1
|
Zhu Y, Sun L, Hou M, Yu J, Yu C, Zhang Z, Yang H, Liu C, Huang L, Jiang D, Zhang Y, Yuan Y, Zhu X. An "inside-out"-guided genetically engineered hydrogel for augmenting aged bone regeneration. Bioact Mater 2025; 51:318-332. [PMID: 40491685 PMCID: PMC12146016 DOI: 10.1016/j.bioactmat.2025.05.003] [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: 03/12/2025] [Revised: 04/24/2025] [Accepted: 05/05/2025] [Indexed: 06/11/2025] Open
Abstract
Senescent bone repair faces significant obstacles due to reduced cellular activity and an unfavorable microenvironment, both of which hinder the osteogenic differentiation of bone marrow-derived stem cells (BMSCs) into osteoblasts (OBs) and subsequent bone formation. Current approaches primarily target senescent cell clearance (senolytics) or suppression of the senescence-associated secretory phenotype (senomorphics), neglecting the complex interactions between BMSCs and the osteogenic microenvironment. In this study, a genetically engineered hydrogel incorporating NAD-dependent deacetylase sirtuins 3 (SIRT3)-loaded nano-vectors and poly (glycerol sebacate)-co-poly (ethylene glycol)/polyacrylic acid (PEGS/PAA) was developed as an "inside-out" strategy for bone regeneration. At the intracellular level, BMSC function is restored, and osteogenesis is promoted through genetically enhanced SIRT3 expression. At the extracellular level, carboxyl functional groups chelate iron ions, simulating a hypoxic environment and promoting synergistic interactions between angiogenesis and osteogenesis. The therapeutic effects of the genetically engineered hydrogel in alleviating senescent damage and enhancing osteogenic differentiation were confirmed in both chemically and naturally induced senescence models in vitro. Local delivery of the hydrogel significantly increased newly formed bone in rat cranial defects. Mechanistically, the central role of SIRT3 in balancing senescence and osteogenesis, as well as its involvement in bone immune signaling pathways, was elucidated through CRISPR/Cas9-mediated editing in mice and transcriptome sequencing. This work presents a novel paradigm that integrates cellular and microenvironmental factors to enhance bone regeneration, offering new hope for treating age-related bone injuries.
Collapse
Affiliation(s)
- Yanrun Zhu
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, 215006, China
- Orthopaedic Institute, Medical College, Soochow University, Suzhou, 215000, China
- Xuzhou Medical University Affiliated Hospital Sihong Branch, The First People's Hospital of Sihong County, Suqian, 223900, China
| | - Lili Sun
- Key Laboratory for Ultrafine Materials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai, 200237, China
- Engineering Research Center for Biomedical Materials of Ministry of Education, East China University of Science and Technology, Shanghai, 200237, China
| | - Mingzhuang Hou
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, 215006, China
- Orthopaedic Institute, Medical College, Soochow University, Suzhou, 215000, China
| | - Jianfeng Yu
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, 215006, China
- Orthopaedic Institute, Medical College, Soochow University, Suzhou, 215000, China
| | - Chenqi Yu
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, 215006, China
- Orthopaedic Institute, Medical College, Soochow University, Suzhou, 215000, China
| | - Zihan Zhang
- Xi'an Jiaotong-Liverpool University, Suzhou, 215123, China
| | - Huilin Yang
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, 215006, China
- Orthopaedic Institute, Medical College, Soochow University, Suzhou, 215000, China
| | - Changsheng Liu
- Key Laboratory for Ultrafine Materials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai, 200237, China
- Engineering Research Center for Biomedical Materials of Ministry of Education, East China University of Science and Technology, Shanghai, 200237, China
| | - Lixin Huang
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, 215006, China
- Orthopaedic Institute, Medical College, Soochow University, Suzhou, 215000, China
| | - Dinghua Jiang
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, 215006, China
- Orthopaedic Institute, Medical College, Soochow University, Suzhou, 215000, China
| | - Yijian Zhang
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, 215006, China
- Orthopaedic Institute, Medical College, Soochow University, Suzhou, 215000, China
| | - Yuan Yuan
- Key Laboratory for Ultrafine Materials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai, 200237, China
- Engineering Research Center for Biomedical Materials of Ministry of Education, East China University of Science and Technology, Shanghai, 200237, China
| | - Xuesong Zhu
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, 215006, China
- Orthopaedic Institute, Medical College, Soochow University, Suzhou, 215000, China
- Department of Orthopaedics, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, 215002, China
| |
Collapse
|
2
|
Ahn B, Wanagat J, Cleary C, Ainsworth HC, Kim E, Kim H. Unacylated Ghrelin Counteracts Contractile and Mitochondrial Dysfunction in Cancer Cachexia. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.29.649515. [PMID: 40376088 PMCID: PMC12080946 DOI: 10.1101/2025.04.29.649515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2025]
Abstract
Background Cancer cachexia is a complex metabolic syndrome that severely impacts patient mobility, treatment strategies, and quality of life. However, no treatments are available to mitigate the debilitating consequences of cancer cachexia. Unacylated ghrelin (UnAG), the main circulating form of ghrelin, enhances muscle growth and mitochondrial function in various diseases, but its effects in cancer cachexia remain to be tested. Methods Male C57Bl6/N mice were assigned to one of three treatment groups: non-tumor-bearing (NTB), tumor-bearing (TB), or tumor-bearing treated with unacylated ghrelin (TB+UnAG). Over four weeks, we monitored body weight, food intake, and tumor size. We assessed muscle mass, contractility, mitochondrial oxygen consumption rate (OCR), and reactive oxygen species (ROS) production. Proteomic analysis was performed to elucidate the downstream effects of UnAG. Cell culture assays were performed to measure the in vitro effects of cancer cell-secreted factors and UnAG on myoblasts. Results Gastrocnemius and quadriceps muscle masses were reduced by 20-30% in TB mice compared to NTB controls; however, UnAG treatment prevented approximately 50% of this loss. Beyond muscle mass, UnAG enhanced the isometric maximum specific force of the extensor digitorum longus by 70% in TB mice. This improvement in muscle quality was associated with preferential upregulation of myosin heavy chain expression in TB+UnAG mice. UnAG also increased mitochondrial OCR while reducing ROS production. Mitochondrial DNA (mtDNA) copy number, which was reduced in TB mice, was restored by UnAG, while the reduced mtDNA mutation frequency in TB mice was maintained with treatment, indicating improved mtDNA integrity. Consistent with enhanced mitochondrial function, treadmill running time was significantly increased in TB+UnAG mice. Proteomic analysis revealed that UnAG downregulated proteins associated with proteolysis, while normalizing antioxidant enzyme thioredoxin and proteins involved in calcium handling. Cancer cell-conditioned medium reduced myotube width in vitro, but UnAG treatment preserved myotube structure.. Conclusion UnAG protects against cancer cachexia by targeting multiple risk factors, including myosin heavy chain expression, mitochondrial bioenergetics, and modulation of protein degradation pathways.
Collapse
|
3
|
Miranda ER, Shahtout JL, Watanabe S, Milam N, Karasawa T, Rout S, Atkinson DL, Holland WL, Drummond MJ, Funai K. Muscle-specific Keap1 deletion enhances force production but does not prevent inactivity-induced muscle atrophy in mice. FASEB J 2025; 39:e70464. [PMID: 40085142 PMCID: PMC11908634 DOI: 10.1096/fj.202402810r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 02/14/2025] [Accepted: 03/05/2025] [Indexed: 03/16/2025]
Abstract
Immobilization-associated muscle atrophy and weakness appear to be driven in part by oxidative stress. Nuclear Factor Erythroid 2-Related Factor 2 (NRF2) is a critical redox rheostat that regulates oxidative stress responses, and its deletion is known to accelerate muscle atrophy and weakness during aging (sarcopenia) or denervation. Conversely, pharmacologic activation of NRF2 extends mouse lifespan and attenuates sarcopenia. Similarly, deletion of Kelch-like ECH-associated Protein 1 (Keap1), a negative regulator of NRF2, enhances exercise capacity. The purpose of this study was to determine whether muscle-specific Keap1 deletion is sufficient to prevent muscle atrophy and weakness in mice following 7 days of hindlimb unloading (HU). To test this hypothesis, control (Ctrl) and tamoxifen-inducible, muscle-specific Keap1 knockout (mKO) mice were subjected to either normal housing (Sham) or HU for 7 days. Activation of NRF2 in muscle was confirmed by increased mRNA of NRF2 targets thioredoxin 1 (Txn1) and NAD(P)H quinone dehydrogenase 1 (NQO1) in mKO mice. Keap1 deletion had an effect to increase force-generating capacity at baseline. However, muscle masses, cross-sectional area, and ex vivo force were not different between mKO and Ctrl HU mice. In addition, muscle 4-hydroxynonenal-modified proteins and protein carbonyls were unaffected by Keap1 deletion. These data suggest that NRF2 activation improves muscle force production during ambulatory conditions but is not sufficient to prevent muscle atrophy or weakness following 7 days of HU.
Collapse
Affiliation(s)
- Edwin R. Miranda
- Diabetes & Metabolism Research CenterUniversity of UtahSalt Lake CityUtahUSA
- Department of Nutrition and Integrative PhysiologyUniversity of UtahSalt Lake CityUtahUSA
- Molecular Medicine ProgramUniversity of UtahSalt Lake CityUtahUSA
| | - Justin L. Shahtout
- Diabetes & Metabolism Research CenterUniversity of UtahSalt Lake CityUtahUSA
- Department of Physical Therapy & Athletic TrainingUniversity of UtahSalt Lake CityUtahUSA
| | - Shinya Watanabe
- Diabetes & Metabolism Research CenterUniversity of UtahSalt Lake CityUtahUSA
- Department of Nutrition and Integrative PhysiologyUniversity of UtahSalt Lake CityUtahUSA
| | - Norah Milam
- Diabetes & Metabolism Research CenterUniversity of UtahSalt Lake CityUtahUSA
| | - Takuya Karasawa
- Diabetes & Metabolism Research CenterUniversity of UtahSalt Lake CityUtahUSA
- Department of Nutrition and Integrative PhysiologyUniversity of UtahSalt Lake CityUtahUSA
- Molecular Medicine ProgramUniversity of UtahSalt Lake CityUtahUSA
| | - Subhasmita Rout
- Diabetes & Metabolism Research CenterUniversity of UtahSalt Lake CityUtahUSA
- Department of Nutrition and Integrative PhysiologyUniversity of UtahSalt Lake CityUtahUSA
| | - Donald L. Atkinson
- Diabetes & Metabolism Research CenterUniversity of UtahSalt Lake CityUtahUSA
- Department of Nutrition and Integrative PhysiologyUniversity of UtahSalt Lake CityUtahUSA
| | - William L. Holland
- Diabetes & Metabolism Research CenterUniversity of UtahSalt Lake CityUtahUSA
- Department of Nutrition and Integrative PhysiologyUniversity of UtahSalt Lake CityUtahUSA
- Molecular Medicine ProgramUniversity of UtahSalt Lake CityUtahUSA
| | - Micah J. Drummond
- Diabetes & Metabolism Research CenterUniversity of UtahSalt Lake CityUtahUSA
- Department of Nutrition and Integrative PhysiologyUniversity of UtahSalt Lake CityUtahUSA
- Department of Physical Therapy & Athletic TrainingUniversity of UtahSalt Lake CityUtahUSA
| | - Katsuhiko Funai
- Diabetes & Metabolism Research CenterUniversity of UtahSalt Lake CityUtahUSA
- Department of Nutrition and Integrative PhysiologyUniversity of UtahSalt Lake CityUtahUSA
- Molecular Medicine ProgramUniversity of UtahSalt Lake CityUtahUSA
- Department of Physical Therapy & Athletic TrainingUniversity of UtahSalt Lake CityUtahUSA
| |
Collapse
|
4
|
Nirmala FS, Lee H, Cho Y, Um MY, Seo HD, Jung CH, Hahm JH, Ahn J. Norharmane prevents muscle aging via activation of SKN-1/NRF2 stress response pathways. Redox Biol 2025; 80:103512. [PMID: 39874928 PMCID: PMC11810848 DOI: 10.1016/j.redox.2025.103512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2024] [Accepted: 01/21/2025] [Indexed: 01/30/2025] Open
Abstract
Sarcopenia, the age-related decline in muscle mass and function, is a significant contributor to increased frailty and mortality in the elderly. Currently, no FDA-approved treatment exists for sarcopenia. Here, we identified norharmane (NR), a β-carboline alkaloid, as a potential therapeutic agent for mitigating muscle aging. We aimed to determine the ability of NR to delay muscle aging in Caenorhabditis elegans (C. elegans), mouse, and muscle cells in mice and humans. NR treatment improved swimming ability and increased the maximum velocity in aged C. elegans. Transcriptomic analysis revealed that NR upregulated detoxification genes in C. elegans, including cytochrome P450, UGT, and GST enzymes. NR-induced benefits were dependent on the SKN-1/Nrf2 stress response pathway. In mammalian models, NR delayed cellular senescence in human skeletal muscle myoblasts and enhanced myogenesis in C2C12 cells and primary aged myoblasts. NR supplementation in aged mice prevented muscle loss, improved muscle function, and reduced markers of cellular senescence. We found that the p38 MAPK pathway mediated NR activation of Nrf2 by disrupting the Nrf2-Keap1 interaction. NR also improved oxygen consumption rates and promoted mitochondrial biogenesis. These findings suggest that NR is a promising candidate for preventing sarcopenia and improving muscle health.
Collapse
Affiliation(s)
- Farida S Nirmala
- Aging and Metabolism Research Group, Korea Food Research Institute, Wanju-gun, South Korea; Department of Food Biotechnology, Korea University of Science and Technology, Daejeon-si, South Korea
| | - Hyunjung Lee
- Aging and Metabolism Research Group, Korea Food Research Institute, Wanju-gun, South Korea
| | - Yejin Cho
- Aging and Metabolism Research Group, Korea Food Research Institute, Wanju-gun, South Korea
| | - Min Young Um
- Functional Food Materials Research Group, Korea Food Research Institute, Wanju-gun, South Korea; Department of Food Biotechnology, Korea University of Science and Technology, Daejeon-si, South Korea
| | - Hyo Deok Seo
- Department of Food Biotechnology, Korea University of Science and Technology, Daejeon-si, South Korea
| | - Chang Hwa Jung
- Aging and Metabolism Research Group, Korea Food Research Institute, Wanju-gun, South Korea; Department of Food Biotechnology, Korea University of Science and Technology, Daejeon-si, South Korea
| | - Jeong-Hoon Hahm
- Aging and Metabolism Research Group, Korea Food Research Institute, Wanju-gun, South Korea.
| | - Jiyun Ahn
- Aging and Metabolism Research Group, Korea Food Research Institute, Wanju-gun, South Korea; Department of Food Biotechnology, Korea University of Science and Technology, Daejeon-si, South Korea.
| |
Collapse
|
5
|
Xu H, Brown JL, Bhaskaran S, Van Remmen H. Reactive oxygen species in the pathogenesis of sarcopenia. Free Radic Biol Med 2025; 227:446-458. [PMID: 39613046 PMCID: PMC11816180 DOI: 10.1016/j.freeradbiomed.2024.11.046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 11/06/2024] [Accepted: 11/21/2024] [Indexed: 12/01/2024]
Abstract
One of the most critical factors impacting healthspan in the elderly is the loss of muscle mass and function, clinically referred to as sarcopenia. Muscle atrophy and weakness lead to loss of mobility, increased risk of injury, metabolic changes and loss of independence. Thus, defining the underlying mechanisms of sarcopenia is imperative to enable the development of effective interventions to preserve muscle function and quality in the elderly and improve healthspan. Over the past few decades, understanding the roles of mitochondrial dysfunction and oxidative stress has been a major focus of studies seeking to reveal critical molecular pathways impacted during aging. In this review, we will highlight how oxidative stress might contribute to sarcopenia by discussing the impact of oxidative stress on the loss of innervation and alteration in the neuromuscular junction (NMJ), on muscle mitochondrial function and atrophy pathways, and finally on muscle contractile function.
Collapse
Affiliation(s)
- Hongyang Xu
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, United States
| | - Jacob L Brown
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, United States; Oklahoma City VA Medical Center, Oklahoma City, OK, 73104, United States
| | - Shylesh Bhaskaran
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, United States
| | - Holly Van Remmen
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, United States; Oklahoma City VA Medical Center, Oklahoma City, OK, 73104, United States.
| |
Collapse
|
6
|
Kim H, Ranjit R, Claflin DR, Georgescu C, Wren JD, Brooks SV, Miller BF, Ahn B. Unacylated Ghrelin Protects Against Age-Related Loss of Muscle Mass and Contractile Dysfunction in Skeletal Muscle. Aging Cell 2024; 23:e14323. [PMID: 39223708 PMCID: PMC11634730 DOI: 10.1111/acel.14323] [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: 03/08/2024] [Revised: 07/20/2024] [Accepted: 08/10/2024] [Indexed: 09/04/2024] Open
Abstract
Sarcopenia, the progressive loss of muscle mass and function, universally affects older adults and is closely associated with frailty and reduced quality of life. Despite the inevitable consequences of sarcopenia and its relevance to healthspan, no pharmacological therapies are currently available. Ghrelin is a gut-released hormone that increases appetite and body weight through acylation. Acylated ghrelin activates its receptor, growth hormone secretagogue receptor 1a (GHSR1a), in the brain by binding to it. Studies have demonstrated that acyl and unacylated ghrelin (UnAG) both have protective effects against acute pathological conditions independent of receptor activation. Here, we investigated the long-term effects of UnAG in age-associated muscle atrophy and contractile dysfunction in mice. Four-month-old and 18-month-old mice were subjected to either UnAG or control treatment for 10 months. UnAG did not affect food consumption or body weight. Gastrocnemius and quadriceps muscle weights were reduced by 20%-30% with age, which was partially protected against by UnAG. Specific force, force per cross-sectional area, measured in isolated extensor digitorum longus muscle was diminished by 30% in old mice; however, UnAG prevented the loss of specific force. UnAG also protected from decreases in mitochondrial respiration and increases in hydrogen peroxide generation of skeletal muscle of old mice. Results of bulk mRNA-seq analysis and our contractile function data show that UnAG reversed neuromuscular junction impairment that occurs with age. Collectively, our data revealed the direct role of UnAG in mitigating sarcopenia in mice, independent of food consumption or body weight, implicating UnAG treatment as a potential therapy against sarcopenia.
Collapse
Affiliation(s)
- Hyunyoung Kim
- Department of Internal MedicineWake Forest University School of MedicineWinston‐SalemNorth CarolinaUSA
| | - Rojina Ranjit
- Aging and Metabolism Research ProgramOklahoma Medical Research FoundationOklahoma CityOklahomaUSA
- Department of BiochemistryUniversity of Oklahoma Health Sciences CenterOklahoma CityOklahomaUSA
| | - Dennis R. Claflin
- Department of Biomedical EngineeringUniversity of MichiganAnn ArborMichiganUSA
- Department of Molecular and Integrative PhysiologyUniversity of MichiganAnn ArborMichiganUSA
| | - Constantin Georgescu
- Genes and Human Disease Research ProgramOklahoma Medical Research FoundationOklahoma CityOklahomaUSA
| | - Jonathan D. Wren
- Genes and Human Disease Research ProgramOklahoma Medical Research FoundationOklahoma CityOklahomaUSA
| | - Susan V. Brooks
- Department of Biomedical EngineeringUniversity of MichiganAnn ArborMichiganUSA
- Department of Molecular and Integrative PhysiologyUniversity of MichiganAnn ArborMichiganUSA
| | - Benjamin F. Miller
- Aging and Metabolism Research ProgramOklahoma Medical Research FoundationOklahoma CityOklahomaUSA
- Oklahoma City VA Medical CenterOklahoma CityOklahomaUSA
| | - Bumsoo Ahn
- Department of Internal MedicineWake Forest University School of MedicineWinston‐SalemNorth CarolinaUSA
| |
Collapse
|
7
|
Nguyen HVM, Ran Q, Salmon AB, Bumsoo A, Chiao YA, Bhaskaran S, Richardson A. Mouse models used to test the role of reactive oxygen species in aging and age-related chronic diseases. Free Radic Biol Med 2024; 225:617-629. [PMID: 39419456 PMCID: PMC11624111 DOI: 10.1016/j.freeradbiomed.2024.10.269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 08/13/2024] [Accepted: 10/02/2024] [Indexed: 10/19/2024]
Abstract
With the development of the technology to generate transgenic and knockout mice in the 1990s, investigators had a powerful tool to directly test the impact of altering a specific gene on a biological process or disease. Over the past three decades, investigators have used transgenic and knockout mouse models, which have altered expression of antioxidant genes, to test the role of oxidative stress/damage in aging and age-related diseases. In this comprehensive review, we describe the studies using transgenic and knockout mouse models to test the role of oxidative stress/damage in aging (longevity) and three age-related diseases, e.g., sarcopenia, cardiac aging, and Alzheimer's Disease. While longevity was consistently altered only by one transgenic and one knockout mouse model as predicted by the Oxidative Stress Theory of Aging, the incidence/progression of the three age-related diseases (especially Alzheimer's disease) were robustly impacted when the expression of various antioxidant genes was altered using transgenic and knockout mouse models.
Collapse
Affiliation(s)
- Hoang Van M Nguyen
- Department of Nutritional Sciences, University of Oklahoma Health Sciences, Oklahoma City, OK, USA
| | - Qitao Ran
- Department of Cell Systems & Anatomy, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA; VA South Texas Health Care System, San Antonio, TX, USA
| | - Adam B Salmon
- Department of Molecular Medicine, Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA; VA South Texas Health Care System, San Antonio, TX, USA
| | - Ahn Bumsoo
- Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Ying Ann Chiao
- Aging & Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Shylesh Bhaskaran
- Aging & Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Arlan Richardson
- Department of Biochemistry & Molecular Biology, University of Oklahoma Health Sciences, Oklahoma City, OK, USA; VA Oklahoma Health Care System, Oklahoma City, OK, USA.
| |
Collapse
|
8
|
Bhaskaran S, Piekarz KM, Brown J, Yang B, Ocañas SR, Wren JD, Georgescu C, Bottoms C, Murphy A, Thomason J, Saunders D, Smith N, Towner R, Van Remmen H. The nitrone compound OKN-007 delays motor neuron loss and disease progression in the G93A mouse model of amyotrophic lateral sclerosis. Front Neurosci 2024; 18:1505369. [PMID: 39633896 PMCID: PMC11614777 DOI: 10.3389/fnins.2024.1505369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Accepted: 10/28/2024] [Indexed: 12/07/2024] Open
Abstract
Our study investigated the therapeutic potential of OKN-007 in the SOD1 G93A mouse model of amyotrophic lateral sclerosis (ALS). The impact of OKN-007, known for its antioxidant, anti-inflammatory, and neuroprotective properties, was tested at two doses (150 mg/kg and 300 mg/kg) at onset and late-stage disease. Results demonstrated a significant delay in disease progression at both doses, with treated mice showing a slower advance to early disease stages compared to untreated controls. Motor neuron counts in the lumbar spinal cord were notably higher in OKN-007 treated mice at the time of disease onset, suggesting neuroprotection. Additionally, OKN-007 reduced microglial activation and preserved reduced neuromuscular junction fragmentation, although it did not significantly alter the increase in astrocyte number or the decline in hindlimb muscle mass. MR spectroscopy (MRS) revealed improved spinal cord perfusion and normalized myo-inositol levels in treated mice, supporting reduced neuroinflammation. While the expression of several proteins associated with inflammation is increased in spinal cord extracts from G93A mice, OKN-007 dampened the expression of IL-1β, IL-1ra and IL-1α. Despite its promising effects on early-stage disease progression, in general, the beneficial effects of OKN-007 diminished over longer treatment durations. Further, we found no improvement in muscle atrophy or weakness phenotypes in OKN-007 treated G93A mice, and no effect on mitochondrial function or lifespan. Overall, our findings suggest that OKN-007 holds potential as a disease-modifying treatment for ALS, although further research is needed to optimize dosing regimens and understand its long-term effects.
Collapse
Affiliation(s)
- Shylesh Bhaskaran
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, United States
| | - Katarzyna M. Piekarz
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, United States
- Oklahoma Center for Neuroscience, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Jacob Brown
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, United States
| | - Brian Yang
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, United States
| | - Sarah R. Ocañas
- Oklahoma Center for Neuroscience, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
- Genes & Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, United States
| | - Jonathan D. Wren
- Genes & Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, United States
| | - Constantin Georgescu
- Genes & Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, United States
| | - Christopher Bottoms
- Center for Biomedical Data Sciences, Oklahoma Medical Research Foundation, Oklahoma City, OK, United States
| | - Ashley Murphy
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, United States
| | - Jessica Thomason
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, United States
| | - Debra Saunders
- Advanced Magnetic Resonance Center, Oklahoma Medical Research Foundation, Oklahoma City, OK, United States
| | - Nataliya Smith
- Advanced Magnetic Resonance Center, Oklahoma Medical Research Foundation, Oklahoma City, OK, United States
| | - Rheal Towner
- Advanced Magnetic Resonance Center, Oklahoma Medical Research Foundation, Oklahoma City, OK, United States
- Department of Chemistry, University of Prince Edward Island, Charlottetown, Prince Edward Island, Charlottetown, PE, Canada
| | - Holly Van Remmen
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, United States
- Oklahoma City VA Medical Center, Oklahoma City, OK, United States
| |
Collapse
|
9
|
Wang L, Guo D, Huang Y, Long P, Zhang X, Bai L, Liu J, Hu X, Pang R, Gou X. Scientific landscape of oxidative stress in sarcopenia: from bibliometric analysis to hotspots review. Front Med (Lausanne) 2024; 11:1472413. [PMID: 39588187 PMCID: PMC11586176 DOI: 10.3389/fmed.2024.1472413] [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: 07/29/2024] [Accepted: 10/28/2024] [Indexed: 11/27/2024] Open
Abstract
OBJECTIVE Sarcopenia is a significant healthcare challenge in the aging population. Oxidative stress (OS) is acknowledged to play a pivotal role in the pathological progression of sarcopenia. Numerous studies have demonstrated that mitigating or eliminating OS can ameliorate the pathological manifestations associated with sarcopenia. However, current clinical antioxidant therapies often fall short of anticipated outcomes. This bibliometric analysis aims to delineate prevailing research trends, thematic emphases, focal points, and developmental trajectories within the domain of OS in sarcopenia, while also endeavoring to explore prospective anti-oxidative stress strategies for future clinical interventions. METHODS Relevant publications were retrieved from the Web of Science (WOS) Core Collection database for the period 2000-2024. Citespace was employed for retrieving and analyzing trends and emerging topics. RESULTS In the field of OS in sarcopenia, the number of publications has significantly increased from 2000 to 2024. The United States and China are the primary contributors to global publication output. The most productive research institution is INRAE. The most prolific author is Holly Van Remmen from the United States, while the most frequently cited author is Cruz-Jentoft AJ from Spain. Experimental Gerontology is the journal with the highest volume of published articles, whereas the Journal of Gerontology Series A: Biological Sciences and Medical Sciences holds the record for the highest number of citations. The research keywords in this field can be categorized into eight domains: "Physiology and anatomy", "Physiological mechanisms", "Pathology associations", "Experimental studies", "Nutrition and metabolism", "Sports and physical activities", "Age" and "Oxidation and antioxidation". Moreover, recent years have seen the emergence of "TNF-α," "insulin resistance", "mitochondrial autophagy", "signal pathways", and "mechanisms" as focal points in the realm of OS in sarcopenia, encompassing related fundamental research and clinical translation. CONCLUSION This bibliometric and visualization provides a comprehensive analysis of the global research landscape in the field of OS in sarcopenia, identifies priorities, summarizes the current research status and suggests possible future research priorities. In addition, in order to benefit more sarcopenia patients, strengthening cooperation and communication between institutions and research teams is the key to the future development of this field. Given the expectation that research on OS in sarcopenia will remain a prominent area of interest in the future, this article could serve as a valuable resource for scholars seeking to shape future studies through an understanding of influential scholarly contributions and key research findings. SYSTEMATIC REVIEW REGISTRATION https://www.crd.york.ac.uk, identifier CRD42024528628.
Collapse
Affiliation(s)
- Linjie Wang
- Department of Rehabilitation Medicine, The General Hospital of Western Theater Command, Sichuan, Chengdu, China
- Sichuan Clinical Medical Research Center for Traditional Chinese Medicine Orthopedics and Sports Medicine Rehabilitation, Sichuan, Chengdu, China
| | - Dongliang Guo
- Department of Rehabilitation Medicine, The General Hospital of Western Theater Command, Sichuan, Chengdu, China
- Sichuan Clinical Medical Research Center for Traditional Chinese Medicine Orthopedics and Sports Medicine Rehabilitation, Sichuan, Chengdu, China
| | - Yi Huang
- Department of Rehabilitation Medicine, The General Hospital of Western Theater Command, Sichuan, Chengdu, China
- Sichuan Clinical Medical Research Center for Traditional Chinese Medicine Orthopedics and Sports Medicine Rehabilitation, Sichuan, Chengdu, China
| | - Pan Long
- Sichuan Clinical Medical Research Center for Traditional Chinese Medicine Orthopedics and Sports Medicine Rehabilitation, Sichuan, Chengdu, China
- Department of Ophthalmology, The General Hospital of Western Theater Command, Sichuan, Chengdu, China
| | - Xin Zhang
- Department of Rehabilitation Medicine, The General Hospital of Western Theater Command, Sichuan, Chengdu, China
- Sichuan Clinical Medical Research Center for Traditional Chinese Medicine Orthopedics and Sports Medicine Rehabilitation, Sichuan, Chengdu, China
| | - Ling Bai
- Department of Rehabilitation Medicine, The General Hospital of Western Theater Command, Sichuan, Chengdu, China
- Sichuan Clinical Medical Research Center for Traditional Chinese Medicine Orthopedics and Sports Medicine Rehabilitation, Sichuan, Chengdu, China
| | - Jiancheng Liu
- Department of Rehabilitation Medicine, The General Hospital of Western Theater Command, Sichuan, Chengdu, China
- Sichuan Clinical Medical Research Center for Traditional Chinese Medicine Orthopedics and Sports Medicine Rehabilitation, Sichuan, Chengdu, China
| | - Xiaomin Hu
- Department of Rehabilitation Medicine, The General Hospital of Western Theater Command, Sichuan, Chengdu, China
- Sichuan Clinical Medical Research Center for Traditional Chinese Medicine Orthopedics and Sports Medicine Rehabilitation, Sichuan, Chengdu, China
| | - Rizhao Pang
- Department of Rehabilitation Medicine, The General Hospital of Western Theater Command, Sichuan, Chengdu, China
- Sichuan Clinical Medical Research Center for Traditional Chinese Medicine Orthopedics and Sports Medicine Rehabilitation, Sichuan, Chengdu, China
| | - Xiang Gou
- Department of Rehabilitation Medicine, The General Hospital of Western Theater Command, Sichuan, Chengdu, China
- Sichuan Clinical Medical Research Center for Traditional Chinese Medicine Orthopedics and Sports Medicine Rehabilitation, Sichuan, Chengdu, China
| |
Collapse
|
10
|
Xu H, Piekarz KM, Brown JL, Bhaskaran S, Smith N, Towner RA, Van Remmen H. Neuroprotective treatment with the nitrone compound OKN-007 mitigates age-related muscle weakness in aging mice. GeroScience 2024; 46:4263-4273. [PMID: 38512579 PMCID: PMC11336152 DOI: 10.1007/s11357-024-01134-y] [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: 11/27/2023] [Accepted: 03/12/2024] [Indexed: 03/23/2024] Open
Abstract
Despite the universal impact of sarcopenia on compromised health and quality of life in the elderly, promising pharmaceutical approaches that can effectively mitigate loss of muscle and function during aging have been limited. Our group and others have reported impairments in peripheral motor neurons and loss of muscle innervation as initiating factors in sarcopenia, contributing to mitochondrial dysfunction and elevated oxidative stress in muscle. We recently reported a reduction in α motor neuron loss in aging mice in response to the compound OKN-007, a proposed antioxidant and anti-inflammatory agent. In the current study, we asked whether OKN-007 treatment in wildtype male mice for 8-9 months beginning at 16 months of age can also protect muscle mass and function. At 25 months of age, we observed a reduction in the loss of whole-body lean mass, a reduced loss of innervation at the neuromuscular junction and well-preserved neuromuscular junction morphology in OKN-007 treated mice versus age matched wildtype untreated mice. The loss in muscle force generation in aging mice (~ 25%) is significantly improved with OKN-007 treatment. In contrast, OKN-007 treatment provided no protection in loss of muscle mass in aging mice. Mitochondrial function was improved by OKN-007 treatment, consistent with its potential antioxidative properties. Together, these exciting findings are the first to demonstrate that interventions through neuroprotection can be an effective therapy to counter aging-related muscle dysfunction.
Collapse
Affiliation(s)
- Hongyang Xu
- Aging & Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Katarzyna M Piekarz
- Aging & Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Jacob L Brown
- Aging & Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
- Oklahoma City VA Medical Center, Oklahoma City, OK, USA
| | - Shylesh Bhaskaran
- Aging & Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Nataliya Smith
- Advanced Magnetic Resonance Center, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Rheal A Towner
- Advanced Magnetic Resonance Center, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Holly Van Remmen
- Aging & Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA.
- Department of Biochemistry & Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
- Oklahoma City VA Medical Center, Oklahoma City, OK, USA.
| |
Collapse
|
11
|
Yang Z, Wei J, Liu H, Zhang H, Liu R, Tang N, Yang X. Changes in muscle strength and risk of cardiovascular disease among middle-aged and older adults in China: Evidence from a prospective cohort study. Chin Med J (Engl) 2024; 137:1343-1350. [PMID: 38407330 PMCID: PMC11191030 DOI: 10.1097/cm9.0000000000002968] [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: 10/22/2023] [Indexed: 02/27/2024] Open
Abstract
BACKGROUND Evidence indicates that low muscle strength is associated with an increased cardiovascular diseases (CVDs) risk. However, the association between muscle strength changes based on repeated measurements and CVD incidence remains unclear. METHODS The study used data from the China Health and Retirement Longitudinal Study in 2011 (Wave 1), 2013 (Wave 2), 2015 (Wave 3), and 2018 (Wave 4). Low muscle strength was defined as handgrip strength <28 kg for men or <18 kg for women, or chair-rising time ≥12 s. Based on changes in muscle strength from Waves 1 to 2, participants were categorized into four groups of Normal-Normal, Low-Normal, Normal-Low, and Low-Low. CVD events, including heart disease and stroke, were recorded using a self-reported questionnaire during Waves 3 and 4 visits. Cox proportional hazards models were used to investigate the association between muscle strength changes and CVD incidence after multivariable adjustments. Hazard ratios (HRs) and 95% confidence intervals (95% CIs) were estimated with the Normal-Normal group as the reference. RESULTS A total of 1164 CVD cases were identified among 6608 participants. Compared to participants with sustained normal muscle strength, the CVD risks increased progressively across groups of the Low-Normal (HR = 1.20, 95% CI: 1.01-1.43), the Normal-Low (HR = 1.35, 95% CI: 1.14-1.60), and the Low-Low (HR = 1.76, 95% CI: 1.49-2.07). Similar patterns were observed for the significant associations between muscle strength status and the incidence risks of heart disease and stroke. Subgroup analyses showed that the significant associations between CVD and muscle strength changes were consistent across age, sex, and body mass index (BMI) categories. CONCLUSIONS The study found that muscle strength changes were associated with CVD risk. This suggests that continuous tracking of muscle status may be helpful in screening cardiovascular risk.
Collapse
Affiliation(s)
- Ze Yang
- Department of Occupational and Environmental Health, School of Public Health, Tianjin Medical University, Tianjin 300070, China
- Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin Medical University, Tianjin 300070, China
- Center for International Collaborative Research on Environment, Nutrition and Public Health, Tianjin 300070, China
| | - Jiemin Wei
- Department of Occupational and Environmental Health, School of Public Health, Tianjin Medical University, Tianjin 300070, China
- Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin Medical University, Tianjin 300070, China
- Center for International Collaborative Research on Environment, Nutrition and Public Health, Tianjin 300070, China
| | - Hongbo Liu
- Department of Occupational and Environmental Health, School of Public Health, Tianjin Medical University, Tianjin 300070, China
- Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin Medical University, Tianjin 300070, China
- Center for International Collaborative Research on Environment, Nutrition and Public Health, Tianjin 300070, China
| | - Honglu Zhang
- Department of Occupational and Environmental Health, School of Public Health, Tianjin Medical University, Tianjin 300070, China
- Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin Medical University, Tianjin 300070, China
- Center for International Collaborative Research on Environment, Nutrition and Public Health, Tianjin 300070, China
| | - Ruifang Liu
- Department of Occupational and Environmental Health, School of Public Health, Tianjin Medical University, Tianjin 300070, China
- Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin Medical University, Tianjin 300070, China
- Center for International Collaborative Research on Environment, Nutrition and Public Health, Tianjin 300070, China
| | - Naijun Tang
- Department of Occupational and Environmental Health, School of Public Health, Tianjin Medical University, Tianjin 300070, China
- Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin Medical University, Tianjin 300070, China
- Center for International Collaborative Research on Environment, Nutrition and Public Health, Tianjin 300070, China
| | - Xueli Yang
- Department of Occupational and Environmental Health, School of Public Health, Tianjin Medical University, Tianjin 300070, China
- Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin Medical University, Tianjin 300070, China
- Center for International Collaborative Research on Environment, Nutrition and Public Health, Tianjin 300070, China
| |
Collapse
|
12
|
Borowik AK, Lawrence MM, Peelor FF, Piekarz KM, Crosswhite A, Richardson A, Miller BF, Van Remmen H, Brown JL. Senolytic treatment does not mitigate oxidative stress-induced muscle atrophy but improves muscle force generation in CuZn superoxide dismutase knockout mice. GeroScience 2024; 46:3219-3233. [PMID: 38233728 PMCID: PMC11009189 DOI: 10.1007/s11357-024-01070-x] [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: 10/16/2023] [Accepted: 01/05/2024] [Indexed: 01/19/2024] Open
Abstract
Oxidative stress is associated with tissue dysfunctions that can lead to reduced health. Prior work has shown that oxidative stress contributes to both muscle atrophy and cellular senescence, which is a hallmark of aging that may drive in muscle atrophy and muscle contractile dysfunction. The purpose of the study was to test the hypothesis that cellular senescence contributes to muscle atrophy or weakness. To increase potential senescence in skeletal muscle, we used a model of oxidative stress-induced muscle frailty, the CuZn superoxide dismutase knockout (Sod1KO) mouse. We treated 6-month-old wildtype (WT) and Sod1KO mice with either vehicle or a senolytic treatment of combined dasatinib (5 mg/kg) + quercetin (50 mg/kg) (D + Q) for 3 consecutive days every 15 days. We continued treatment for 7 months and sacrificed the mice at 13 months of age. Treatment with D + Q did not preserve muscle mass, reduce NMJ fragmentation, or alter muscle protein synthesis in Sod1KO mice when compared to the vehicle-treated group. However, we observed an improvement in muscle-specific force generation in Sod1KO mice treated with D + Q when compared to Sod1KO-vehicle mice. Overall, these data suggest that reducing cellular senescence via D + Q is not sufficient to mitigate loss of muscle mass in a mouse model of oxidative stress-induced muscle frailty but may mitigate some aspects of oxidative stress-induced muscle dysfunction.
Collapse
Affiliation(s)
- Agnieszka K Borowik
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, USA
| | - Marcus M Lawrence
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, USA
- Department of Kinesiology and Outdoor Recreation, Southern Utah University, Cedar City, Utah, USA
| | - Frederick F Peelor
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, USA
| | - Katarzyna M Piekarz
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, USA
- Oklahoma Center for Neuroscience, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Abby Crosswhite
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, USA
| | - Arlan Richardson
- Oklahoma City VA Medical Center, Oklahoma City, OK, 73104, USA
- Department of Biochemistry & Molecular Biology, Oklahoma University Health Science Center, Oklahoma City, OK, 73104, USA
| | - Benjamin F Miller
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, USA
- Oklahoma City VA Medical Center, Oklahoma City, OK, 73104, USA
| | - Holly Van Remmen
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, USA
- Oklahoma City VA Medical Center, Oklahoma City, OK, 73104, USA
| | - Jacob L Brown
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, USA.
- Oklahoma City VA Medical Center, Oklahoma City, OK, 73104, USA.
| |
Collapse
|
13
|
Dowling P, Gargan S, Zweyer M, Henry M, Meleady P, Swandulla D, Ohlendieck K. Proteomic reference map for sarcopenia research: mass spectrometric identification of key muscle proteins of organelles, cellular signaling, bioenergetic metabolism and molecular chaperoning. Eur J Transl Myol 2024; 34:12565. [PMID: 38787292 PMCID: PMC11264233 DOI: 10.4081/ejtm.2024.12565] [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: 04/12/2024] [Accepted: 04/12/2024] [Indexed: 05/25/2024] Open
Abstract
During the natural aging process, frailty is often associated with abnormal muscular performance. Although inter-individual differences exit, in most elderly the tissue mass and physiological functionality of voluntary muscles drastically decreases. In order to study age-related contractile decline, animal model research is of central importance in the field of biogerontology. Here we have analyzed wild type mouse muscle to establish a proteomic map of crude tissue extracts. Proteomics is an advanced and large-scale biochemical method that attempts to identify all accessible proteins in a given biological sample. It is a technology-driven approach that uses mass spectrometry for the characterization of individual protein species. Total protein extracts were used in this study in order to minimize the potential introduction of artefacts due to excess subcellular fractionation procedures. In this report, the proteomic survey of aged muscles has focused on organellar marker proteins, as well as proteins that are involved in cellular signaling, the regulation of ion homeostasis, bioenergetic metabolism and molecular chaperoning. Hence, this study has establish a proteomic reference map of a highly suitable model system for future aging research.
Collapse
Affiliation(s)
- Paul Dowling
- Department of Biology, Maynooth University, National University of Ireland, Maynooth, Co. Kildare, Ireland; Kathleen Lonsdale Institute for Human Health Research, Maynooth University, Maynooth, Co. Kildare.
| | - Stephen Gargan
- Department of Biology, Maynooth University, National University of Ireland, Maynooth, Co. Kildare, Ireland; Kathleen Lonsdale Institute for Human Health Research, Maynooth University, Maynooth, Co. Kildare.
| | - Margit Zweyer
- Department of Neonatology and Paediatric Intensive Care, Children's Hospital, University of Bonn, Bonn, Germany; German Center for Neurodegenerative Diseases, Bonn.
| | - Michael Henry
- National Institute for Cellular Biotechnology, Dublin City University, Dublin.
| | - Paula Meleady
- National Institute for Cellular Biotechnology, Dublin City University, Dublin.
| | - Dieter Swandulla
- Institute of Physiology, Medical Faculty, University of Bonn, Bonn.
| | - Kay Ohlendieck
- Department of Biology, Maynooth University, National University of Ireland, Maynooth, Co. Kildare, Ireland; Kathleen Lonsdale Institute for Human Health Research, Maynooth University, Maynooth, Co. Kildare.
| |
Collapse
|
14
|
Kim K, Fazzone B, Cort TA, Kunz EM, Alvarez S, Moerschel J, Palzkill VR, Dong G, Anderson EM, O'Malley KA, Berceli SA, Ryan TE, Scali ST. Mitochondrial targeted catalase improves muscle strength following arteriovenous fistula creation in mice with chronic kidney disease. Sci Rep 2024; 14:8288. [PMID: 38594299 PMCID: PMC11004135 DOI: 10.1038/s41598-024-58805-1] [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: 11/21/2023] [Accepted: 04/03/2024] [Indexed: 04/11/2024] Open
Abstract
Hand dysfunction is a common observation after arteriovenous fistula (AVF) creation for hemodialysis access and has a variable clinical phenotype; however, the underlying mechanism responsible is unclear. Grip strength changes are a common metric used to assess AVF-associated hand disability but has previously been found to poorly correlate with the hemodynamic perturbations post-AVF placement implicating other tissue-level factors as drivers of hand outcomes. In this study, we sought to test if expression of a mitochondrial targeted catalase (mCAT) in skeletal muscle could reduce AVF-related limb dysfunction in mice with chronic kidney disease (CKD). Male and female C57BL/6J mice were fed an adenine-supplemented diet to induce CKD prior to placement of an AVF in the iliac vascular bundle. Adeno-associated virus was used to drive expression of either a green fluorescent protein (control) or mCAT using the muscle-specific human skeletal actin (HSA) gene promoter prior to AVF creation. As expected, the muscle-specific AAV-HSA-mCAT treatment did not impact blood urea nitrogen levels (P = 0.72), body weight (P = 0.84), or central hemodynamics including infrarenal aorta and inferior vena cava diameters (P > 0.18) or velocities (P > 0.38). Hindlimb perfusion recovery and muscle capillary densities were also unaffected by AAV-HSA-mCAT treatment. In contrast to muscle mass and myofiber size which were not different between groups, both absolute and specific muscle contractile forces measured via a nerve-mediated in-situ preparation were significantly greater in AAV-HSA-mCAT treated mice (P = 0.0012 and P = 0.0002). Morphological analysis of the post-synaptic neuromuscular junction uncovered greater acetylcholine receptor cluster areas (P = 0.0094) and lower fragmentation (P = 0.0010) in AAV-HSA-mCAT treated mice. Muscle mitochondrial oxidative phosphorylation was not different between groups, but AAV-HSA-mCAT treated mice had lower succinate-fueled mitochondrial hydrogen peroxide emission compared to AAV-HSA-GFP mice (P < 0.001). In summary, muscle-specific scavenging of mitochondrial hydrogen peroxide significantly improves neuromotor function in mice with CKD following AVF creation.
Collapse
Affiliation(s)
- Kyoungrae Kim
- Department of Applied Physiology and Kinesiology, University of Florida, 1864 Stadium Rd, Gainesville, FL, 32611, USA
| | - Brian Fazzone
- Division of Vascular Surgery and Endovascular Therapy, University of Florida, P.O. Box 100128, Gainesville, FL, 32610, USA
- Malcom Randall Veteran Affairs Medical Center, Gainesville, FL, USA
| | - Tomas A Cort
- Department of Applied Physiology and Kinesiology, University of Florida, 1864 Stadium Rd, Gainesville, FL, 32611, USA
| | - Eric M Kunz
- Department of Applied Physiology and Kinesiology, University of Florida, 1864 Stadium Rd, Gainesville, FL, 32611, USA
| | - Samuel Alvarez
- Department of Applied Physiology and Kinesiology, University of Florida, 1864 Stadium Rd, Gainesville, FL, 32611, USA
| | - Jack Moerschel
- Department of Applied Physiology and Kinesiology, University of Florida, 1864 Stadium Rd, Gainesville, FL, 32611, USA
| | - Victoria R Palzkill
- Department of Applied Physiology and Kinesiology, University of Florida, 1864 Stadium Rd, Gainesville, FL, 32611, USA
| | - Gengfu Dong
- Department of Applied Physiology and Kinesiology, University of Florida, 1864 Stadium Rd, Gainesville, FL, 32611, USA
| | - Erik M Anderson
- Division of Vascular Surgery and Endovascular Therapy, University of Florida, P.O. Box 100128, Gainesville, FL, 32610, USA
- Malcom Randall Veteran Affairs Medical Center, Gainesville, FL, USA
| | - Kerri A O'Malley
- Division of Vascular Surgery and Endovascular Therapy, University of Florida, P.O. Box 100128, Gainesville, FL, 32610, USA
- Malcom Randall Veteran Affairs Medical Center, Gainesville, FL, USA
| | - Scott A Berceli
- Division of Vascular Surgery and Endovascular Therapy, University of Florida, P.O. Box 100128, Gainesville, FL, 32610, USA
- Malcom Randall Veteran Affairs Medical Center, Gainesville, FL, USA
| | - Terence E Ryan
- Department of Applied Physiology and Kinesiology, University of Florida, 1864 Stadium Rd, Gainesville, FL, 32611, USA.
- Center for Exercise Science, University of Florida, Gainesville, FL, USA.
| | - Salvatore T Scali
- Division of Vascular Surgery and Endovascular Therapy, University of Florida, P.O. Box 100128, Gainesville, FL, 32610, USA.
- Malcom Randall Veteran Affairs Medical Center, Gainesville, FL, USA.
| |
Collapse
|
15
|
Germani S, Van Ho AT, Cherubini A, Varone E, Chernorudskiy A, Renna GM, Fumagalli S, Gobbi M, Lucchetti J, Bolis M, Guarrera L, Craparotta I, Rastelli G, Piccoli G, de Napoli C, Nogara L, Poggio E, Brini M, Cattaneo A, Bachi A, Simmen T, Calì T, Quijano-Roy S, Boncompagni S, Blaauw B, Ferreiro A, Zito E. SEPN1-related myopathy depends on the oxidoreductase ERO1A and is druggable with the chemical chaperone TUDCA. Cell Rep Med 2024; 5:101439. [PMID: 38402623 PMCID: PMC10982971 DOI: 10.1016/j.xcrm.2024.101439] [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: 05/09/2023] [Revised: 12/06/2023] [Accepted: 01/31/2024] [Indexed: 02/27/2024]
Abstract
Selenoprotein N (SEPN1) is a protein of the endoplasmic reticulum (ER) whose inherited defects originate SEPN1-related myopathy (SEPN1-RM). Here, we identify an interaction between SEPN1 and the ER-stress-induced oxidoreductase ERO1A. SEPN1 and ERO1A, both enriched in mitochondria-associated membranes (MAMs), are involved in the redox regulation of proteins. ERO1A depletion in SEPN1 knockout cells restores ER redox, re-equilibrates short-range MAMs, and rescues mitochondrial bioenergetics. ERO1A knockout in a mouse background of SEPN1 loss blunts ER stress and improves multiple MAM functions, including Ca2+ levels and bioenergetics, thus reversing diaphragmatic weakness. The treatment of SEPN1 knockout mice with the ER stress inhibitor tauroursodeoxycholic acid (TUDCA) mirrors the results of ERO1A loss. Importantly, muscle biopsies from patients with SEPN1-RM exhibit ERO1A overexpression, and TUDCA-treated SEPN1-RM patient-derived primary myoblasts show improvement in bioenergetics. These findings point to ERO1A as a biomarker and a viable target for intervention and to TUDCA as a pharmacological treatment for SEPN1-RM.
Collapse
Affiliation(s)
- Serena Germani
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy; Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Andrew Tri Van Ho
- Basic and Translational Myology Laboratory, Université Paris, BFA, UMR 8251, CNRS, 75013 Paris, France
| | | | - Ersilia Varone
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | | | | | | | - Marco Gobbi
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Jacopo Lucchetti
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Marco Bolis
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy; Bioinformatics Core Unit, Institute of Oncology Research (IOR), 6500 Bellinzona, Switzerland
| | - Luca Guarrera
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | | | - Giorgia Rastelli
- CAST, Center for Advanced Studies and Technology & DNICS, Department of Neuroscience, Imaging and Clinical Sciences, University G. D'Annunzio of Chieti-Pescara, 66100 Chieti, Italy
| | - Giorgia Piccoli
- Department of Biomedical Sciences, University of Padua, Padua, Italy
| | - Cosimo de Napoli
- Department of Biomedical Sciences, University of Padua, Padua, Italy
| | - Leonardo Nogara
- Department of Biomedical Sciences, University of Padua, Padua, Italy; Department of Pharmaceutical Sciences, University of Padova, Padova, Italy
| | - Elena Poggio
- Department of Biology, University of Padova, Padova, Italy
| | - Marisa Brini
- Department of Pharmaceutical Sciences, University of Padova, Padova, Italy; Department of Biology, University of Padova, Padova, Italy; Study Center for Neurodegeneration (CESNE), University of Padova, Padova, Italy
| | | | - Angela Bachi
- IFOM-ETS AIRC Institute of Molecular Oncology, Milan, Italy
| | - Thomas Simmen
- Department of Cell Biology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Tito Calì
- Department of Biomedical Sciences, University of Padua, Padua, Italy; Study Center for Neurodegeneration (CESNE), University of Padova, Padova, Italy; Padova Neuroscience Center, University of Padova, Padova, Italy
| | - Susana Quijano-Roy
- APHP-Université Paris-Saclay, Reference Center for Neuromuscular Disorders Nord-Est-Ile de France, FILNEMUS, ERN-Euro-NMD, Creteil, France; Pediatric Neurology and ICU Department, DMU Santé Enfant Adolescent (SEA), Raymond Poincaré University Hospital, Garches, France
| | - Simona Boncompagni
- CAST, Center for Advanced Studies and Technology & DNICS, Department of Neuroscience, Imaging and Clinical Sciences, University G. D'Annunzio of Chieti-Pescara, 66100 Chieti, Italy
| | - Bert Blaauw
- Department of Biomedical Sciences, University of Padua, Padua, Italy; Venetian Institute of Molecular Medicine, Padova, Italy.
| | - Ana Ferreiro
- Basic and Translational Myology Laboratory, Université Paris, BFA, UMR 8251, CNRS, 75013 Paris, France; APHP, Reference Center for Neuromuscular Disorders Nord-Est-Ile de France, Neuromyology Department, Groupe Hospitalier Pitié-Salpêtrière, Paris, France.
| | - Ester Zito
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy; Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy.
| |
Collapse
|
16
|
Xu H, Czyżowska A, Van Remmen H, Brown JL. Modulation of sarcopenia phenotypes by glutathione peroxidase 4 overexpression in mice. J Physiol 2023; 601:5277-5293. [PMID: 37878529 PMCID: PMC10871152 DOI: 10.1113/jp285259] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 09/15/2023] [Indexed: 10/27/2023] Open
Abstract
Our laboratory previously showed lipid hydroperoxides and oxylipin levels are elevated in response to loss of skeletal muscle innervation and are associated with muscle pathologies. To elucidate the pathological impact of lipid hydroperoxides, we overexpressed glutathione peroxidase 4 (GPx4), an enzyme that targets reduction of lipid hydroperoxides in membranes, in adult CuZn superoxide dismutase knockout (Sod1KO) mice that show accelerated muscle atrophy associated with loss of innervation. The gastrocnemius muscle from Sod1KO mice shows reduced mitochondrial respiration and elevated oxidative stress (F2 -isoprostanes and hydroperoxides) compared to wild-type (WT) mice. Overexpression of GPx4 improved mitochondrial respiration and reduced hydroperoxide generation in Sod1KO mice, but did not attenuate the muscle loss that occurs in Sod1KO mice. In contrast, contractile force generation is reduced in EDL muscle in Sod1KO mice relative to WT mice, and overexpression of GPx4 restored force generation to WT levels in Sod1KO mice. GPx4 overexpression also prevented loss of muscle contractility at the single fibre level in fast-twitch fibres from Sod1KO mice. Muscle fibres from Sod1KO mice were less sensitive to both depolarization and calcium at the single fibre level and exhibited a reduced activation by S-glutathionylation. GPx4 overexpression in Sod1KO mice rescued the deficits in both membrane excitability and calcium sensitivity of fast-twitch muscle fibres. Overexpression of GPx4 also restored the sarco/endoplasmic reticulum Ca2+ -ATPase activity in Sod1KO gastrocnemius muscles. These data suggest that GPx4 plays an important role in preserving excitation-contraction coupling function and Ca2+ homeostasis, and in maintaining muscle and mitochondrial function in oxidative stress-induced sarcopenia. KEY POINTS: Knockout of CuZn superoxide dismutase (Sod1KO) induces elevated oxidative stress with accelerated muscle atrophy and weakness. Glutathione peroxidase 4 (GPx4) plays a fundamental role in the reduction of lipid hydroperoxides in membranes, and overexpression of GPx4 improves mitochondrial respiration and reduces hydroperoxide generation in Sod1KO mice. Muscle contractile function deficits in Sod1KO mice are alleviated by the overexpression of GPx4. GPx4 overexpression in Sod1KO mice rescues the impaired muscle membrane excitability of fast-twitch muscle fibres and improves their calcium sensitivity. Sarco/endoplasmic reticulum Ca2+ -ATPase activity in Sod1KO muscles is decreased, and it is restored by the overexpression of GPx4. Our results confirm that GPx4 plays an important role in preserving excitation-contraction coupling function and Ca2+ homeostasis, and maintaining muscle and mitochondrial function in oxidative stress-induced sarcopenia.
Collapse
Affiliation(s)
- Hongyang Xu
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Agnieszka Czyżowska
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Holly Van Remmen
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
- Oklahoma City VA Medical Center, Oklahoma City, OK, USA
| | - Jacob L Brown
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
- Oklahoma City VA Medical Center, Oklahoma City, OK, USA
| |
Collapse
|
17
|
Gökçe Y, Danisman B, Akcay G, Derin N, Yaraş N. L-Carnitine improves mechanical responses of cardiomyocytes and restores Ca 2+ homeostasis during aging. Histochem Cell Biol 2023; 160:341-347. [PMID: 37329457 DOI: 10.1007/s00418-023-02215-3] [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] [Accepted: 05/29/2023] [Indexed: 06/19/2023]
Abstract
L-Carnitine (β-hydroxy-γ-trimethylaminobutyric acid, LC) is a crucial molecule for the mitochondrial oxidation of fatty acids. It facilitates the transport of long-chain fatty acids into the mitochondrial matrix. The reduction in LC levels during the aging process has been linked to numerous cardiovascular disorders, including contractility dysfunction, and disrupted intracellular Ca2+ homeostasis. The aim of this study was to examine the effects of long-term (7 months) LC administration on cardiomyocyte contraction and intracellular Ca2+ transients ([Ca2+]i) in aging rats. Male albino Wistar rats were randomly assigned to either the control or LC-treated groups. LC (50 mg/kg body weight/day) was dissolved in distilled water and orally administered for a period of 7 months. The control group received distilled water alone. Subsequently, ventricular single cardiomyocytes were isolated, and the contractility and Ca2+ transients were recorded in aging (18 months) rats. This study demonstrates, for the first time, a novel inotropic effect of long-term LC treatment on rat ventricular cardiomyocyte contraction. LC increased cardiomyocyte cell shortening and resting sarcomere length. Furthermore, LC supplementation led to a reduction in resting [Ca2+]i level and an increase in the amplitude of [Ca2+]i transients, indicative of enhanced contraction. Consistent with these results, decay time of Ca2+ transients also decreased significantly in the LC-treated group. The long-term administration of LC may help restore the Ca2+ homeostasis altered during aging and could be used as a cardioprotective medication in cases where myocyte contractility is diminished.
Collapse
Affiliation(s)
- Yasin Gökçe
- Faculty of Medicine, Department of Biophysics, Harran University, Sanliurfa, Turkey.
| | - Betul Danisman
- Faculty of Medicine, Department of Biophysics, Ataturk University, Erzurum, Turkey
| | - Guven Akcay
- Faculty of Medicine, Department of Biophysics, Hitit University, Corum, Turkey
| | - Narin Derin
- Faculty of Medicine, Department of Biophysics, Akdeniz University, Antalya, Turkey
| | - Nazmi Yaraş
- Faculty of Medicine, Department of Biophysics, Akdeniz University, Antalya, Turkey
| |
Collapse
|
18
|
Dhureja M, Arthur R, Soni D, Upadhayay S, Temgire P, Kumar P. Calcium channelopathies in neurodegenerative disorder: an untold story of RyR and SERCA. Expert Opin Ther Targets 2023; 27:1159-1172. [PMID: 37971192 DOI: 10.1080/14728222.2023.2277863] [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: 08/26/2023] [Accepted: 10/27/2023] [Indexed: 11/19/2023]
Abstract
INTRODUCTION Recent neuroscience breakthroughs have shed light on the sophisticated relationship between calcium channelopathies and movement disorders, exposing a previously undiscovered tale focusing on the Ryanodine Receptor (RyR) and the Sarco/Endoplasmic Reticulum Calcium ATPase (SERCA). Calcium signaling mainly orchestrates neural communication, which regulates synaptic transmission and total network activity. It has been determined that RyR play a significant role in managing neuronal functions, most notably in releasing intracellular calcium from the endoplasmic reticulum. AREAS COVERED It highlights the involvement of calcium channels such as RyR and SERCA in physiological and pathophysiological conditions. EXPERT OPINION Links between RyR and SERCA activity dysregulation, aberrant calcium levels, motor and cognitive dysfunction have brought attention to the importance of RyR and SERCA modulation in neurodegenerative disorders. Understanding the obscure function of these proteins will open up new therapeutic possibilities to address the underlying causes of neurodegenerative diseases. The unreported RyR and SERCA narrative broadens the understanding of calcium channelopathies in movement disorders and calls for more research into cutting-edge therapeutic approaches.
Collapse
Affiliation(s)
- Maanvi Dhureja
- Department of Pharmacology, Central University of Punjab, Bathinda, India
| | - Richmond Arthur
- Department of Pharmacology, Central University of Punjab, Bathinda, India
| | - Divya Soni
- Department of Pharmacology, Central University of Punjab, Bathinda, India
| | - Shubham Upadhayay
- Department of Pharmacology, Central University of Punjab, Bathinda, India
| | - Pooja Temgire
- Department of Pharmacology, Central University of Punjab, Bathinda, India
| | - Puneet Kumar
- Department of Pharmacology, Central University of Punjab, Bathinda, India
| |
Collapse
|