1
|
Qin M, Zhu J, Xing L, Fan Y, Luo J, Sun J, Chen T, Zhang Y, Xi Q. Adipose-derived exosomes ameliorate skeletal muscle atrophy via miR-146a-5p/IGF-1R signaling. J Nanobiotechnology 2024; 22:754. [PMID: 39696303 DOI: 10.1186/s12951-024-02983-7] [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: 10/10/2023] [Accepted: 11/04/2024] [Indexed: 12/20/2024] Open
Abstract
The study of muscle disorders has gained popularity, with a particular emphasis on the relationship between adipose tissue and skeletal muscle. In our investigation, we discovered that the deletion of miR-146a-5p specifically in adipose tissue (aKO) led to a notable rise in mice's mass and adiposity. In contrast, it led to a decline in lean mass, ability to exercise, diameter of muscle fibers, and the levels of genes associated with differentiation. The co-culture experiment showed that the transfection of miR-146a-5p mimics to 3T3-L1 significantly suppressive cell growth and promotes myotube differentiation in C2C12 cells. Exosomes from white adipose tissue (WAT) of aKO mice (aKO-WAT-Exos) significantly promoted muscle atrophy and inhibited differentiation of C2C12 cells but were reversed by co-incubation with miR-146a-5p-mimics. The miR-146a-5p can specifically target IGF-1R to improve skeletal muscle wasting. In this process, the PI3K/AKT/mTOR pathway is activated or the FoxO3 pathway is inhibited to enhance the synthesis of skeletal muscle proteins. Significantly, miR-146a-5p serves a crucial function as a microRNA in the communication of the fat-muscle connection. It can be transported through the pathway of exosomes derived from adipose tissue, ultimately ameliorating skeletal muscle atrophy and modulating body mass index (BMI).
Collapse
Affiliation(s)
- Mengran Qin
- State Key Laboratory of Livestock and Poultry Breeding, Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, No. 483 Wushan Road, Guangzhou, 510642, China
- Tianjin Hospital, Tianjin University, Tianjin, 300211, China
- Tianjin Orthopedic Institute, Tianjin Key Laboratory of Orthopedic Biomechanics and Medical Engineering, Tianjin, 300050, China
| | - Jiahao Zhu
- State Key Laboratory of Livestock and Poultry Breeding, Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, No. 483 Wushan Road, Guangzhou, 510642, China
| | - Lipeng Xing
- State Key Laboratory of Livestock and Poultry Breeding, Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, No. 483 Wushan Road, Guangzhou, 510642, China
| | - Yaotian Fan
- State Key Laboratory of Livestock and Poultry Breeding, Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, No. 483 Wushan Road, Guangzhou, 510642, China
| | - Junyi Luo
- State Key Laboratory of Livestock and Poultry Breeding, Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, No. 483 Wushan Road, Guangzhou, 510642, China
| | - Jiajie Sun
- State Key Laboratory of Livestock and Poultry Breeding, Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, No. 483 Wushan Road, Guangzhou, 510642, China
| | - Ting Chen
- State Key Laboratory of Livestock and Poultry Breeding, Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, No. 483 Wushan Road, Guangzhou, 510642, China
| | - Yongliang Zhang
- State Key Laboratory of Livestock and Poultry Breeding, Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, No. 483 Wushan Road, Guangzhou, 510642, China
| | - Qianyun Xi
- State Key Laboratory of Livestock and Poultry Breeding, Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, No. 483 Wushan Road, Guangzhou, 510642, China.
| |
Collapse
|
2
|
Zhao Q, Huang Y, Fu N, Cui C, Peng X, Kang H, Xiao J, Ke G. Podocyte senescence: from molecular mechanisms to therapeutics. Ren Fail 2024; 46:2398712. [PMID: 39248407 PMCID: PMC11385655 DOI: 10.1080/0886022x.2024.2398712] [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: 12/21/2023] [Revised: 08/25/2024] [Accepted: 08/26/2024] [Indexed: 09/10/2024] Open
Abstract
As an important component of the glomerular filtration membrane, the state of the podocytes is closely related to kidney function, they are also key cells involved in aging and play a central role in the damage caused by renal aging. Therefore, understanding the aging process of podocytes will allow us to understand their susceptibility to injury and identify targeted protective mechanisms. In fact, the process of physiological aging itself can induce podocyte senescence. Pathological stresses, such as oxidative stress, mitochondrial damage, secretion of senescence-associated secretory phenotype, reduced autophagy, oncogene activation, altered transcription factors, DNA damage response, and other factors, play a crucial role in inducing premature senescence and accelerating aging. Senescence-associated-β-galactosidase (SA-β-gal) is a marker of aging, and β-hydroxybutyric acid treatment can reduce SA-β-gal activity to alleviate cellular senescence and damage. In addition, CCAAT/enhancer-binding protein-α, transforming growth factor-β signaling, glycogen synthase kinase-3β, cycle-dependent kinase, programmed cell death protein 1, and plasminogen activator inhibitor-1 are closely related to aging. The absence or elevation of these factors can affect aging through different mechanisms. Podocyte injury is not an independent process, and injured podocytes interact with the surrounding epithelial cells or other kidney cells to mediate the injury or loss of podocytes. In this review, we discuss the manifestations, molecular mechanisms, biomarkers, and therapeutic drugs for podocyte senescence. We included elamipretide, lithium, calorie restriction, rapamycin; and emerging treatment strategies, such as gene and immune therapies. More importantly, we summarize how podocyte interact with other kidney cells.
Collapse
Affiliation(s)
- Qian Zhao
- Department of Nephrology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yongzhang Huang
- Department of Nephrology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Ningying Fu
- Department of Nephrology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Caixia Cui
- Department of Nephrology, The First Affiliated Hospital of Bengbu Medical University, Bengbu, China
| | - Xuan Peng
- Department of Nephrology, Affiliated Hospital/Clinical Medical College of Chengdu University, Chengdu, China
| | - Haiyan Kang
- Department of Nephrology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jie Xiao
- Department of Nephrology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Guibao Ke
- Department of Nephrology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
3
|
Lay AC, Tran VDT, Nair V, Betin V, Hurcombe JA, Barrington AF, Pope RJ, Burdet F, Mehl F, Kryvokhyzha D, Ahmad A, Sinton MC, Lewis P, Wilson MC, Menon R, Otto E, Heesom KJ, Ibberson M, Looker HC, Nelson RG, Ju W, Kretzler M, Satchell SC, Gomez MF, Coward RJM. Profiling of insulin-resistant kidney models and human biopsies reveals common and cell-type-specific mechanisms underpinning Diabetic Kidney Disease. Nat Commun 2024; 15:10018. [PMID: 39562547 PMCID: PMC11576882 DOI: 10.1038/s41467-024-54089-1] [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: 05/15/2023] [Accepted: 11/01/2024] [Indexed: 11/21/2024] Open
Abstract
Diabetic kidney disease (DKD) is the leading cause of end stage kidney failure worldwide, of which cellular insulin resistance is a major driver. Here, we study key human kidney cell types implicated in DKD (podocytes, glomerular endothelial, mesangial and proximal tubular cells) in insulin sensitive and resistant conditions, and perform simultaneous transcriptomics and proteomics for integrated analysis. Our data is further compared with bulk- and single-cell transcriptomic kidney biopsy data from early- and advanced-stage DKD patient cohorts. We identify several consistent changes (individual genes, proteins, and molecular pathways) occurring across all insulin-resistant kidney cell types, together with cell-line-specific changes occurring in response to insulin resistance, which are replicated in DKD biopsies. This study provides a rich data resource to direct future studies in elucidating underlying kidney signalling pathways and potential therapeutic targets in DKD.
Collapse
Affiliation(s)
- Abigail C Lay
- Bristol Renal, Bristol Medical School, University of Bristol, Bristol, UK
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Van Du T Tran
- Vital-IT group, SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Viji Nair
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Virginie Betin
- Bristol Renal, Bristol Medical School, University of Bristol, Bristol, UK
| | | | | | - Robert Jp Pope
- Bristol Renal, Bristol Medical School, University of Bristol, Bristol, UK
| | - Frédéric Burdet
- Vital-IT group, SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Florence Mehl
- Vital-IT group, SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Dmytro Kryvokhyzha
- Department of Clinical Sciences, Lund University Diabetes Centre, Lund University, Malmö, Sweden
| | - Abrar Ahmad
- Department of Clinical Sciences, Lund University Diabetes Centre, Lund University, Malmö, Sweden
| | - Matthew C Sinton
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Philip Lewis
- Proteomics Facility, University of Bristol, Bristol, UK
| | | | - Rajasree Menon
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Edgar Otto
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Kate J Heesom
- Proteomics Facility, University of Bristol, Bristol, UK
| | - Mark Ibberson
- Vital-IT group, SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Helen C Looker
- Chronic Kidney Disease Section, National Institute of Diabetes and Digestive and Kidney Diseases, National Institute of Health, Phoenix, AZ, USA
| | - Robert G Nelson
- Chronic Kidney Disease Section, National Institute of Diabetes and Digestive and Kidney Diseases, National Institute of Health, Phoenix, AZ, USA
| | - Wenjun Ju
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Matthias Kretzler
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Simon C Satchell
- Bristol Renal, Bristol Medical School, University of Bristol, Bristol, UK
| | - Maria F Gomez
- Department of Clinical Sciences, Lund University Diabetes Centre, Lund University, Malmö, Sweden
| | - Richard J M Coward
- Bristol Renal, Bristol Medical School, University of Bristol, Bristol, UK.
| |
Collapse
|
4
|
Moustafa Mahmoud M, Abdel Hameed NQ, Adel Al Dreny Abd Al Latef B, Samir Kamar S, Ahmed Rashed L, Abdelhameed Gouda SA. High-intensity exercise alongside insulin alleviates muscle atrophy in type 1 diabetes mellitus concomitant with modulation of mitophagy-related proteins in skeletal muscle. Arch Physiol Biochem 2024:1-13. [PMID: 39382178 DOI: 10.1080/13813455.2024.2410791] [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: 04/26/2024] [Revised: 08/08/2024] [Accepted: 09/15/2024] [Indexed: 10/10/2024]
Abstract
Background: Diabetes patients' quality of life can be severely impacted by diabetic muscle atrophy.Aim: This study aimed to explore the impact of high-intensity exercise (HIE) alongside insulin treatment on muscle atrophy in a rat model of type 1 diabetes mellitus (T1DM).Methodology: Fifty rats were allocated into five groups; Group 1, control sedentary (CS), T1DM was elicited in the rest of the groups by giving them Streptozotocin (STZ) (60 mg/kg), where group 2 (DS) remained sedentary, while groups 3,4,5 were treated with insulin after induction of diabetes. Group 4 (DI+MIE) and 5 (DI+ HIE) underwent moderate and high-intensity exercise, respectively.Results: HIE for 14 days combined with insulin treatment significantly restored muscle strength and mass with a significant modification in the mitophagy-related proteins and fibroblast growth factor 21 (FGF 21) compared to other treated groups.Conclusion: This study concluded that there is a therapeutic role for HIE with insulin against T1DM-induced muscle atrophy.
Collapse
Affiliation(s)
| | | | | | - Samaa Samir Kamar
- Histology department, Faculty of Medicine- Cairo University, Cairo, Egypt
- Histology department, Armed Forces College of Medicine, Cairo, Egypt
| | - Laila Ahmed Rashed
- Department of Medical Biochemistry, Faculty of Medicine, Cairo University, Cairo, Egypt
| | | |
Collapse
|
5
|
Park S, Cha HN, Shin MG, Park S, Kim Y, Kim MS, Shin KH, Thoudam T, Lee EJ, Wolfe RR, Dan J, Koh JH, Kim IY, Choi I, Lee IK, Sung HK, Park SY. Inhibitory Regulation of FOXO1 in PPARδ Expression Drives Mitochondrial Dysfunction and Insulin Resistance. Diabetes 2024; 73:1084-1098. [PMID: 38656552 DOI: 10.2337/db23-0432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Accepted: 04/09/2024] [Indexed: 04/26/2024]
Abstract
Forkhead box O1 (FOXO1) regulates muscle growth, but the metabolic role of FOXO1 in skeletal muscle and its mechanisms remain unclear. To explore the metabolic role of FOXO1 in skeletal muscle, we generated skeletal muscle-specific Foxo1 inducible knockout (mFOXO1 iKO) mice and fed them a high-fat diet to induce obesity. We measured insulin sensitivity, fatty acid oxidation, mitochondrial function, and exercise capacity in obese mFOXO1 iKO mice and assessed the correlation between FOXO1 and mitochondria-related protein in the skeletal muscle of patients with diabetes. Obese mFOXO1 iKO mice exhibited improved mitochondrial respiratory capacity, which was followed by attenuated insulin resistance, enhanced fatty acid oxidation, and improved skeletal muscle exercise capacity. Transcriptional inhibition of FOXO1 in peroxisome proliferator-activated receptor δ (PPARδ) expression was confirmed in skeletal muscle, and deletion of PPARδ abolished the beneficial effects of FOXO1 deficiency. FOXO1 protein levels were higher in the skeletal muscle of patients with diabetes and negatively correlated with PPARδ and electron transport chain protein levels. These findings highlight FOXO1 as a new repressor in PPARδ gene expression in skeletal muscle and suggest that FOXO1 links insulin resistance and mitochondrial dysfunction in skeletal muscle via PPARδ. ARTICLE HIGHLIGHTS
Collapse
Affiliation(s)
- Soyoung Park
- Department of Physiology, College of Medicine, Yeungnam University, Daegu, Republic of Korea
- Senotherapy-Based Metabolic Diseases Control Research Center, College of Medicine, Yeungnam University, Daegu, Republic of Korea
| | - Hye-Na Cha
- Department of Physiology, College of Medicine, Yeungnam University, Daegu, Republic of Korea
- Senotherapy-Based Metabolic Diseases Control Research Center, College of Medicine, Yeungnam University, Daegu, Republic of Korea
| | - Min-Gyeong Shin
- Department of Physiology, College of Medicine, Yeungnam University, Daegu, Republic of Korea
| | - Sanghee Park
- Department of Molecular Medicine, College of Medicine, Gachon University, Incheon, Republic of Korea
| | - Yeongmin Kim
- Department of Health Sciences and Technology, Gachon Advanced Institute for Health Sciences and Technology, Gachon University, Incheon, Republic of Korea
| | - Min-Seob Kim
- Department of Fundamental Environment Research, Environmental Measurement and Analysis Center, National Institute of Environmental Research, Incheon, Republic of Korea
| | - Kyung-Hoon Shin
- Department of Marine Sciences and Convergent Technology, Hanyang University, Ansan, Republic of Korea
| | - Themis Thoudam
- Research Institute of Aging and Metabolism, Kyungpook National University, Daegu, Republic of Korea
| | - Eun Ju Lee
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, Republic of Korea
| | - Robert R Wolfe
- Department of Geriatrics, University of Arkansas for Medical Sciences, Little Rock, AR
| | - Jinmyoung Dan
- Department of Orthopedic Surgery, College of Medicine, CHA University, Gumi, Republic of Korea
| | - Jin-Ho Koh
- Department of Convergence Medicine, Wonju College of Medicine, Yonsei University, Wonju, Republic of Korea
| | - Il-Young Kim
- Department of Molecular Medicine, College of Medicine, Gachon University, Incheon, Republic of Korea
| | - Inho Choi
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, Republic of Korea
| | - In-Kyu Lee
- Research Institute of Aging and Metabolism, Kyungpook National University, Daegu, Republic of Korea
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Republic of Korea
| | - Hoon-Ki Sung
- The Hospital for Sick Children Research Institute & Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - So-Young Park
- Department of Physiology, College of Medicine, Yeungnam University, Daegu, Republic of Korea
- Senotherapy-Based Metabolic Diseases Control Research Center, College of Medicine, Yeungnam University, Daegu, Republic of Korea
| |
Collapse
|
6
|
Lee MC, Hsu YJ, Chen MT, Kuo YW, Lin JH, Hsu YC, Huang YY, Li CM, Tsai SY, Hsia KC, Ho HH, Huang CC. Efficacy of Lactococcus lactis subsp. lactis LY-66 and Lactobacillus plantarum PL-02 in Enhancing Explosive Strength and Endurance: A Randomized, Double-Blinded Clinical Trial. Nutrients 2024; 16:1921. [PMID: 38931275 PMCID: PMC11206817 DOI: 10.3390/nu16121921] [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: 05/23/2024] [Revised: 06/14/2024] [Accepted: 06/15/2024] [Indexed: 06/28/2024] Open
Abstract
Probiotics are posited to enhance exercise performance by influencing muscle protein synthesis, augmenting glycogen storage, and reducing inflammation. This double-blind study randomized 88 participants to receive a six-week intervention with either a placebo, Lactococcus lactis subsp. lactis LY-66, Lactobacillus plantarum PL-02, or a combination of both strains, combined with a structured exercise training program. We assessed changes in maximal oxygen consumption (VO2max), exercise performance, and gut microbiota composition before and after the intervention. Further analyses were conducted to evaluate the impact of probiotics on exercise-induced muscle damage (EIMD), muscle integrity, and inflammatory markers in the blood, 24 and 48 h post-intervention. The results demonstrated that all probiotic groups exhibited significant enhancements in exercise performance and attenuation of muscle strength decline post-exercise exhaustion (p < 0.05). Notably, PL-02 intake significantly increased muscle mass, whereas LY-66 and the combination therapy significantly reduced body fat percentage (p < 0.05). Analysis of intestinal microbiota revealed an increase in beneficial bacteria, especially a significant rise in Akkermansia muciniphila following supplementation with PL-02 and LY-66 (p < 0.05). Overall, the combination of exercise training and supplementation with PL-02, LY-66, and their combination improved muscle strength, explosiveness, and endurance performance, and had beneficial effects on body composition and gastrointestinal health, as evidenced by data obtained from non-athlete participants.
Collapse
Affiliation(s)
- Mon-Chien Lee
- Graduate Institute of Sports Science, National Taiwan Sport University, Taoyuan 333325, Taiwan; (M.-C.L.); (Y.-J.H.)
- Center for General Education, Taipei Medical University, Taipei 110301, Taiwan
| | - Yi-Ju Hsu
- Graduate Institute of Sports Science, National Taiwan Sport University, Taoyuan 333325, Taiwan; (M.-C.L.); (Y.-J.H.)
| | - Mu-Tsung Chen
- Committee on General Studies, Shih Chien University, Taipei City 104, Taiwan;
| | - Yi-Wei Kuo
- Functional R&D Department, Research and Design Center, Glac Biotech Co., Ltd., Tainan City 744, Taiwan; (Y.-W.K.); (J.-H.L.); (Y.-Y.H.); (C.-M.L.); (H.-H.H.)
| | - Jia-Hung Lin
- Functional R&D Department, Research and Design Center, Glac Biotech Co., Ltd., Tainan City 744, Taiwan; (Y.-W.K.); (J.-H.L.); (Y.-Y.H.); (C.-M.L.); (H.-H.H.)
| | - Yu-Chieh Hsu
- Research Product Department, Research and Design Center, Glac Biotech Co., Ltd., Tainan City 744, Taiwan; (Y.-C.H.); (S.-Y.T.); (K.-C.H.)
| | - Yen-Yu Huang
- Functional R&D Department, Research and Design Center, Glac Biotech Co., Ltd., Tainan City 744, Taiwan; (Y.-W.K.); (J.-H.L.); (Y.-Y.H.); (C.-M.L.); (H.-H.H.)
| | - Ching-Min Li
- Functional R&D Department, Research and Design Center, Glac Biotech Co., Ltd., Tainan City 744, Taiwan; (Y.-W.K.); (J.-H.L.); (Y.-Y.H.); (C.-M.L.); (H.-H.H.)
| | - Shin-Yu Tsai
- Research Product Department, Research and Design Center, Glac Biotech Co., Ltd., Tainan City 744, Taiwan; (Y.-C.H.); (S.-Y.T.); (K.-C.H.)
| | - Ko-Chiang Hsia
- Research Product Department, Research and Design Center, Glac Biotech Co., Ltd., Tainan City 744, Taiwan; (Y.-C.H.); (S.-Y.T.); (K.-C.H.)
| | - Hsieh-Hsun Ho
- Functional R&D Department, Research and Design Center, Glac Biotech Co., Ltd., Tainan City 744, Taiwan; (Y.-W.K.); (J.-H.L.); (Y.-Y.H.); (C.-M.L.); (H.-H.H.)
- Research Product Department, Research and Design Center, Glac Biotech Co., Ltd., Tainan City 744, Taiwan; (Y.-C.H.); (S.-Y.T.); (K.-C.H.)
| | - Chi-Chang Huang
- Graduate Institute of Sports Science, National Taiwan Sport University, Taoyuan 333325, Taiwan; (M.-C.L.); (Y.-J.H.)
- Tajen University, Pingtung 907101, Taiwan
| |
Collapse
|
7
|
Liu D, Wang S, Liu S, Wang Q, Che X, Wu G. Frontiers in sarcopenia: Advancements in diagnostics, molecular mechanisms, and therapeutic strategies. Mol Aspects Med 2024; 97:101270. [PMID: 38583268 DOI: 10.1016/j.mam.2024.101270] [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: 10/28/2023] [Revised: 03/05/2024] [Accepted: 03/19/2024] [Indexed: 04/09/2024]
Abstract
The onset of sarcopenia is intimately linked with aging, posing significant implications not only for individual patient quality of life but also for the broader societal healthcare framework. Early and accurate identification of sarcopenia and a comprehensive understanding of its mechanistic underpinnings and therapeutic targets paramount to addressing this condition effectively. This review endeavors to present a cohesive overview of recent advancements in sarcopenia research and diagnosis. We initially delve into the contemporary diagnostic criteria, specifically referencing the European Working Group on Sarcopenia in Older People (EWGSOP) 2 and Asian Working Group on Sarcopenia (AWGS) 2019 benchmarks. Additionally, we elucidate comprehensive assessment techniques for muscle strength, quantity, and physical performance, highlighting tools such as grip strength, chair stand test, dual-energy X-ray Absorptiometry (DEXA), bioelectrical impedance analysis (BIA), gait speed, and short physical performance battery (SPPB), while also discussing their inherent advantages and limitations. Such diagnostic advancements pave the way for early identification and unequivocal diagnosis of sarcopenia. Proceeding further, we provide a deep-dive into sarcopenia's pathogenesis, offering a thorough examination of associated signaling pathways like the Myostatin, AMP-activated protein kinase (AMPK), insulin/IGF-1 Signaling (IIS), and the nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) pathways. Each pathway's role in sarcopenia mediation is detailed, underscoring potential therapeutic target avenues. From a mechanistic perspective, the review also underscores the pivotal role of mitochondrial dysfunction in sarcopenia, emphasizing elements such as mitochondrial oxidative overload, mitochondrial biogenesis, and mitophagy, and highlighting their therapeutic significance. At last, we capture recent strides made in sarcopenia treatment, ranging from nutritional and exercise interventions to potential pharmacological and supplementation strategies. In sum, this review meticulously synthesizes the latest scientific developments in sarcopenia, aiming to enhance diagnostic precision in clinical practice and provide comprehensive insights into refined mechanistic targets and innovative therapeutic interventions, ultimately contributing to optimized patient care and advancements in the field.
Collapse
Affiliation(s)
- Dequan Liu
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, Liaoning, China
| | - Shijin Wang
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, Liaoning, China
| | - Shuang Liu
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, Liaoning, China
| | - Qifei Wang
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, Liaoning, China.
| | - Xiangyu Che
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, Liaoning, China.
| | - Guangzhen Wu
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, Liaoning, China.
| |
Collapse
|
8
|
Bao S, Yin T, Liu S. Ovarian aging: energy metabolism of oocytes. J Ovarian Res 2024; 17:118. [PMID: 38822408 PMCID: PMC11141068 DOI: 10.1186/s13048-024-01427-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: 12/13/2023] [Accepted: 04/30/2024] [Indexed: 06/03/2024] Open
Abstract
In women who are getting older, the quantity and quality of their follicles or oocytes and decline. This is characterized by decreased ovarian reserve function (DOR), fewer remaining oocytes, and lower quality oocytes. As more women choose to delay childbirth, the decline in fertility associated with age has become a significant concern for modern women. The decline in oocyte quality is a key indicator of ovarian aging. Many studies suggest that age-related changes in oocyte energy metabolism may impact oocyte quality. Changes in oocyte energy metabolism affect adenosine 5'-triphosphate (ATP) production, but how related products and proteins influence oocyte quality remains largely unknown. This review focuses on oocyte metabolism in age-related ovarian aging and its potential impact on oocyte quality, as well as therapeutic strategies that may partially influence oocyte metabolism. This research aims to enhance our understanding of age-related changes in oocyte energy metabolism, and the identification of biomarkers and treatment methods.
Collapse
Affiliation(s)
- Shenglan Bao
- Reproductive Medical Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Tailang Yin
- Reproductive Medical Center, Renmin Hospital of Wuhan University, Wuhan, China.
| | - Su Liu
- Shenzhen Key Laboratory of Reproductive Immunology for Peri-Implantation, , Shenzhen Zhongshan Institute for Reproductive Medicine and Genetics, Shenzhen Zhongshan Obstetrics & Gynecology Hospital (Formerly Shenzhen Zhongshan Urology Hospital), Shenzhen, China.
| |
Collapse
|
9
|
Hurcombe JA, Barrington F, Marchetti M, Betin VM, Bowen EE, Lay AC, Ni L, Dayalan L, Pope RJ, Brinkkoetter PT, Holzenberger M, Welsh GI, Coward RJ. Contrasting consequences of podocyte insulin-like growth factor 1 receptor inhibition. iScience 2024; 27:109749. [PMID: 38706850 PMCID: PMC11068853 DOI: 10.1016/j.isci.2024.109749] [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: 01/20/2023] [Revised: 04/12/2023] [Accepted: 04/11/2024] [Indexed: 05/07/2024] Open
Abstract
Insulin signaling to the glomerular podocyte via the insulin receptor (IR) is critical for kidney function. In this study we show that near-complete knockout of the closely related insulin-like growth factor 1 receptor (IGF1R) in podocytes is detrimental, resulting in albuminuria in vivo and podocyte cell death in vitro. In contrast, partial podocyte IGF1R knockdown confers protection against doxorubicin-induced podocyte injury. Proteomic analysis of cultured podocytes revealed that while near-complete loss of podocyte IGF1R results in the downregulation of mitochondrial respiratory complex I and DNA damage repair proteins, partial IGF1R inhibition promotes respiratory complex expression. This suggests that altered mitochondrial function and resistance to podocyte stress depends on the level of IGF1R suppression, the latter determining whether receptor inhibition is protective or detrimental. Our work suggests that the partial suppression of podocyte IGF1R could have therapeutic benefits in treating albuminuric kidney disease.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Lan Ni
- Bristol Renal, University of Bristol, Bristol, UK
| | | | | | - Paul T. Brinkkoetter
- Department II of Internal Medicine and Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | | | | | | |
Collapse
|
10
|
Umek N, Pušnik L, Ugwoke CK, Šink Ž, Horvat S, Janáček J, Cvetko E. Skeletal muscle myosin heavy chain expression and 3D capillary network changes in streptozotocin-induced diabetic female mice. BIOMOLECULES & BIOMEDICINE 2024; 24:582-592. [PMID: 37902457 PMCID: PMC11088899 DOI: 10.17305/bb.2023.9843] [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: 09/20/2023] [Revised: 10/18/2023] [Accepted: 10/30/2023] [Indexed: 10/31/2023]
Abstract
It is not well-understood how type 1 diabetes (T1DM) affects skeletal muscle histological phenotype, particularly capillarisation. This study aimed to analyze skeletal muscle myosin heavy chain (MyHC) fibre type changes and 3D capillary network characteristics in experimental T1DM mice. Female C57BL/6J-OlaHsd mice were categorized into streptozotocin (STZ)-induced diabetic (n = 12) and age-matched non-diabetic controls (n =12). The muscle fibre phenotype of the soleus, gluteus maximus, and gastrocnemius muscles were characterized based on the expression of MyHC isoforms, while capillaries of the gluteus maximus were assessed with immunofluorescence staining, confocal laser microscopy and 3D image analysis. STZ-induced diabetic mice exhibited elevated glucose levels, reduced body weight, and prolonged thermal latency, verifying the T1DM phenotype. In both T1DM and non-diabetic mice, the gluteus maximus and gastrocnemius muscles predominantly expressed fast-twitch type 2b fibers, with no significant differences noted. However, the soleus muscle in non-diabetic mice had a greater proportion of type 2a fibers and comparable type 1 fiber densities (26.2 ± 14.6% vs 21.9 ± 13.5%) relative to diabetic mice. T1DM mice showed reduced fiber diameters (P = 0.026), and the 3D capillary network analysis indicated a higher capillary length per muscle volume in the gluteus maximus of diabetic mice compared to controls (P < 0.05). Overall, T1DM induced significant changes in the skeletal muscle, including shifts in MyHC fibre types, decreased fibre diameters, and increased relative capillarisation, possibly due to muscle fibre atrophy. Our findings emphasize the superior detail provided by the 3D analytical method for characterizing skeletal muscle capillary architecture, highlighting caution in interpreting 2D data for capillary changes in T1DM.
Collapse
Affiliation(s)
- Nejc Umek
- Institute of Anatomy, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Luka Pušnik
- Institute of Anatomy, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | | | - Žiga Šink
- Institute of Anatomy, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Simon Horvat
- Department of Animal Science, Biotechnical Faculty, University of Ljubljana, Ljubljana, Slovenia
| | - Jiří Janáček
- Laboratory of Biomathematics, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Erika Cvetko
- Institute of Anatomy, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| |
Collapse
|
11
|
Kawahara T, Suzuki G, Mizuno S, Tominaga N, Toda M, Toyama N, Inazu T, Kawahara C, Okada Y, Tanaka Y. Active vitamin D treatment in the prevention of sarcopenia in adults with prediabetes (DPVD ancillary study): a randomised controlled trial. THE LANCET. HEALTHY LONGEVITY 2024; 5:e255-e263. [PMID: 38437855 DOI: 10.1016/s2666-7568(24)00009-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Revised: 01/15/2024] [Accepted: 01/18/2024] [Indexed: 03/06/2024] Open
Abstract
BACKGROUND Observational studies show inverse associations between serum 25-hydroxyvitamin D concentrations and sarcopenia incidence; however, it remains unclear whether treatment with vitamin D prevents its development. We aimed to assess whether treatment with active vitamin D (eldecalcitol [0·75 μg per day]) can reduce the development of sarcopenia among adults with prediabetes. METHODS This randomised, double-blind, placebo-controlled, multicenter trial as an ancillary study was conducted at 32 clinics and hospital sites in Japan. Participants were assigned (1:1) by using a central randomisation method in which a randomisation list was made for each hospital separately using a stratified permuted block procedure. The primary endpoint was sarcopenia incidence during 3 years in the intention-to-treat population defined as weak handgrip strength (<28 kg for men and <18 kg for women) and low appendicular skeletal muscle index (<7·0 kg/m2 for men and <5·7 kg/m2 for women in bioelectrical impedance analysis). Although the usual criterion of hypercalcaemia was 10·4 mg/dL (2·6 mmol/L) or higher, hypercalcaemia that was enough to discontinue the study was defined as 11·0 mg/dL or higher. This study is registered with the UMIN clinical trials registry, UMIN000005394. FINDINGS A total of 1094 participants (548 in the eldecalcitol group and 546 in the placebo group; 44·2% [484 of 1094] women; mean age 60·8 [SD 9·2] years) were followed up for a median of 2·9 (IQR 2·8-3·0) years. Eldecalcitol treatment as compared with placebo showed statistically significant preventive effect on sarcopenia incidence (25 [4·6%] of 548 participants in the eldecalcitol group and 48 [8·8%] of 546 participants in the placebo group; hazard ratio 0·51; 95% CI 0·31 to 0·83; p=0·0065). The incidence of adverse events did not differ between the two groups. INTERPRETATION We found that treatment with eldecalcitol has the potential to prevent the onset of sarcopenia among people with prediabetes via increasing skeletal muscle volume and strength, which might lead to a substantial risk reduction of falls. FUNDING Kitakyushu Medical Association. TRANSLATION For the Japanese translation of the abstract see Supplementary Materials section.
Collapse
Affiliation(s)
- Tetsuya Kawahara
- The First Department of Internal Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan; Department of Endocrinology and Diabetes, Shin Komonji Hospital, Kitakyushu, Japan.
| | - Gen Suzuki
- Department of Internal Medicine, International University Health and Welfare Clinic, Ohtawara, Japan
| | - Shoichi Mizuno
- Division of Biostatics, National Cancer Center EPOC, Kashiwa, Japan
| | - Naoki Tominaga
- Department of Endocrinology and Diabetes, Shin Komonji Hospital, Kitakyushu, Japan
| | - Mikio Toda
- Department of Endocrinology and Diabetes, Shin Komonji Hospital, Kitakyushu, Japan
| | - Nagahiro Toyama
- Department of Endocrinology and Diabetes, Shin Komonji Hospital, Kitakyushu, Japan
| | - Tetsuya Inazu
- Department of Pharmacy, College of Pharmaceutical Sciences, Ritsumeikan University, Kusatsu, Japan
| | - Chie Kawahara
- The First Department of Internal Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Yosuke Okada
- The First Department of Internal Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Yoshiya Tanaka
- The First Department of Internal Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| |
Collapse
|
12
|
Cai M, Li S, Cai K, Du X, Han J, Hu J. Empowering mitochondrial metabolism: Exploring L-lactate supplementation as a promising therapeutic approach for metabolic syndrome. Metabolism 2024; 152:155787. [PMID: 38215964 DOI: 10.1016/j.metabol.2024.155787] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 12/08/2023] [Accepted: 01/05/2024] [Indexed: 01/14/2024]
Abstract
Mitochondrial dysfunction plays a critical role in the pathogenesis of metabolic syndrome (MetS), affecting various cell types and organs. In MetS animal models, mitochondria exhibit decreased quality control, characterized by abnormal morphological structure, impaired metabolic activity, reduced energy production, disrupted signaling cascades, and oxidative stress. The aberrant changes in mitochondrial function exacerbate the progression of metabolic syndrome, setting in motion a pernicious cycle. From this perspective, reversing mitochondrial dysfunction is likely to become a novel and powerful approach for treating MetS. Unfortunately, there are currently no effective drugs available in clinical practice to improve mitochondrial function. Recently, L-lactate has garnered significant attention as a valuable metabolite due to its ability to regulate mitochondrial metabolic processes and function. It is highly likely that treating MetS and its related complications can be achieved by correcting mitochondrial homeostasis disorders. In this review, we comprehensively discuss the complex relationship between mitochondrial function and MetS and the involvement of L-lactate in regulating mitochondrial metabolism and associated signaling pathways. Furthermore, it highlights recent findings on the involvement of L-lactate in common pathologies of MetS and explores its potential clinical application and further prospects, thus providing new insights into treatment possibilities for MetS.
Collapse
Affiliation(s)
- Ming Cai
- College of Rehabilitation Sciences, Shanghai University of Medicine and Health Sciences, Shanghai 201318, PR China; Bio-X Institutes, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Shuyao Li
- College of Rehabilitation Sciences, Shanghai University of Medicine and Health Sciences, Shanghai 201318, PR China
| | - Keren Cai
- College of Rehabilitation Sciences, Shanghai University of Medicine and Health Sciences, Shanghai 201318, PR China
| | - Xinlin Du
- College of Rehabilitation Sciences, Shanghai University of Medicine and Health Sciences, Shanghai 201318, PR China
| | - Jia Han
- College of Rehabilitation Sciences, Shanghai University of Medicine and Health Sciences, Shanghai 201318, PR China.
| | - Jingyun Hu
- Central Lab, Shanghai Key Laboratory of Pathogenic Fungi Medical Testing, Shanghai Pudong New Area People's Hospital, Shanghai 201299, PR China.
| |
Collapse
|
13
|
Wu H, Xu T, Yang N, Zhang J, Xu S. Low-Se Diet Increased Mitochondrial ROS to Suppress Myoblasts Proliferation and Promote Apoptosis in Broilers via miR-365-3p/SelT Signaling Axis. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:284-299. [PMID: 38109331 DOI: 10.1021/acs.jafc.3c04406] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2023]
Abstract
microRNA (miRNA) controls the post-transcriptional translation of mRNA to affect the expression of many genes participating in functional interaction pathways. Selenoproteins are characterized by their antioxidant activity, wherein selenoprotein T (SelT) is an essential membrane-bound selenoprotein serving as a guardian of intracellular homeostasis. During muscle development and regeneration, myoblasts enter the cell cycle and rapidly proliferate. However, the role of SelT in muscle development and selenium (Se) deficiency-induced muscle damage remains poorly investigated. This study established Se deficient broiler models, chicken embryos models, and cultured chicken primary myoblasts in vitro. We showed that Se deficiency induced skeletal muscle damage in broilers, promoted miR-365-3p expression, and downregulated the level of SelT, significantly. The absence of SelT led to the accumulation of mitochondrial superoxide and downregulated mitochondrial dynamics gene expression, which, in turn, induced the disruption of mitochondria potential and blocked the oxidative phosphorylation (OXPHOS) process. Limited ATP production rate caused by mitochondrial ROS overproduction went along with cell cycle arrest, cell proliferation slowness, and myocyte apoptosis increase. Using Mito-TEMPO for mitochondrial ROS elimination could effectively mitigate the above adverse reactions and significantly restore the proliferation potential of myoblasts. Moreover, we identified miR-365-3p, a miRNA that targeted SelT mRNA to inhibit myoblast proliferation by disrupting intracellular redox balance. The omics analysis results showed that Se deficiency led to the significant enrichment of "cell cycle", "oxidative stress response", and "oxidative phosphorylation" pathway genes. Finally, we proved that the effect of the miR-365-3p/SelT signaling axis on muscle development did exist in the chicken embryo stage. In summary, our findings revealed that miR-365-3p was involved in broiler skeletal muscle damage in Se deficiency by targeting SelT, and SelT, serving as an intracellular homeostasis guardian, resisted mitochondrial oxidative stress, and protected ATP generation, promoting myoblast proliferation and inhibiting apoptosis. This study provides an attractive target for the cultivated meat industry and regenerative medicine.
Collapse
Affiliation(s)
- Hao Wu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, P. R. China
| | - Tong Xu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, P. R. China
| | - Naixi Yang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, P. R. China
| | - Jiuli Zhang
- Heilongjiang Polytechnic, Harbin 150080, P. R. China
| | - Shiwen Xu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, P. R. China
- Key Laboratory of the Provincial Education Department of Heilongjiang for Common Animal Disease Prevention and Treatment, College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, P. R. China
| |
Collapse
|
14
|
Li X, Hu S, Cai Y, Liu X, Luo J, Wu T. Revving the engine: PKB/AKT as a key regulator of cellular glucose metabolism. Front Physiol 2024; 14:1320964. [PMID: 38264327 PMCID: PMC10804622 DOI: 10.3389/fphys.2023.1320964] [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: 10/13/2023] [Accepted: 12/12/2023] [Indexed: 01/25/2024] Open
Abstract
Glucose metabolism is of critical importance for cell growth and proliferation, the disorders of which have been widely implicated in cancer progression. Glucose uptake is achieved differently by normal cells and cancer cells. Even in an aerobic environment, cancer cells tend to undergo metabolism through glycolysis rather than the oxidative phosphorylation pathway. Disordered metabolic syndrome is characterized by elevated levels of metabolites that can cause changes in the tumor microenvironment, thereby promoting tumor recurrence and metastasis. The activation of glycolysis-related proteins and transcription factors is involved in the regulation of cellular glucose metabolism. Changes in glucose metabolism activity are closely related to activation of protein kinase B (PKB/AKT). This review discusses recent findings on the regulation of glucose metabolism by AKT in tumors. Furthermore, the review summarizes the potential importance of AKT in the regulation of each process throughout glucose metabolism to provide a theoretical basis for AKT as a target for cancers.
Collapse
Affiliation(s)
- Xia Li
- General Practice Medical Center, West China Hospital, Sichuan University, Chengdu, China
| | - Shuying Hu
- General Practice Medical Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yaoting Cai
- General Practice Medical Center, West China Hospital, Sichuan University, Chengdu, China
| | - Xuelian Liu
- General Practice Medical Center, West China Hospital, Sichuan University, Chengdu, China
| | - Jing Luo
- General Practice Medical Center, West China Hospital, Sichuan University, Chengdu, China
| | - Tao Wu
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
15
|
Zhang Q, Ye X, Xu X, Yan J. Placenta-derived exosomal miR-135a-5p promotes gestational diabetes mellitus pathogenesis by activating PI3K/AKT signalling pathway via SIRT1. J Cell Mol Med 2023; 27:3729-3743. [PMID: 37667545 PMCID: PMC10718144 DOI: 10.1111/jcmm.17941] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 08/17/2023] [Accepted: 08/24/2023] [Indexed: 09/06/2023] Open
Abstract
Most people are aware of gestational diabetes mellitus (GDM), a dangerous pregnancy complication in which pregnant women who have never been diagnosed with diabetes develop chronic hyperglycaemia. Exosomal microRNA (miRNA) dysregulation has been shown to be a key player in the pathophysiology of GDM. In this study, we looked into how placental exosomes and their miRNAs may contribute to GDM. When compared to exosomes from healthy pregnant women, it was discovered that miR-135a-5p was elevated in placenta-derived exosomes that were isolated from the maternal peripheral plasma of GDM women. Additionally, we discovered that miR-135a-5p encouraged HTR-8/SVneo cell growth, invasion and migration. Further research revealed that miR-135a-5p activates HTR-8/SVneo cells' proliferation, invasion and migration by promoting PI3K/AKT pathway activity via Sirtuin 1 (SIRT1). The transfer of exosomal miR-135a-5p generated from the placenta could be viewed as a promising agent for targeting genes and pertinent pathways involved in GDM, according to our findings.
Collapse
Affiliation(s)
- Qiuyu Zhang
- Department of Obstetrics and Gynecology, Fujian Maternity and Child Health HospitalAffiliated Hospital of Fujian Medical UniversityFuzhouChina
| | - Xu Ye
- Department of Obstetrics and Gynecology, Fujian Maternity and Child Health HospitalAffiliated Hospital of Fujian Medical UniversityFuzhouChina
| | - Xia Xu
- Department of Obstetrics and Gynecology, Fujian Maternity and Child Health HospitalAffiliated Hospital of Fujian Medical UniversityFuzhouChina
| | - Jianying Yan
- Department of Obstetrics and Gynecology, Fujian Maternity and Child Health HospitalAffiliated Hospital of Fujian Medical UniversityFuzhouChina
| |
Collapse
|
16
|
Li J, Zhou X, Chen J, Eliasson P, Kingham PJ, Backman LJ. Secretome from myoblasts statically loaded at low intensity promotes tenocyte proliferation via the IGF-1 receptor pathway. FASEB J 2023; 37:e23203. [PMID: 37732638 DOI: 10.1096/fj.202301097r] [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: 06/02/2023] [Revised: 08/16/2023] [Accepted: 09/05/2023] [Indexed: 09/22/2023]
Abstract
Exercise is widely recognized as beneficial for tendon healing. Recently, it has been described that muscle-derived molecules secreted in response to static exercise influence tendon healing. In this study, the optimal static loading intensity for tendon healing and the composition of secretome released by myoblasts in response to different intensities of static strain were investigated. In an in vitro coculture model, myoblasts were mechanically loaded using a Flexcell Tension System. Tenocytes were seeded on transwell inserts that allowed communication between the tenocytes and myoblasts without direct contact. Proliferation and migration assays, together with RNA sequencing, were used to determine potential cellular signaling pathways. The secretome from myoblasts exposed to 2% static loading increased the proliferation and migration of the cocultured tenocytes. RNA-seq analysis revealed that this loading condition upregulated the expression of numerous genes encoding secretory proteins, including insulin-like growth factor-1 (IGF-1). Confirmation of IGF-1 expression and secretion was carried out using qPCR and enzyme-linked immunosorbt assay (ELISA), revealing a statistically significant upregulation in response to 2% static loading in comparison to both control conditions and higher loading intensities of 5% and 10%. Addition of an inhibitor of the IGF-1 receptor (PQ401) to the tenocytes significantly reduced myoblast secretome-induced tenocyte proliferation. In conclusion, IGF-1 may be an important molecule in the statically loaded myoblast secretome, which is responsible for influencing tenocytes during exercise-induced healing.
Collapse
Affiliation(s)
- Junhong Li
- Department of Integrative Medical Biology, Umeå University, Umeå, Sweden
- Department of Community Medicine and Rehabilitation, Physiotherapy, Umeå University, Umeå, Sweden
| | - Xin Zhou
- Department of Community Medicine and Rehabilitation, Physiotherapy, Umeå University, Umeå, Sweden
| | - Jialin Chen
- School of Medicine, Southeast University, Nanjing, China
| | - Pernilla Eliasson
- Department of Orthopaedics, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Paul J Kingham
- Department of Integrative Medical Biology, Umeå University, Umeå, Sweden
| | - Ludvig J Backman
- Department of Integrative Medical Biology, Umeå University, Umeå, Sweden
- Department of Community Medicine and Rehabilitation, Physiotherapy, Umeå University, Umeå, Sweden
| |
Collapse
|
17
|
Chen ZL, Guo C, Zou YY, Feng C, Yang DX, Sun CC, Wen W, Jian ZJ, Zhao Z, Xiao Q, Zheng L, Peng XY, Zhou ZQ, Tang CF. Aerobic exercise enhances mitochondrial homeostasis to counteract D-galactose-induced sarcopenia in zebrafish. Exp Gerontol 2023; 180:112265. [PMID: 37482108 DOI: 10.1016/j.exger.2023.112265] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Revised: 07/18/2023] [Accepted: 07/20/2023] [Indexed: 07/25/2023]
Abstract
Sarcopenia is a common skeletal muscle degenerative disease characterized by decreased skeletal muscle mass and mitochondrial dysfunction that involves microRNAs (miR) as regulatory factors in various pathways. Exercise reduces age-related oxidative damage and chronic inflammation and increases autophagy, among others. Moreover, whether aerobic exercise can regulate mitochondrial homeostasis by modulating the miR-128/insulin-like growth factor-1 (IGF-1) signaling pathway and can improve sarcopenia requires further investigation. Interestingly, zebrafish have been used as a model for aging research for over a decade due to their many outstanding advantages. Therefore, we established a model of zebrafish sarcopenia using d-galactose immersion and observed substantial changes, including reduced skeletal muscle cross-sectional area, increased tissue fibrosis, decreased motility, increased skeletal muscle reactive oxygen species, and notable alterations in mitochondrial morphology and function. We found that miR-128 expression was considerably upregulated, where as Igf1 and peroxisome proliferator-activated receptor gamma coactivator 1-alpha were significantly downregulated; moreover, mitochondrial homeostasis was reduced. Four weeks of aerobic exercise delayed sarcopenia progression and prevented the disruption of mitochondrial function and homeostasis. The genes related to atrophy and miR-128 were downregulated, Igf1 expression was considerably upregulated, and the phosphorylation levels of Pi3k, Akt, and Foxo3a were upregulated. Furthermore, mitochondrial respiration and homeostasis were enhanced. In conclusion, aerobic exercise improved skeletal muscle quality and function via the miR-128/IGF-1 signaling pathway, consequently ameliorating mitochondrial homeostasis in aging skeletal muscle.
Collapse
Affiliation(s)
- Zhang-Lin Chen
- Key Laboratory of Physical Fitness and Exercise Rehabilitation of Hunan Province, College of Physical Education, Hunan Normal University, 410012 Changsha, China
| | - Cheng Guo
- Key Laboratory of Physical Fitness and Exercise Rehabilitation of Hunan Province, College of Physical Education, Hunan Normal University, 410012 Changsha, China
| | - Yun-Yi Zou
- Key Laboratory of Physical Fitness and Exercise Rehabilitation of Hunan Province, College of Physical Education, Hunan Normal University, 410012 Changsha, China
| | - Chen Feng
- Key Laboratory of Physical Fitness and Exercise Rehabilitation of Hunan Province, College of Physical Education, Hunan Normal University, 410012 Changsha, China
| | - Di-Xuan Yang
- Key Laboratory of Physical Fitness and Exercise Rehabilitation of Hunan Province, College of Physical Education, Hunan Normal University, 410012 Changsha, China
| | - Chen-Chen Sun
- Key Laboratory of Physical Fitness and Exercise Rehabilitation of Hunan Province, College of Physical Education, Hunan Normal University, 410012 Changsha, China
| | - Wei Wen
- Key Laboratory of Physical Fitness and Exercise Rehabilitation of Hunan Province, College of Physical Education, Hunan Normal University, 410012 Changsha, China
| | - Zhen-Jie Jian
- Key Laboratory of Physical Fitness and Exercise Rehabilitation of Hunan Province, College of Physical Education, Hunan Normal University, 410012 Changsha, China
| | - Zhe Zhao
- Key Laboratory of Physical Fitness and Exercise Rehabilitation of Hunan Province, College of Physical Education, Hunan Normal University, 410012 Changsha, China
| | - Qin Xiao
- Key Laboratory of Physical Fitness and Exercise Rehabilitation of Hunan Province, College of Physical Education, Hunan Normal University, 410012 Changsha, China
| | - Lan Zheng
- Key Laboratory of Physical Fitness and Exercise Rehabilitation of Hunan Province, College of Physical Education, Hunan Normal University, 410012 Changsha, China
| | - Xi-Yang Peng
- Key Laboratory of Physical Fitness and Exercise Rehabilitation of Hunan Province, College of Physical Education, Hunan Normal University, 410012 Changsha, China.
| | - Zuo-Qiong Zhou
- Key Laboratory of Physical Fitness and Exercise Rehabilitation of Hunan Province, College of Physical Education, Hunan Normal University, 410012 Changsha, China.
| | - Chang-Fa Tang
- Key Laboratory of Physical Fitness and Exercise Rehabilitation of Hunan Province, College of Physical Education, Hunan Normal University, 410012 Changsha, China.
| |
Collapse
|
18
|
Chen X, Ji Y, Liu R, Zhu X, Wang K, Yang X, Liu B, Gao Z, Huang Y, Shen Y, Liu H, Sun H. Mitochondrial dysfunction: roles in skeletal muscle atrophy. J Transl Med 2023; 21:503. [PMID: 37495991 PMCID: PMC10373380 DOI: 10.1186/s12967-023-04369-z] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 07/18/2023] [Indexed: 07/28/2023] Open
Abstract
Mitochondria play important roles in maintaining cellular homeostasis and skeletal muscle health, and damage to mitochondria can lead to a series of pathophysiological changes. Mitochondrial dysfunction can lead to skeletal muscle atrophy, and its molecular mechanism leading to skeletal muscle atrophy is complex. Understanding the pathogenesis of mitochondrial dysfunction is useful for the prevention and treatment of skeletal muscle atrophy, and finding drugs and methods to target and modulate mitochondrial function are urgent tasks in the prevention and treatment of skeletal muscle atrophy. In this review, we first discussed the roles of normal mitochondria in skeletal muscle. Importantly, we described the effect of mitochondrial dysfunction on skeletal muscle atrophy and the molecular mechanisms involved. Furthermore, the regulatory roles of different signaling pathways (AMPK-SIRT1-PGC-1α, IGF-1-PI3K-Akt-mTOR, FoxOs, JAK-STAT3, TGF-β-Smad2/3 and NF-κB pathways, etc.) and the roles of mitochondrial factors were investigated in mitochondrial dysfunction. Next, we analyzed the manifestations of mitochondrial dysfunction in muscle atrophy caused by different diseases. Finally, we summarized the preventive and therapeutic effects of targeted regulation of mitochondrial function on skeletal muscle atrophy, including drug therapy, exercise and diet, gene therapy, stem cell therapy and physical therapy. This review is of great significance for the holistic understanding of the important role of mitochondria in skeletal muscle, which is helpful for researchers to further understanding the molecular regulatory mechanism of skeletal muscle atrophy, and has an important inspiring role for the development of therapeutic strategies for muscle atrophy targeting mitochondria in the future.
Collapse
Affiliation(s)
- Xin Chen
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Department of Neurology, Affiliated Hospital of Nantong University, Nantong University, Nantong, 226001, Jiangsu, People's Republic of China
| | - Yanan Ji
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Department of Neurology, Affiliated Hospital of Nantong University, Nantong University, Nantong, 226001, Jiangsu, People's Republic of China
| | - Ruiqi Liu
- Department of Clinical Medicine, Medical College, Nantong University, Nantong, Jiangsu, 226001, People's Republic of China
| | - Xucheng Zhu
- Department of Clinical Medicine, Medical College, Nantong University, Nantong, Jiangsu, 226001, People's Republic of China
| | - Kexin Wang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Department of Neurology, Affiliated Hospital of Nantong University, Nantong University, Nantong, 226001, Jiangsu, People's Republic of China
| | - Xiaoming Yang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Department of Neurology, Affiliated Hospital of Nantong University, Nantong University, Nantong, 226001, Jiangsu, People's Republic of China
| | - Boya Liu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Department of Neurology, Affiliated Hospital of Nantong University, Nantong University, Nantong, 226001, Jiangsu, People's Republic of China
| | - Zihui Gao
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Department of Neurology, Affiliated Hospital of Nantong University, Nantong University, Nantong, 226001, Jiangsu, People's Republic of China
| | - Yan Huang
- Department of Clinical Medicine, Medical College, Nantong University, Nantong, Jiangsu, 226001, People's Republic of China
| | - Yuntian Shen
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Department of Neurology, Affiliated Hospital of Nantong University, Nantong University, Nantong, 226001, Jiangsu, People's Republic of China.
| | - Hua Liu
- Department of Orthopedics, Haian Hospital of Traditional Chinese Medicine, 55 Ninghai Middle Road, Nantong, Jiangsu, 226600, People's Republic of China.
| | - Hualin Sun
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Department of Neurology, Affiliated Hospital of Nantong University, Nantong University, Nantong, 226001, Jiangsu, People's Republic of China.
| |
Collapse
|
19
|
Yu J, Li T, Zhu J. Gene Therapy Strategies Targeting Aging-Related Diseases. Aging Dis 2023; 14:398-417. [PMID: 37008065 PMCID: PMC10017145 DOI: 10.14336/ad.2022.00725] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 07/25/2022] [Indexed: 11/18/2022] Open
Abstract
Rapid advancements have taken place in gene therapy technology. However, effective methods for treating aging- or age-related chronic diseases, which are often closely related to genes or even multiple genes, are still lacking. The path to developing cures is winding, while gene therapy that targets genes related to aging represents an exciting research direction with tremendous potential. Among aging-related genes, some candidates have been studied at different levels, from cell to organismal levels (e.g., mammalian models) with different methods, from overexpression to gene editing. The TERT and APOE have even entered the stage of clinical trials. Even those displaying only a preliminary association with diseases have potential applications. This article discusses the foundations and recent breakthroughs in the field of gene therapy, providing a summary of current mainstream strategies and gene therapy products with clinical and preclinical applications. Finally, we review representative target genes and their potential for treating aging or age-related diseases.
Collapse
Affiliation(s)
| | | | - Jianhong Zhu
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, National Center for Neurological Disorders, National Key Laboratory for Medical Neurobiology, Institutes of Brain Science, Shanghai Key Laboratory of Brain Function and Regeneration, Institute of Neurosurgery, MOE Frontiers Center for Brain Science, Shanghai, China.
| |
Collapse
|
20
|
Ahmadi A, Huda MN, Bennett BJ, Gamboa J, Zelnick LR, Smith LR, Chondronikola M, Raftery D, de Boer IH, Roshanravan B. Chronic Kidney Disease is Associated With Attenuated Plasma Metabolome Response to Oral Glucose Tolerance Testing. J Ren Nutr 2023; 33:316-325. [PMID: 36270479 PMCID: PMC11196097 DOI: 10.1053/j.jrn.2022.09.013] [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: 05/02/2022] [Revised: 08/01/2022] [Accepted: 09/25/2022] [Indexed: 12/15/2022] Open
Abstract
OBJECTIVE Chronic kidney disease (CKD) is associated with decreased anabolic response to insulin contributing to protein-energy wasting. Targeted metabolic profiling of oral glucose tolerance testing (OGTT) may help identify metabolic pathways contributing to disruptions to insulin response in CKD. METHODS Using targeted metabolic profiling, we studied the plasma metabolome response in 41 moderate-to-severe nondiabetic CKD patients and 20 healthy controls at fasting and 2 hours after an oral glucose load. We used linear mixed modeling with random intercepts, adjusting for age, gender, race/ethnicity, body weight, and batch to assess heterogeneity in response to OGTT by CKD status. RESULTS Mean estimated glomerular filtration rate among CKD participants was 38.9 ± 12.7 mL/min per 1.73 m2 compared to 87.2 ± 17.7 mL/min per 1.73 m2 among controls. Glucose ingestion induced an anabolic response resulting in increased glycolysis products and a reduction in a wide range of metabolites including amino acids, tricarboxylic acid cycle intermediates, and purine nucleotides compared to fasting. Participants with CKD demonstrated a blunted anabolic response to OGTT evidenced by significant changes in 13 metabolites compared to controls. The attenuated metabolome response predominant involved mitochondrial energy metabolism, vitamin B family, and purine nucleotides. Compared to controls, CKD participants had elevated lactate:pyruvate (L:P) ratio and decreased guanosine diphosphate:guanosine triphosphate ratio during OGTT. CONCLUSION Metabolic profiling of OGTT response suggests a broad disruption of mitochondrial energy metabolism in CKD patients. These findings motivate further investigation into the impact of insulin sensitizers and mitochondrial targeted therapeutics on energy metabolism in patients with nondiabetic CKD.
Collapse
Affiliation(s)
- Armin Ahmadi
- Department of Medicine, Division of Nephrology, University of California Davis, Davis, California
| | - M Nazmul Huda
- Obesity and Metabolism Research Unit, Western Human Nutrition Research Center, USDA, ARS, Davis, California; Department of Nutrition, University of California Davis, Davis, California
| | - Brian J Bennett
- Obesity and Metabolism Research Unit, Western Human Nutrition Research Center, USDA, ARS, Davis, California; Department of Nutrition, University of California Davis, Davis, California
| | - Jorge Gamboa
- Division of Clinical Pharmacology, Department of Medicine, School of Medicine, Vanderbilt University, Nashville, Tennessee
| | - Leila R Zelnick
- Division of Nephrology and Kidney Research Institute, University of Washington, Seattle, Washington
| | - Lucas R Smith
- Department of Physical Medicine and Rehabilitation, School of Medicine, UCD, Davis, California
| | | | - Daniel Raftery
- Anesthesiology and Pain Medicine, University of Washington, Seattle, Washington
| | - Ian H de Boer
- Division of Nephrology and Kidney Research Institute, University of Washington, Seattle, Washington; Puget Sound Health Care System, Seattle, Washington
| | - Baback Roshanravan
- Department of Medicine, Division of Nephrology, University of California Davis, Davis, California.
| |
Collapse
|
21
|
Penniman CM, Bhardwaj G, Nowers CJ, Brown CU, Junck TL, Boyer CK, Jena J, Fuqua JD, Lira VA, O'Neill BT. Loss of FoxOs in muscle increases strength and mitochondrial function during aging. J Cachexia Sarcopenia Muscle 2023; 14:243-259. [PMID: 36442857 PMCID: PMC9891940 DOI: 10.1002/jcsm.13124] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 10/13/2022] [Accepted: 10/25/2022] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND Muscle mitochondrial decline is associated with aging-related muscle weakness and insulin resistance. FoxO transcription factors are targets of insulin action and deletion of FoxOs improves mitochondrial function in diabetes. However, disruptions in proteostasis and autophagy are hallmarks of aging and the effect of chronic inhibition of FoxOs in aged muscle is unknown. This study investigated the role of FoxOs in regulating muscle strength and mitochondrial function with age. METHODS We measured muscle strength, cross-sectional area, muscle fibre-type, markers of protein synthesis/degradation, central nuclei, glucose/insulin tolerance, and mitochondrial bioenergetics in 4.5-month (Young) and 22-24-month-old (Aged) muscle-specific FoxO1/3/4 triple KO (TKO) and littermate control (Ctrl) mice. RESULTS Lean mass was increased in Aged TKO compared with both Aged Ctrl and younger groups by 26-33% (P < 0.01). Muscle strength, measured by max force of tibialis anterior (TA) contraction, was 20% lower in Aged Ctrl compared with Young Ctrls (P < 0.01) but was not decreased in Aged TKOs. Increased muscle strength in Young and Aged TKO was associated with 18-48% increased muscle weights compared with Ctrls (P < 0.01). Muscle cross-sectional analysis of TA, soleus, and plantaris revealed increases in fibre size distribution and a 2.5-10-fold increase in central nuclei in Young and Aged TKO mice, without histologic signs of muscle damage. Age-dependent increases in Gadd45a and Ube4a expression as well accumulation of K48 polyubiquitinated proteins were observed in quad and TA but were prevented by FoxO deletion. Young and Aged TKO muscle showed minimal changes in autophagy flux and no accumulation of autophagosomes compared with Ctrl groups. Increased strength in Young and Aged TKO was associated with a 10-20% increase in muscle mitochondrial respiration using glutamate/malate/succinate compared with controls (P < 0.05). OXPHOS subunit expression and complex I activity were decreased 16-34% in Aged Ctrl compared with Young Ctrl but were prevented in Aged TKO. Both Aged Ctrl and Aged TKO showed impaired glucose tolerance by 33% compared to young groups (P < 0.05) indicating improved strength and mitochondrial respiration are not due to improved glycemia. CONCLUSIONS FoxO deletion increases muscle strength even during aging. Deletion of FoxOs maintains muscle strength in part by mild suppression of atrophic pathways, including inhibition of Gadd45a and Ube4a expression, without accumulation of autophagosomes in muscle. Deletion of FoxOs also improved mitochondrial function by maintenance of OXPHOS in both young and aged TKO.
Collapse
Affiliation(s)
- Christie M Penniman
- Fraternal Order of Eagles Diabetes Research Center and Division of Endocrinology and Metabolism, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, 52242, Iowa City, Iowa, USA
| | - Gourav Bhardwaj
- Fraternal Order of Eagles Diabetes Research Center and Division of Endocrinology and Metabolism, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, 52242, Iowa City, Iowa, USA
| | - Colette J Nowers
- Fraternal Order of Eagles Diabetes Research Center and Division of Endocrinology and Metabolism, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, 52242, Iowa City, Iowa, USA
| | - Chandler U Brown
- Fraternal Order of Eagles Diabetes Research Center and Division of Endocrinology and Metabolism, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, 52242, Iowa City, Iowa, USA
| | - Taylor L Junck
- Fraternal Order of Eagles Diabetes Research Center and Division of Endocrinology and Metabolism, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, 52242, Iowa City, Iowa, USA
| | - Cierra K Boyer
- Fraternal Order of Eagles Diabetes Research Center and Division of Endocrinology and Metabolism, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, 52242, Iowa City, Iowa, USA
| | - Jayashree Jena
- Fraternal Order of Eagles Diabetes Research Center and Division of Endocrinology and Metabolism, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, 52242, Iowa City, Iowa, USA
| | - Jordan D Fuqua
- Fraternal Order of Eagles Diabetes Research Center and Department of Health and Human Physiology, College of Liberal Arts and Sciences, University of Iowa, Iowa City, Iowa, USA
| | - Vitor A Lira
- Fraternal Order of Eagles Diabetes Research Center and Department of Health and Human Physiology, College of Liberal Arts and Sciences, University of Iowa, Iowa City, Iowa, USA
| | - Brian T O'Neill
- Fraternal Order of Eagles Diabetes Research Center and Division of Endocrinology and Metabolism, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, 52242, Iowa City, Iowa, USA.,Veterans Affairs Health Care System, 52242, Iowa City, Iowa, USA
| |
Collapse
|
22
|
He Q, Gao L, Zhang F, Yao W, Wu J, Song N, Luo J, Zhang Y. The FoxO1-ATGL axis alters milk lipolysis homeostasis through PI3K/AKT signaling pathway in dairy goat mammary epithelial cells. J Anim Sci 2023; 101:skad286. [PMID: 37638641 PMCID: PMC10699848 DOI: 10.1093/jas/skad286] [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: 04/10/2023] [Accepted: 08/25/2023] [Indexed: 08/29/2023] Open
Abstract
Goat milk is enriched in fatty acids which are beneficial to human health. Previous research has revealed that 98% of milk fat is composed of triglycerides. However, the mechanisms regulating milk fat composition remain unclear. Forkhead box protein O1 (FoxO1) is a crucial regulatory factor involved in lipid metabolism across various cell types. Chromatin immunoprecipitation sequencing (ChIP)-seq data) and RNA sequencing (RNA-seq) data revealed that have indicated a close association between FoxO1 was closely related to lipid metabolism during lactation in dairy goats. The objective of this study was to investigate the mechanisms by which FoxO1 regulates lipid metabolism in goat mammary epithelial cells (GMECs). FoxO1 knockdown significantly downregulated the expression of adipose triglyceride lipase (ATGL) and suppressed the activity of the ATGL promoter. Consistently, the number of lipid droplets decreased significantly in FoxO1-overexpressing cells and increased in ATGL-knockdown cells. To further verify the effect of FoxO1 on ATGL promoter activity, cells were transfected with four promoter fragments of different lengths. We found that the core region of the ATGL promoter was located between -882 bp and -524 bp, encompassing two FoxO1 binding sites (FKH1 and FKH2). Mutations in the FoxO1 binding sites significantly downregulated ATGL promoter activity in GMECs. Luciferase reporter assays demonstrated that FoxO1 overexpression markedly enhanced ATGL promoter activity. Furthermore, site-directed mutation confirmed that FKH1 and FKH2 sites were simultaneously mutated significantly attenuated the stimulatory effect of FoxO1 on ATGL promoter activities simultaneous mutation of FKH1 and FKH2 sites significantly attenuated the stimulatory effect of FoxO1 on ATGL promoter activity. ChIP assays showed that FoxO1 directly binds to the FKH2 element located in the ATGL promoter in vivo. Finally, immunofluorescence staining revealed that insulin promotes the translocation of FoxO1 from the nucleus to the cytoplasm, thereby attenuating the FoxO1-induced activation of the ATGL promoter. Collectively, these findings uncover a novel pathway where by FoxO1 may regulate lipid metabolism in GMECs specifically by modulating the transcriptional activity of ATGL.
Collapse
Affiliation(s)
- Qiuya He
- Shaanxi Key Laboratory of Molecular Biology for Agriculture, College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China
| | - Liangjiahui Gao
- Shaanxi Key Laboratory of Molecular Biology for Agriculture, College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China
| | - Fuhong Zhang
- Shaanxi Key Laboratory of Molecular Biology for Agriculture, College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China
| | - Weiwei Yao
- Shaanxi Key Laboratory of Molecular Biology for Agriculture, College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China
| | - Jiao Wu
- Shaanxi Key Laboratory of Molecular Biology for Agriculture, College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China
| | - Ning Song
- College of Animal Science and Technology, Anhui Province Key Laboratory of Local Livestock and Poultry Genetic Resource Conservation and Bio-breeding, Anhui Agricultural University, Hefei 230036, China
| | - Jun Luo
- Shaanxi Key Laboratory of Molecular Biology for Agriculture, College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China
| | - Yong Zhang
- Shaanxi Key Laboratory of Molecular Biology for Agriculture, College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China
| |
Collapse
|
23
|
Spinal Canal and Spinal Cord in Rat Continue to Grow Even after Sexual Maturation: Anatomical Study and Molecular Proposition. Int J Mol Sci 2022; 23:ijms232416076. [PMID: 36555713 PMCID: PMC9781254 DOI: 10.3390/ijms232416076] [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: 11/06/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022] Open
Abstract
Although rodents have been widely used for experimental models of spinal cord diseases, the details of the growth curves of their spinal canal and spinal cord, as well as the molecular mechanism of the growth of adult rat spinal cords remain unavailable. They are particularly important when conducting the experiments of cervical spondylotic myelopathy (CSM), since the disease condition depends on the size of the spinal canal and the spinal cord. Thus, the purposes of the present study were to obtain accurate growth curves for the spinal canal and spinal cord in rats; to define the appropriate age in weeks for their use as a CSM model; and to propose a molecular mechanism of the growth of the adult spinal cord in rats. CT myelography was performed on Lewis rats from 4 weeks to 40 weeks of age. The vertical growth of the spinal canal at C5 reached a plateau after 20 and 12 weeks, and at T8 after 20 and 16 weeks, in males and females, respectively. The vertical growth of the C5 and T8 spinal cord reached a plateau after 24 weeks in both sexes. The vertical space available for the cord (SAC) of C5 and T8 did not significantly change after 8 weeks in either sex. Western blot analyses showed that VEGFA, FGF2, and BDNF were highly expressed in the cervical spinal cords of 4-week-old rats, and that the expression of these growth factors declined as rats grew. These findings indicate that the spinal canal and the spinal cord in rats continue to grow even after sexual maturation and that rats need to be at least 8 weeks of age for use in experimental models of CSM. The present study, in conjunction with recent evidence, proposes the hypothetical model that the growth of rat spinal cord after the postnatal period is mediated at least in part by differentiation of neural progenitor cells and that their differentiation potency is maintained by VEGFA, FGF2, and BDNF.
Collapse
|
24
|
Chen K, Gao P, Li Z, Dai A, Yang M, Chen S, Su J, Deng Z, Li L. Forkhead Box O Signaling Pathway in Skeletal Muscle Atrophy. THE AMERICAN JOURNAL OF PATHOLOGY 2022; 192:1648-1657. [PMID: 36174679 DOI: 10.1016/j.ajpath.2022.09.003] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 09/01/2022] [Accepted: 09/13/2022] [Indexed: 06/16/2023]
Abstract
Skeletal muscle atrophy is the consequence of protein degradation exceeding protein synthesis because of disease, aging, and physical inactivity. Patients with skeletal muscle atrophy have decreased muscle mass and fiber cross-sectional area, and experience reduced survival quality and motor function. The forkhead box O (FOXO) signaling pathway plays an important role in the pathogenesis of skeletal muscle atrophy by regulating E3 ubiquitin ligases and some autophagy factors. However, the mechanism of FOXO signaling pathway leading to skeletal muscle atrophy is still unclear. The development of treatment strategies for skeletal muscle atrophy has been a thorny clinical problem. FOXO-targeted therapy to treat skeletal muscle atrophy is a promising approach, and an increasing number of relevant studies have been reported. This article reviews the mechanism and therapeutic targets of the FOXO signaling pathway mediating skeletal muscle atrophy, and provides ideas for the clinical treatment of this condition.
Collapse
Affiliation(s)
- Kun Chen
- Department of Orthopaedics, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, China
| | - Peng Gao
- Department of Orthopaedics, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, China
| | - Zongchao Li
- Department of Orthopaedics, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, China
| | - Aonan Dai
- Department of Orthopaedics, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, China
| | - Ming Yang
- Department of Orthopaedics, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, China
| | - Siyu Chen
- Department of Sports Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China; School of Medicine, Guangxi University of Chinese Medicine, Nanning, China
| | - Jingyue Su
- Department of Sports Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China; School of Medicine, Guangxi University of Chinese Medicine, Nanning, China
| | - Zhenhan Deng
- Department of Sports Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China; School of Medicine, Guangxi University of Chinese Medicine, Nanning, China.
| | - Liangjun Li
- Department of Orthopaedics, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, China.
| |
Collapse
|
25
|
Zhao Y, Yu Y, Ommati MM, Xu J, Wang J, Zhang J, Sun Z, Niu R, Wang J. Multiomics Analysis Revealed the Molecular Mechanism of miRNAs in Fluoride-Induced Hepatic Glucose and Lipid Metabolism Disorders. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:14284-14295. [PMID: 36222057 DOI: 10.1021/acs.jafc.2c03049] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Fluoride-induced liver injury seriously endangers human and animal health and animal food safety, but the underlying mechanism remains unclear. This study aims to explore the mechanism of miRNAs in fluoride-induced hepatic glycolipid metabolism disorders. C57 male mice were used to establish the fluorosis model (22.62 mg/L F-, 12 weeks). The results indicated that fluoride increased fluoride levels, impaired the structure and function, and disrupted the glycolipid metabolism in the liver. Furthermore, the sequencing results showed that fluoride exposure resulted in the differential expression of 35 miRNAs and 480 mRNAs, of which 23 miRNAs were related to glycolipid metabolism. miRNA-mRNA network analyses and RT-PCR revealed that miRNAs mediated fluoride-induced disturbances in the hepatic glycolipid metabolism. Its possible mechanism was to regulate the insulin pathway, PPAR pathway, and FOXO pathway, which in turn affected the bile secretion, the metabolic processes of glucose, the decomposition of lipids, and the synthesis of unsaturated fatty acids in the liver. This study provides a theoretical basis for miRNAs as diagnostic indicators and target drugs for the treatment of fluoride-induced liver injury.
Collapse
Affiliation(s)
- Yangfei Zhao
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi 030801, China
| | - Yanghuan Yu
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi 030801, China
| | - Mohammad Mehdi Ommati
- College of Life Sciences, Shanxi Agricultural University, Taigu, Shanxi 030801, China
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz 71348-14336, Iran
| | - Jipeng Xu
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi 030801, China
| | - Jinming Wang
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi 030801, China
| | - Jianhai Zhang
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi 030801, China
| | - Zilong Sun
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi 030801, China
| | - Ruiyan Niu
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi 030801, China
| | - Jundong Wang
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi 030801, China
| |
Collapse
|
26
|
Skeletal muscle mitochondrial remodeling in heart failure: An update on mechanisms and therapeutic opportunities. Biomed Pharmacother 2022; 155:113833. [DOI: 10.1016/j.biopha.2022.113833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 10/04/2022] [Accepted: 10/06/2022] [Indexed: 11/22/2022] Open
|
27
|
Zhao M, Banhos Danneskiold-Samsøe N, Ulicna L, Nguyen Q, Voilquin L, Lee DE, White JP, Jiang Z, Cuthbert N, Paramasivam S, Bielczyk-Maczynska E, Van Rechem C, Svensson KJ. Phosphoproteomic mapping reveals distinct signaling actions and activation of muscle protein synthesis by Isthmin-1. eLife 2022; 11:e80014. [PMID: 36169399 PMCID: PMC9592085 DOI: 10.7554/elife.80014] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 09/27/2022] [Indexed: 12/02/2022] Open
Abstract
The secreted protein isthmin-1 (Ism1) mitigates diabetes by increasing adipocyte and skeletal muscle glucose uptake by activating the PI3K-Akt pathway. However, while both Ism1 and insulin converge on these common targets, Ism1 has distinct cellular actions suggesting divergence in downstream intracellular signaling pathways. To understand the biological complexity of Ism1 signaling, we performed phosphoproteomic analysis after acute exposure, revealing overlapping and distinct pathways of Ism1 and insulin. We identify a 53% overlap between Ism1 and insulin signaling and Ism1-mediated phosphoproteome-wide alterations in ~450 proteins that are not shared with insulin. Interestingly, we find several unknown phosphorylation sites on proteins related to protein translation, mTOR pathway, and, unexpectedly, muscle function in the Ism1 signaling network. Physiologically, Ism1 ablation in mice results in altered proteostasis, including lower muscle protein levels under fed and fasted conditions, reduced amino acid incorporation into proteins, and reduced phosphorylation of the key protein synthesis effectors Akt and downstream mTORC1 targets. As metabolic disorders such as diabetes are associated with accelerated loss of skeletal muscle protein content, these studies define a non-canonical mechanism by which this antidiabetic circulating protein controls muscle biology.
Collapse
Affiliation(s)
- Meng Zhao
- Department of Pathology, Stanford University School of MedicineStanfordUnited States
- Stanford Diabetes Research Center, Stanford University School of MedicineStanfordUnited States
- Stanford Cardiovascular Institute, Stanford University School of MedicineStanfordUnited States
| | | | - Livia Ulicna
- Department of Pathology, Stanford University School of MedicineStanfordUnited States
| | - Quennie Nguyen
- Department of Pathology, Stanford University School of MedicineStanfordUnited States
| | - Laetitia Voilquin
- Department of Pathology, Stanford University School of MedicineStanfordUnited States
- Stanford Diabetes Research Center, Stanford University School of MedicineStanfordUnited States
- Stanford Cardiovascular Institute, Stanford University School of MedicineStanfordUnited States
| | - David E Lee
- Duke Molecular Physiology Institute, Duke University School of MedicineDurhamUnited States
- Department of Medicine, Duke University School of MedicineDurhamUnited States
| | - James P White
- Duke Molecular Physiology Institute, Duke University School of MedicineDurhamUnited States
- Department of Medicine, Duke University School of MedicineDurhamUnited States
- Duke Center for the Study of Aging and Human Development, Duke University School of MedicineDurhamUnited States
| | - Zewen Jiang
- Department of Pathology, Stanford University School of MedicineStanfordUnited States
- Department of Laboratory Medicine, University of California, San FranciscoSan FranciscoUnited States
- Diabetes Center, University of California, San FranciscoSan FranciscoUnited States
| | - Nickeisha Cuthbert
- Department of Pathology, Stanford University School of MedicineStanfordUnited States
| | - Shrika Paramasivam
- Department of Pathology, Stanford University School of MedicineStanfordUnited States
| | - Ewa Bielczyk-Maczynska
- Stanford Diabetes Research Center, Stanford University School of MedicineStanfordUnited States
- Stanford Cardiovascular Institute, Stanford University School of MedicineStanfordUnited States
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of MedicineStanfordUnited States
| | - Capucine Van Rechem
- Department of Pathology, Stanford University School of MedicineStanfordUnited States
| | - Katrin J Svensson
- Department of Pathology, Stanford University School of MedicineStanfordUnited States
- Stanford Diabetes Research Center, Stanford University School of MedicineStanfordUnited States
- Stanford Cardiovascular Institute, Stanford University School of MedicineStanfordUnited States
| |
Collapse
|
28
|
Yoh K, Ikeda K, Nagai S, Horie K, Takeda S, Inoue S. Constitutive activation of estrogen receptor α signaling in muscle prolongs exercise endurance in mice. Biochem Biophys Res Commun 2022; 628:11-17. [DOI: 10.1016/j.bbrc.2022.08.064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 08/19/2022] [Accepted: 08/22/2022] [Indexed: 11/16/2022]
|
29
|
Pleiotropic Effects of IGF1 on the Oocyte. Cells 2022; 11:cells11101610. [PMID: 35626647 PMCID: PMC9140015 DOI: 10.3390/cells11101610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 05/08/2022] [Accepted: 05/09/2022] [Indexed: 12/10/2022] Open
Abstract
A woman’s endocrine system plays a crucial role in orchestrating cellular interactions throughout her life. The growth hormone (GH) and insulin-like growth factor (IGF) system appears to impact crucial reproductive events and cell types of the ovary, such as granulosa cells, theca cells, and oocytes. Further, IGF1 is a cornerstone during embryonic development and influences predominantly developing and pre-antral follicles. In this commentary, we will emphasize the pleiotropic effects of IGF1 on physiological processes inside the egg. Herein, we will provide a brief overview on IGF1 related cell signal transduction pathways during the maturation and aging of oocytes. We aim to elucidate from a molecular and biochemical point of view if IGF1 in women with metabolic imbalances such as obesity or diabetes could be used in clinics as a novel, reliable estimator for the developmental competence of an oocyte.
Collapse
|
30
|
Bhardwaj G, Penniman CM, Klaus K, Weatherford ET, Pan H, Dreyfuss JM, Nair KS, Kahn CR, O’Neill BT. Transcriptomic Regulation of Muscle Mitochondria and Calcium Signaling by Insulin/IGF-1 Receptors Depends on FoxO Transcription Factors. Front Physiol 2022; 12:779121. [PMID: 35185597 PMCID: PMC8855073 DOI: 10.3389/fphys.2021.779121] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 12/23/2021] [Indexed: 02/02/2023] Open
Abstract
Insulin and IGF-1, acting through the insulin receptor (IR) and IGF-1 receptor (IGF1R), maintain muscle mass and mitochondrial function, at least part of which occurs via their action to regulate gene expression. Here, we show that while muscle-specific deletion of IR or IGF1R individually results in only modest changes in the muscle transcriptome, combined deletion of IR/IGF1R (MIGIRKO) altered > 3000 genes, including genes involved in mitochondrial dysfunction, fibrosis, cardiac hypertrophy, and pathways related to estrogen receptor, protein kinase A (PKA), and calcium signaling. Functionally, this was associated with decreased mitochondrial respiration and increased ROS production in MIGIRKO muscle. To determine the role of FoxOs in these changes, we performed RNA-Seq on mice with muscle-specific deletion of FoxO1/3/4 (M-FoxO TKO) or combined deletion of IR, IGF1R, and FoxO1/3/4 in a muscle quintuple knockout (M-QKO). This revealed that among IR/IGF1R regulated genes, >97% were FoxO-dependent, and their expression was normalized in M-FoxO TKO and M-QKO muscle. FoxO-dependent genes were related to oxidative phosphorylation, inflammatory signaling, and TCA cycle. Metabolomic analysis showed accumulation of TCA cycle metabolites in MIGIRKO, which was reversed in M-QKO muscle. Likewise, calcium signaling genes involved in PKA signaling and sarcoplasmic reticulum calcium homeostasis were markedly altered in MIGIRKO muscle but normalized in M-QKO. Thus, combined loss of insulin and IGF-1 action in muscle transcriptionally alters mitochondrial function and multiple regulatory and signaling pathways, and these changes are mediated by FoxO transcription factors.
Collapse
Affiliation(s)
- Gourav Bhardwaj
- Fraternal Order of Eagles Diabetes Research Center and Division of Endocrinology and Metabolism, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, United States
| | - Christie M. Penniman
- Fraternal Order of Eagles Diabetes Research Center and Division of Endocrinology and Metabolism, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, United States
| | - Katherine Klaus
- Division of Endocrinology and Metabolism, Mayo Clinic College of Medicine and Science, Rochester, MN, United States
| | - Eric T. Weatherford
- Fraternal Order of Eagles Diabetes Research Center and Division of Endocrinology and Metabolism, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, United States
| | - Hui Pan
- Bioinformatics and Biostatistics Core, Joslin Diabetes Center, Harvard Medical School, Boston, MA, United States
| | - Jonathan M. Dreyfuss
- Bioinformatics and Biostatistics Core, Joslin Diabetes Center, Harvard Medical School, Boston, MA, United States
| | - K. Sreekumaran Nair
- Division of Endocrinology and Metabolism, Mayo Clinic College of Medicine and Science, Rochester, MN, United States
| | - C. Ronald Kahn
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA, United States
| | - Brian T. O’Neill
- Fraternal Order of Eagles Diabetes Research Center and Division of Endocrinology and Metabolism, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, United States
- Veterans Affairs Health Care System, Iowa City, IA, United States
| |
Collapse
|
31
|
A theoretical argument to support the biological benefits for insulin stimulating mitochondrial oxidative phosphorylation. CURRENT OPINION IN PHYSIOLOGY 2022. [DOI: 10.1016/j.cophys.2022.100491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
32
|
Transforming Growth Factor-Beta in Skeletal Muscle Wasting. Int J Mol Sci 2022; 23:ijms23031167. [PMID: 35163088 PMCID: PMC8835446 DOI: 10.3390/ijms23031167] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 01/11/2022] [Accepted: 01/19/2022] [Indexed: 12/21/2022] Open
Abstract
Transforming growth factor-beta (TGF-β) is part of a family of molecules that is present in many body tissues and performs many different functions. Evidence has been obtained from mice and human cancer patients with bony metastases and non-metastatic disease, as well as pediatric burn patients, that inflammation leads to bone resorption and release of TGF-β from the bone matrix with paracrine effects on muscle protein balance, possibly mediated by the generation of reactive oxygen species. Whether immobilization, which confounds the etiology of bone resorption in burn injury, also leads to the release of TGF-β from bone contributing to muscle wasting in other conditions is unclear. The use of anti-resorptive therapy in both metastatic cancer patients and pediatric burn patients has been successful in the prevention of muscle wasting, thereby creating an additional therapeutic niche for this class of drugs. The liberation of TGF-β may be one way in which bone helps to control muscle mass, but further investigation will be necessary to assess whether the rate of bone resorption is the determining factor for the release of TGF-β. Moreover, whether different resorptive conditions, such as immobilization and hyperparathyroidism, also involve TGF-β release in the pathogenesis of muscle wasting needs to be investigated.
Collapse
|
33
|
Ferraz RS, Santos LCB, da-Silva-Cruz RL, Braga-da-Silva CH, Magalhães L, Ribeiro-dos-Santos A, Vidal A, Vinasco-Sandoval T, Reis-das-Mercês L, Sena-dos-Santos C, Pereira AL, Silva LSD, de Melo FTC, de Souza ACCB, Leal VSG, de Figueiredo PBB, Neto JFA, de Moraes LV, de Lemos GN, de Queiroz NNM, Felício KM, Cavalcante GC, Ribeiro-dos-Santos Â, Felício JS. Global miRNA expression reveals novel nuclear and mitochondrial interactions in Type 1 diabetes mellitus. Front Endocrinol (Lausanne) 2022; 13:1033809. [PMID: 36506063 PMCID: PMC9731375 DOI: 10.3389/fendo.2022.1033809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 11/03/2022] [Indexed: 11/27/2022] Open
Abstract
BACKGROUND Considering the potential role of miRNAs as biomarkers and their interaction with both nuclear and mitochondrial genes, we investigated the miRNA expression profile in type 1 diabetes (T1DM) patients, including the pathways in which they are involved considering both nuclear and mitochondrial functions. METHODS We analyzed samples of T1DM patients and control individuals (normal glucose tolerance) by high throughput miRNA sequencing (miRNome). Next, five miRNAs - hsa-miR-26b-5p, hsa-let-7i-5p, hsa-miR-143-3p, hsa-miR-501-3p and hsa-miR-100-5p - were validated by RT-qPCR. The identification of target genes was extracted from miRTarBase and mitoXplorer database. We also performed receiver operating characteristic (ROC) curves and miRNAs that had an AUC > 0.85 were considered potential biomarkers. RESULTS Overall, 41 miRNAs were differentially expressed in T1DM patients compared to control. Hsa-miR-21-5p had the highest number of predicted target genes and was associated with several pathways, including insulin signaling and apoptosis. 34.1% (14/41) of the differentially expressed miRNAs also targeted mitochondrial genes, and 80.5% (33/41) of them targeted nuclear genes involved in the mitochondrial metabolism. All five validated miRNAs were upregulated in T1DM. Among them, hsa-miR-26b-5p showed AUC>0.85, being suggested as potential biomarker to T1DM. CONCLUSION Our results demonstrated 41 DE miRNAs that had a great accuracy in discriminating T1DM and control group. Furthermore, we demonstrate the influence of these miRNAs on numerous metabolic pathways, including mitochondrial metabolism. Hsa-miR-26b-5p and hsa-miR-21-5p were highlighted in our results, possibly acting on nuclear and mitochondrial dysfunction and, subsequently, T1DM dysregulation.
Collapse
Affiliation(s)
- Rafaella Sousa Ferraz
- Laboratory of Human and Medical Genetics, Graduate Program in Genetics and Molecular Biology, Federal University of Para, Belem, PA, Brazil
| | - Lucas Cauê Bezerra Santos
- Laboratory of Human and Medical Genetics, Graduate Program in Genetics and Molecular Biology, Federal University of Para, Belem, PA, Brazil
| | - Rebecca Lais da-Silva-Cruz
- Laboratory of Human and Medical Genetics, Graduate Program in Genetics and Molecular Biology, Federal University of Para, Belem, PA, Brazil
| | - Cintia Helena Braga-da-Silva
- Laboratory of Human and Medical Genetics, Graduate Program in Genetics and Molecular Biology, Federal University of Para, Belem, PA, Brazil
| | - Leandro Magalhães
- Laboratory of Human and Medical Genetics, Graduate Program in Genetics and Molecular Biology, Federal University of Para, Belem, PA, Brazil
| | - Arthur Ribeiro-dos-Santos
- Laboratory of Human and Medical Genetics, Graduate Program in Genetics and Molecular Biology, Federal University of Para, Belem, PA, Brazil
| | - Amanda Vidal
- Laboratory of Human and Medical Genetics, Graduate Program in Genetics and Molecular Biology, Federal University of Para, Belem, PA, Brazil
- Instituto Tecnológico Vale Desenvolvimento Sustentável Vale, Institute of Technology, Belem, PA, Brazil
| | - Tatiana Vinasco-Sandoval
- Laboratoire de Génomique et Radiobiologie de la Kératinopoïèse, Institut de Biologie François Jacob, CEA/DRF/IRCM, Evry, France
| | - Laís Reis-das-Mercês
- Laboratory of Human and Medical Genetics, Graduate Program in Genetics and Molecular Biology, Federal University of Para, Belem, PA, Brazil
| | - Camille Sena-dos-Santos
- Laboratory of Human and Medical Genetics, Graduate Program in Genetics and Molecular Biology, Federal University of Para, Belem, PA, Brazil
| | | | | | - Franciane T. Cunha de Melo
- Endocrinology Research Center, Joao de Barros Barreto University Hospital, Federal University of Para, Belem, PA, Brazil
| | - Ana Carolina C. Braga de Souza
- Endocrinology Research Center, Joao de Barros Barreto University Hospital, Federal University of Para, Belem, PA, Brazil
| | - Valéria S. Galvão Leal
- Endocrinology Research Center, Joao de Barros Barreto University Hospital, Federal University of Para, Belem, PA, Brazil
| | | | - João F. Abrahão Neto
- Endocrinology Research Center, Joao de Barros Barreto University Hospital, Federal University of Para, Belem, PA, Brazil
| | - Lorena Vilhena de Moraes
- Endocrinology Research Center, Joao de Barros Barreto University Hospital, Federal University of Para, Belem, PA, Brazil
| | - Gabriela Nascimento de Lemos
- Endocrinology Research Center, Joao de Barros Barreto University Hospital, Federal University of Para, Belem, PA, Brazil
| | | | - Karem Miléo Felício
- Endocrinology Research Center, Joao de Barros Barreto University Hospital, Federal University of Para, Belem, PA, Brazil
| | - Giovanna C. Cavalcante
- Laboratory of Human and Medical Genetics, Graduate Program in Genetics and Molecular Biology, Federal University of Para, Belem, PA, Brazil
| | - Ândrea Ribeiro-dos-Santos
- Laboratory of Human and Medical Genetics, Graduate Program in Genetics and Molecular Biology, Federal University of Para, Belem, PA, Brazil
- Oncology Research Center, Graduate Program in Oncology and Medical Sciences, Joao de Barros Barreto University Hospital, Federal University of Para, Belem, PA, Brazil
| | - João Soares Felício
- Endocrinology Research Center, Joao de Barros Barreto University Hospital, Federal University of Para, Belem, PA, Brazil
- *Correspondence: João Soares Felício,
| |
Collapse
|