, Beijing, 100048, P. R. China
| | - Yiqiong Liu
- Research Center for Tissue Repair and Regeneration Affiliated to Medical Innovation Research Department and 4th Medical Center, PLA General Hospital and PLA Medical College; PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration; Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, Beijing, 100048, P. R. China
| | - Laixian Zhou
- Research Center for Tissue Repair and Regeneration Affiliated to Medical Innovation Research Department and 4th Medical Center, PLA General Hospital and PLA Medical College; PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration; Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, Beijing, 100048, P. R. China
| | - Yiyue Hong
- Research Center for Tissue Repair and Regeneration Affiliated to Medical Innovation Research Department and 4th Medical Center, PLA General Hospital and PLA Medical College; PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration; Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, Beijing, 100048, P. R. China
| | - Mengyang Wang
- Research Center for Tissue Repair and Regeneration Affiliated to Medical Innovation Research Department and 4th Medical Center, PLA General Hospital and PLA Medical College; PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration; Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, Beijing, 100048, P. R. China
| | - Chunming Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, 999078, Macau SAR, China.
| | - Xiaobing Fu
- Research Center for Tissue Repair and Regeneration Affiliated to Medical Innovation Research Department and 4th Medical Center, PLA General Hospital and PLA Medical College; PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration; Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, Beijing, 100048, P. R. China.
| | - Xiaoyan Sun
- Research Center for Tissue Repair and Regeneration Affiliated to Medical Innovation Research Department and 4th Medical Center, PLA General Hospital and PLA Medical College; PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration; Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, Beijing, 100048, P. R. China.
| |
Collapse
|
43
|
Zhao Y, Li H, Guo Q, Hui H. Multiple characteristic alterations and available therapeutic strategies of cellular senescence. J Zhejiang Univ Sci B 2023; 24:101-114. [PMID: 36751697 PMCID: PMC9936135 DOI: 10.1631/jzus.b2200178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
Given its state of stable proliferative inhibition, cellular senescence is primarily depicted as a critical mechanism by which organisms delay the progression of carcinogenesis. Cells undergoing senescence are often associated with the alteration of a series of specific features and functions, such as metabolic shifts, stemness induction, and microenvironment remodeling. However, recent research has revealed more complexity associated with senescence, including adverse effects on both physiological and pathological processes. How organisms evade these harmful consequences and survive has become an urgent research issue. Several therapeutic strategies targeting senescence, including senolytics, senomorphics, immunotherapy, and function restoration, have achieved initial success in certain scenarios. In this review, we describe in detail the characteristic changes associated with cellular senescence and summarize currently available countermeasures.
Collapse
Affiliation(s)
- Yunzi Zhao
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, 210009 China
| | - Hui Li
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, 210009 China
| | - Qinglong Guo
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, 210009 China
| | - Hui Hui
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
44
|
The Anti-Aging Hormone Klotho Promotes Retinal Pigment Epithelium Cell Viability and Metabolism by Activating the AMPK/PGC-1α Pathway. Antioxidants (Basel) 2023; 12:antiox12020385. [PMID: 36829944 PMCID: PMC9952846 DOI: 10.3390/antiox12020385] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 01/30/2023] [Accepted: 01/30/2023] [Indexed: 02/08/2023] Open
Abstract
Initially discovered by Makuto Kuro-o in 1997, Klotho is a putative aging-suppressor gene when overexpressed and accelerates aging when deleted in mice. Previously, we showed that α-Klotho regulates retinal pigment epithelium (RPE) functions and protects against oxidative stress. However, the mechanisms by which Klotho influences RPE and retinal homeostasis remain elusive. Here, by performing a series of in vitro and in vivo experiments, we demonstrate that Klotho regulates cell viability under oxidative stress, mitochondrial gene expression and activity by inducing the phosphorylation of AMPK and p38MAPK, which in turn phosphorylate and activate CREB and ATF2, respectively, triggering PGC-1α transcription. The inhibition of Klotho in human RPE cells using CRISPR-Cas9 gene editing confirmed that a lack of Klotho negatively affects RPE functions, including mitochondrial activity and cell viability. Proteomic analyses showed that myelin sheath and mitochondrial-related proteins are downregulated in the RPE/retina of Kl-/- compared to WT mice, further supporting our biochemical observations. We conclude that Klotho acts upstream of the AMPK/PGC-1α pathway and regulates RPE/retinal resistance to oxidative stress, mitochondrial function, and gene and protein expressions. Thus, KL decline during aging could negatively impact retinal health, inducing age-related retinal degeneration.
Collapse
|
45
|
Donate-Correa J, Martín-Carro B, Cannata-Andía JB, Mora-Fernández C, Navarro-González JF. Klotho, Oxidative Stress, and Mitochondrial Damage in Kidney Disease. Antioxidants (Basel) 2023; 12:239. [PMID: 36829798 PMCID: PMC9952437 DOI: 10.3390/antiox12020239] [Citation(s) in RCA: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 01/12/2023] [Accepted: 01/17/2023] [Indexed: 01/26/2023] Open
Abstract
Reducing oxidative stress stands at the center of a prevention and control strategy for mitigating cellular senescence and aging. Kidney disease is characterized by a premature aging syndrome, and to find a modulator targeting against oxidative stress, mitochondrial dysfunction, and cellular senescence in kidney cells could be of great significance to prevent and control the progression of this disease. This review focuses on the pathogenic mechanisms related to the appearance of oxidative stress damage and mitochondrial dysfunction in kidney disease. In this scenario, the anti-aging Klotho protein plays a crucial role by modulating signaling pathways involving the manganese-containing superoxide dismutase (Mn-SOD) and the transcription factors FoxO and Nrf2, known antioxidant systems, and other known mitochondrial function regulators, such as mitochondrial uncoupling protein 1 (UCP1), B-cell lymphoma-2 (BCL-2), Wnt/β-catenin, peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1 alpha), transcription factor EB, (TFEB), and peroxisome proliferator-activated receptor gamma (PPAR-gamma). Therefore, Klotho is postulated as a very promising new target for future therapeutic strategies against oxidative stress, mitochondria abnormalities, and cellular senescence in kidney disease patients.
Collapse
Affiliation(s)
- Javier Donate-Correa
- Unidad de Investigación, Hospital Universitario Nuestra Señora de Candelaria, 38010 Santa Cruz de Tenerife, Spain
- GEENDIAB (Grupo Español para el Estudio de la Nefropatía Diabética), Sociedad Española de Nefrología, 39008 Santander, Spain
- Instituto de Tecnologías Biomédicas, Universidad de La Laguna, 38010 San Cristóbal de La Laguna, Spain
- RICORS2040 (RD21/0005/0013), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Beatriz Martín-Carro
- RICORS2040 (RD21/0005/0019), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Bone and Mineral Research Unit, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
| | - Jorge B. Cannata-Andía
- RICORS2040 (RD21/0005/0019), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Bone and Mineral Research Unit, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
| | - Carmen Mora-Fernández
- Unidad de Investigación, Hospital Universitario Nuestra Señora de Candelaria, 38010 Santa Cruz de Tenerife, Spain
- GEENDIAB (Grupo Español para el Estudio de la Nefropatía Diabética), Sociedad Española de Nefrología, 39008 Santander, Spain
- RICORS2040 (RD21/0005/0013), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Juan F. Navarro-González
- Unidad de Investigación, Hospital Universitario Nuestra Señora de Candelaria, 38010 Santa Cruz de Tenerife, Spain
- GEENDIAB (Grupo Español para el Estudio de la Nefropatía Diabética), Sociedad Española de Nefrología, 39008 Santander, Spain
- Instituto de Tecnologías Biomédicas, Universidad de La Laguna, 38010 San Cristóbal de La Laguna, Spain
- RICORS2040 (RD21/0005/0013), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Servicio de Nefrología, Hospital Universitario Nuestra Señora de Candelaria, 38010 Santa Cruz de Tenerife, Spain
| |
Collapse
|
46
|
Iijima H, Gilmer G, Wang K, Bean AC, He Y, Lin H, Tang WY, Lamont D, Tai C, Ito A, Jones JJ, Evans C, Ambrosio F. Age-related matrix stiffening epigenetically regulates α-Klotho expression and compromises chondrocyte integrity. Nat Commun 2023; 14:18. [PMID: 36627269 PMCID: PMC9832042 DOI: 10.1038/s41467-022-35359-2] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Accepted: 11/29/2022] [Indexed: 01/12/2023] Open
Abstract
Extracellular matrix stiffening is a quintessential feature of cartilage aging, a leading cause of knee osteoarthritis. Yet, the downstream molecular and cellular consequences of age-related biophysical alterations are poorly understood. Here, we show that epigenetic regulation of α-Klotho represents a novel mechanosensitive mechanism by which the aged extracellular matrix influences chondrocyte physiology. Using mass spectrometry proteomics followed by a series of genetic and pharmacological manipulations, we discovered that increased matrix stiffness drove Klotho promoter methylation, downregulated Klotho gene expression, and accelerated chondrocyte senescence in vitro. In contrast, exposing aged chondrocytes to a soft matrix restored a more youthful phenotype in vitro and enhanced cartilage integrity in vivo. Our findings demonstrate that age-related alterations in extracellular matrix biophysical properties initiate pathogenic mechanotransductive signaling that promotes Klotho promoter methylation and compromises cellular health. These findings are likely to have broad implications even beyond cartilage for the field of aging research.
Collapse
Affiliation(s)
- Hirotaka Iijima
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, PA, USA.
- Japan Society for the Promotion of Science, Tokyo, Japan.
- Institute for Advanced Research, Nagoya University, Nagoya, Japan.
| | - Gabrielle Gilmer
- Medical Scientist Training Program, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA
- Cellular and Molecular Pathology Graduate Program, University of Pittsburgh, Pittsburgh, PA, USA
- Discovery Center for Musculoskeletal Recovery, Schoen Adams Research Institute at Spaulding, Boston, MA, USA
- Department of Physical Medicine & Rehabilitation, Harvard Medical School, Boston, MA, USA
| | - Kai Wang
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, PA, USA
- Discovery Center for Musculoskeletal Recovery, Schoen Adams Research Institute at Spaulding, Boston, MA, USA
- Department of Physical Medicine & Rehabilitation, Harvard Medical School, Boston, MA, USA
| | - Allison C Bean
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, PA, USA
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Yuchen He
- Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Hang Lin
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Wan-Yee Tang
- Department of Environmental and Occupational Health, University of Pittsburgh School of Public Health, Pittsburgh, PA, USA
| | - Daniel Lamont
- Petersen Institute of Nanoscience and Engineering, University of Pittsburgh, Pittsburgh, PA, USA
| | - Chia Tai
- Department of Motor Function Analysis, Human Health Sciences, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Akira Ito
- Department of Motor Function Analysis, Human Health Sciences, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Jeffrey J Jones
- Proteome Exploration Laboratory, Beckman Institute, California Institute of Technology, Pasadena, CA, USA
| | - Christopher Evans
- Department of Physical Medicine & Rehabilitation, Mayo Clinic, Rochester, MN, USA
| | - Fabrisia Ambrosio
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, PA, USA.
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA.
- Discovery Center for Musculoskeletal Recovery, Schoen Adams Research Institute at Spaulding, Boston, MA, USA.
- Department of Physical Medicine & Rehabilitation, Harvard Medical School, Boston, MA, USA.
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA.
- Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA, USA.
- Department of Environmental and Occupational Health, University of Pittsburgh School of Public Health, Pittsburgh, PA, USA.
| |
Collapse
|
47
|
Millozzi F, Papait A, Bouché M, Parolini O, Palacios D. Nano-Immunomodulation: A New Strategy for Skeletal Muscle Diseases and Aging? Int J Mol Sci 2023; 24:1175. [PMID: 36674691 PMCID: PMC9862642 DOI: 10.3390/ijms24021175] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 12/23/2022] [Accepted: 12/24/2022] [Indexed: 01/11/2023] Open
Abstract
The skeletal muscle has a very remarkable ability to regenerate upon injury under physiological conditions; however, this regenerative capacity is strongly diminished in physio-pathological conditions, such as those present in diseased or aged muscles. Many muscular dystrophies (MDs) are characterized by aberrant inflammation due to the deregulation of both the lymphoid and myeloid cell populations and the production of pro-inflammatory cytokines. Pathological inflammation is also observed in old muscles due to a systemic change in the immune system, known as "inflammaging". Immunomodulation represents, therefore, a promising therapeutic opportunity for different skeletal muscle conditions. However, the use of immunomodulatory drugs in the clinics presents several caveats, including their low stability in vivo, the need for high doses to obtain therapeutically relevant effects, and the presence of strong side effects. Within this context, the emerging field of nanomedicine provides the powerful tools needed to control the immune response. Nano-scale materials are currently being explored as biocarriers to release immunomodulatory agents in the damaged tissues, allowing therapeutic doses with limited off-target effects. In addition, the intrinsic immunomodulatory properties of some nanomaterials offer further opportunities for intervention that still need to be systematically explored. Here we exhaustively review the state-of-the-art regarding the use of nano-sized materials to modulate the aberrant immune response that characterizes some physio-pathological muscle conditions, such as MDs or sarcopenia (the age-dependent loss of muscle mass). Based on our learnings from cancer and immune tolerance induction, we also discuss further opportunities, challenges, and limitations of the emerging field of nano-immunomodulation.
Collapse
Affiliation(s)
- Francesco Millozzi
- Department of Anatomical, Histological, Forensic Medicine and Orthopaedic Sciences, Section of Histology and Embryology, Sapienza University of Rome, 00161 Rome, Italy
- IRCCS Fondazione Santa Lucia, Via del Fosso di Fiorano, 64, 00143 Rome, Italy
| | - Andrea Papait
- Department of Life Sciences and Public Health, Università Cattolica del Sacro Cuore, Largo Vito, 1, 00168 Rome, Italy
- IRCCS Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo Vito, 1, 00168 Rome, Italy
| | - Marina Bouché
- Department of Anatomical, Histological, Forensic Medicine and Orthopaedic Sciences, Section of Histology and Embryology, Sapienza University of Rome, 00161 Rome, Italy
| | - Ornella Parolini
- Department of Life Sciences and Public Health, Università Cattolica del Sacro Cuore, Largo Vito, 1, 00168 Rome, Italy
- IRCCS Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo Vito, 1, 00168 Rome, Italy
| | - Daniela Palacios
- Department of Life Sciences and Public Health, Università Cattolica del Sacro Cuore, Largo Vito, 1, 00168 Rome, Italy
- IRCCS Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo Vito, 1, 00168 Rome, Italy
| |
Collapse
|
48
|
Cai J, Zhang L, Chen C, Ge J, Li M, Zhang Y, Liu H, Song B. Association between serum Klotho concentration and heart failure in adults, a cross-sectional study from NHANES 2007-2016. Int J Cardiol 2023; 370:236-243. [PMID: 36351541 DOI: 10.1016/j.ijcard.2022.11.010] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 10/08/2022] [Accepted: 11/02/2022] [Indexed: 11/08/2022]
Abstract
BACKGROUND Limited data exist on the association between serum Klotho concentration and heart failure (HF). METHODS AND RESULTS We conducted a cross-sectional study of 13,625 participants aged 40-79 years in the National Health and Nutrition Examination Survey (NHANES) 2007-2016. Multivariable logistic regression models were used to examine the association between serum Klotho concentration (ln transformation) and HF. A total of 533 (2.9%) participants were identified to have HF, and participants with the lowest tertiles of serum Klotho concentration had the highest percentage of HF (T1: 3.8% vs. T2: 2.8% and T3: 2.1%, P < 0.001). After adjusting for potential confounders, ln (Klotho) was negatively and independently associated with the risk of HF (OR= 0.55, 95% CI 0.36-0.84). Meanwhile, compared with the T1 group, a higher serum Klotho concentration was associated with a lower risk of HF (tertile 2: OR = 0.93, 95% CI: 0.69-1.29, tertile 3: OR = 0.75, 95% CI: 0.52-1.09, P for trend 0.022). Finally, subgroup analyses indicated that lower Klotho concentrations significantly correlated with an increased risk of HF in half of the subgroups. CONCLUSION Serum Klotho concentration was consistently and negatively associated with the presence of HF among US middle-aged and older adults.
Collapse
Affiliation(s)
- Jiasheng Cai
- Departments of Cardiology, Fudan University Zhongshan Hospital, Qingpu Branch, 1158 Park East Road, Shanghai 60518120, China.
| | - Lingyun Zhang
- Department of Nephrology, Huadong Hospital affiliated to Fudan University, 221 Yanan West Road, Shanghai 200040, China.
| | - Conggai Chen
- Department of Stroke Center, the First Affiliated Hospital of Soochow University, 188 Shizi Street Suzhou, Jiangsu Province 215006, China
| | - Jin Ge
- Department of General medicine, Renji Hospital affiliated to JiaoTong University, 1630 Pujian Road, Shanghai 200127, China
| | - Mingxuan Li
- Departments of Cardiology, Fudan University Zhongshan Hospital, Qingpu Branch, 1158 Park East Road, Shanghai 60518120, China
| | - Yi Zhang
- Departments of Cardiology, Fudan University Zhongshan Hospital, Qingpu Branch, 1158 Park East Road, Shanghai 60518120, China
| | - Haibo Liu
- Departments of Cardiology, Fudan University Zhongshan Hospital, Qingpu Branch, 1158 Park East Road, Shanghai 60518120, China.
| | - Bin Song
- Department of Chronic Diseases Management, Clinical Medical College, Yangzhou University, 98 Nantong West Road, Yangzhou 225001, China.
| |
Collapse
|
49
|
Advances in Human Mitochondria-Based Therapies. Int J Mol Sci 2022; 24:ijms24010608. [PMID: 36614050 PMCID: PMC9820658 DOI: 10.3390/ijms24010608] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Revised: 12/19/2022] [Accepted: 12/23/2022] [Indexed: 12/31/2022] Open
Abstract
Mitochondria are the key biological generators of eukaryotic cells, controlling the energy supply while providing many important biosynthetic intermediates. Mitochondria act as a dynamic, functionally and structurally interconnected network hub closely integrated with other cellular compartments via biomembrane systems, transmitting biological information by shuttling between cells and tissues. Defects and dysregulation of mitochondrial functions are critically involved in pathological mechanisms contributing to aging, cancer, inflammation, neurodegenerative diseases, and other severe human diseases. Mediating and rejuvenating the mitochondria may therefore be of significant benefit to prevent, reverse, and even treat such pathological conditions in patients. The goal of this review is to present the most advanced strategies using mitochondria to manage such disorders and to further explore innovative approaches in the field of human mitochondria-based therapies.
Collapse
|
50
|
Luthra NS, Clow A, Corcos DM. The Interrelated Multifactorial Actions of Cortisol and Klotho: Potential Implications in the Pathogenesis of Parkinson's Disease. Brain Sci 2022; 12:1695. [PMID: 36552155 PMCID: PMC9775285 DOI: 10.3390/brainsci12121695] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 12/06/2022] [Accepted: 12/07/2022] [Indexed: 12/14/2022] Open
Abstract
The pathogenesis of Parkinson's disease (PD) is complex, multilayered, and not fully understood, resulting in a lack of effective disease-modifying treatments for this prevalent neurodegenerative condition. Symptoms of PD are heterogenous, including motor impairment as well as non-motor symptoms such as depression, cognitive impairment, and circadian disruption. Aging and stress are important risk factors for PD, leading us to explore pathways that may either accelerate or protect against cellular aging and the detrimental effects of stress. Cortisol is a much-studied hormone that can disrupt mitochondrial function and increase oxidative stress and neuroinflammation, which are recognized as key underlying disease mechanisms in PD. The more recently discovered klotho protein, considered a general aging-suppressor, has a similarly wide range of actions but in the opposite direction to cortisol: promoting mitochondrial function while reducing oxidative stress and inflammation. Both hormones also converge on pathways of vitamin D metabolism and insulin resistance, also implicated to play a role in PD. Interestingly, aging, stress and PD associate with an increase in cortisol and decrease in klotho, while physical exercise and certain genetic variations lead to a decrease in cortisol response and increased klotho. Here, we review the interrelated opposite actions of cortisol and klotho in the pathogenesis of PD. Together they impact powerful and divergent mechanisms that may go on to influence PD-related symptoms. Better understanding of these hormones in PD would facilitate the design of effective interventions that can simultaneously impact the multiple systems involved in the pathogenesis of PD.
Collapse
Affiliation(s)
- Nijee S. Luthra
- Department of Neurology, University of California San Francisco, San Francisco, CA 94127, USA
| | - Angela Clow
- Department of Psychology, School of Social Sciences, University of Westminster, London W1B 2HW, UK
| | - Daniel M. Corcos
- Department of Physical Therapy & Human Movement Sciences, Feinberg School of Medicine, Northwestern University, Chicago, IL 60208, USA
| |
Collapse
|