1
|
Han G, Li D, Zhang H, Li C, Yang L, Ma T, Wang X, Ma B, Wu X, Tao Y, Wang Z, Wang A, Chao HW, Jin Y, Chen H. A Transcriptomic Dataset of Liver Tissues from Global and Liver-Specific Bmal1 Knockout Mice. Sci Data 2025; 12:199. [PMID: 39900971 PMCID: PMC11790919 DOI: 10.1038/s41597-025-04545-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 01/28/2025] [Indexed: 02/05/2025] Open
Abstract
The circadian clock regulates various physiological processes in mammals. The core circadian clock gene Bmal1 is crucial for maintaining the oscillations of the circadian clock system by controlling the rhythmic expression of numerous circadian clock-controlled genes. To explore the transcriptional changes associated with Bmal1 deletion in liver tissues, we collected liver tissues from global and liver-specific Bmal1 knockout mice, along with their respective control groups, at two circadian time points (CT2 and CT14) and used them for transcriptome sequencing analysis. Genotyping, locomotor activity analysis, and comprehensive quality control analyses, including base quality scores, GC content, and mapping rates, confirmed the high quality of sequencing data. Differential expression analysis and RT-qPCR validation confirmed the reliability and validity of the dataset. These data offer a valuable resource for researchers investigating the role of BMAL1 in liver physiology, pathology, and the broader field of circadian biology.
Collapse
Affiliation(s)
- Guohao Han
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, 712100, China
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture and Rural Affairs, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Dan Li
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, 712100, China
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture and Rural Affairs, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Haisen Zhang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, 712100, China
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture and Rural Affairs, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Chao Li
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, 712100, China
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture and Rural Affairs, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Luda Yang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, 712100, China
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture and Rural Affairs, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Tiantian Ma
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, 712100, China
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture and Rural Affairs, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Xuerong Wang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, 712100, China
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture and Rural Affairs, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Bairong Ma
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, 712100, China
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture and Rural Affairs, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Xiaodie Wu
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, 712100, China
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture and Rural Affairs, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Yang Tao
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, 712100, China
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture and Rural Affairs, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Ziang Wang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, 712100, China
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture and Rural Affairs, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Aihua Wang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, 712100, China
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture and Rural Affairs, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Hsu-Wen Chao
- Department of Physiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, 11031, Taiwan.
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, 11031, Taiwan.
- Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan.
| | - Yaping Jin
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, 712100, China.
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture and Rural Affairs, Northwest A&F University, Yangling, Shaanxi, 712100, China.
| | - Huatao Chen
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, 712100, China.
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture and Rural Affairs, Northwest A&F University, Yangling, Shaanxi, 712100, China.
| |
Collapse
|
2
|
Prabakaran AD, Chung HJ, McFarland K, Govindarajan T, Soussi FEA, Durumutla HB, Villa C, Piczer K, Latimer H, Werbrich C, Akinborewa O, Horning R, Quattrocelli M. The human genetic variant rs6190 unveils Foxc1 and Arid5a as novel pro-metabolic targets of the glucocorticoid receptor in muscle. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.03.28.586997. [PMID: 38585940 PMCID: PMC10996618 DOI: 10.1101/2024.03.28.586997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
The genetic determinants of the glucocorticoid receptor (GR) metabolic action remain largely unelucidated. This is a compelling gap in knowledge for the GR single nucleotide polymorphism (SNP) rs6190 (p.R23K), which has been associated in humans with enhanced metabolic health but whose mechanism of action remains completely unknown. We generated transgenic knock-in mice genocopying this polymorphism to elucidate how the mutant GR impacts metabolism. Compared to non-mutant littermates, mutant mice showed increased insulin sensitivity on regular chow and high-fat diet, blunting the diet-induced adverse effects on adiposity and exercise intolerance. Overlay of RNA-seq and ChIP-seq profiling in skeletal muscle revealed increased transactivation of Foxc1 and Arid5A genes by the mutant GR. Using myotropic adeno-associated viruses for in vivo overexpression or knockdown in muscle, we found that Foxc1 was required and sufficient for normal expression levels of insulin response pathway genes Insr and Irs1 , promoting muscle insulin sensitivity. In parallel, Arid5a was required and sufficient to transcriptionally repress the lipid uptake genes C 36 and Fabp4 , reducing muscle triacylglycerol accumulation. Moreover, the Foxc1 and Arid5a programs in muscle were divergently changed by glucocorticoid regimens with opposite metabolic outcomes in muscle. Finally, we found a direct human relevance for our mechanism of SNP action in the UK Biobank and All of Us datasets, where the rs6190 SNP correlated with pro-metabolic changes in BMI, lean mass, strength and glucose control according to zygosity. Collectively, our study leveraged a human nuclear receptor coding variant to unveil novel epigenetic regulators of muscle metabolism. Graphical abstract
Collapse
|
3
|
Quinlan JI, Viggars MR. Tendinopathy: A 'timely' matter. J Physiol 2024; 602:6525-6526. [PMID: 36869867 DOI: 10.1113/jp284518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 03/02/2023] [Indexed: 03/05/2023] Open
Affiliation(s)
- Jonathan I Quinlan
- NIHR Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust and University of Birmingham, Birmingham, UK
- School of Sport, Exercise and Rehabilitation Sciences, University of Birmingham, Birmingham, UK
| | - Mark R Viggars
- Department of Physiology and Aging, University of Florida, Gainesville, FL, USA
- Myology Institute, University of Florida, Gainesville, FL, USA
| |
Collapse
|
4
|
Durumutla HB, Prabakaran AD, El Abdellaoui Soussi F, Akinborewa O, Latimer H, McFarland K, Piczer K, Werbrich C, Jain MK, Haldar SM, Quattrocelli M. Glucocorticoid chronopharmacology promotes glucose metabolism in heart through a cardiomyocyte-autonomous transactivation program. JCI Insight 2024; 9:e182599. [PMID: 39378111 PMCID: PMC11601906 DOI: 10.1172/jci.insight.182599] [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: 05/01/2024] [Accepted: 10/04/2024] [Indexed: 10/10/2024] Open
Abstract
Circadian time of intake gates the cardioprotective effects of glucocorticoid administration in both healthy and infarcted hearts. The cardiomyocyte-specific glucocorticoid receptor (GR) and its cofactor, Krüppel-like factor 15 (KLF15), play critical roles in maintaining normal heart function in the long term and serve as pleiotropic regulators of cardiac metabolism. Despite this understanding, the cardiomyocyte-autonomous metabolic targets influenced by the concerted epigenetic action of the GR/KLF15 axis remain undefined. Here, we demonstrated the critical roles of the cardiomyocyte-specific GR and KLF15 in orchestrating a circadian-dependent glucose oxidation program within the heart. Combining integrated transcriptomics and epigenomics with cardiomyocyte-specific inducible ablation of GR or KLF15, we identified their synergistic role in the activation of adiponectin receptor expression (Adipor1) and the mitochondrial pyruvate complex (Mpc1/2), thereby enhancing insulin-stimulated glucose uptake and pyruvate oxidation. Furthermore, in obese diabetic (db/db) mice exhibiting insulin resistance and impaired glucose oxidation, light-phase prednisone administration, as opposed to dark-phase prednisone dosing, restored cardiomyocyte glucose oxidation and improved diastolic function. These effects were blocked by combined in vivo knockdown of GR and KLF15 levels in db/db hearts. In summary, this study leveraged the circadian-dependent cardioprotective effects of glucocorticoids to identify cardiomyocyte-autonomous targets for the GR/KLF15 axis in glucose metabolism.
Collapse
Affiliation(s)
- Hima Bindu Durumutla
- Molecular Cardiovascular Biology, Heart Institute, Cincinnati Children’s Hospital Medical Center and Department of Pediatrics, and
| | - Ashok Daniel Prabakaran
- Molecular Cardiovascular Biology, Heart Institute, Cincinnati Children’s Hospital Medical Center and Department of Pediatrics, and
| | - Fadoua El Abdellaoui Soussi
- Molecular Cardiovascular Biology, Heart Institute, Cincinnati Children’s Hospital Medical Center and Department of Pediatrics, and
| | - Olukunle Akinborewa
- Molecular Cardiovascular Biology, Heart Institute, Cincinnati Children’s Hospital Medical Center and Department of Pediatrics, and
- Department of Pharmacology, Physiology and Neurobiology, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Hannah Latimer
- Molecular Cardiovascular Biology, Heart Institute, Cincinnati Children’s Hospital Medical Center and Department of Pediatrics, and
| | - Kevin McFarland
- Molecular Cardiovascular Biology, Heart Institute, Cincinnati Children’s Hospital Medical Center and Department of Pediatrics, and
| | - Kevin Piczer
- Molecular Cardiovascular Biology, Heart Institute, Cincinnati Children’s Hospital Medical Center and Department of Pediatrics, and
| | - Cole Werbrich
- Molecular Cardiovascular Biology, Heart Institute, Cincinnati Children’s Hospital Medical Center and Department of Pediatrics, and
| | - Mukesh K. Jain
- Department of Cell Biology and Biochemistry, Brown University, Providence, Rhode Island, USA
| | - Saptarsi M. Haldar
- Amgen Research, South San Francisco, California, USA
- Gladstone Institutes, San Francisco, California, USA
- Department of Medicine, Cardiology Division, UCSF, San Francisco, California, USA
| | - Mattia Quattrocelli
- Molecular Cardiovascular Biology, Heart Institute, Cincinnati Children’s Hospital Medical Center and Department of Pediatrics, and
| |
Collapse
|
5
|
Durumutla HB, Prabakaran AD, Soussi FEA, Akinborewa O, Latimer H, McFarland K, Piczer K, Werbrich C, Jain MK, Haldar SM, Quattrocelli M. Glucocorticoid chrono-pharmacology unveils novel targets for the cardiomyocyte-specific GR-KLF15 axis in cardiac glucose metabolism. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.12.18.572210. [PMID: 38187555 PMCID: PMC10769285 DOI: 10.1101/2023.12.18.572210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Circadian time-of-intake gates the cardioprotective effects of glucocorticoid administration in both healthy and infarcted hearts. The cardiomyocyte-specific glucocorticoid receptor (GR) and its co-factor, Krüppel-like factor (Klf15), play critical roles in maintaining normal heart function in the long-term and serve as pleiotropic regulators of cardiac metabolism. Despite this understanding, the cardiomyocyte-autonomous metabolic targets influenced by the concerted epigenetic action of GR-Klf15 axis remain undefined. Here, we demonstrate the critical roles of the cardiomyocyte-specific GR and Klf15 in orchestrating a circadian-dependent glucose oxidation program within the heart. Combining integrated transcriptomics and epigenomics with cardiomyocyte-specific inducible ablation of GR or Klf15, we identified their synergistic role in the activation of adiponectin receptor expression (Adipor1) and the mitochondrial pyruvate complex (Mpc1/2), thereby enhancing insulin-stimulated glucose uptake and pyruvate oxidation. Furthermore, in obese diabetic (db/db) mice exhibiting insulin resistance and impaired glucose oxidation, light-phase prednisone administration, as opposed to dark-phase prednisone dosing, effectively restored cardiomyocyte glucose oxidation and improved diastolic function towards control-like levels in a sex-independent manner. Collectively, our findings uncover novel cardiomyocyte-autonomous metabolic targets of the GR-Klf15 axis. This study highlights the circadian-dependent cardioprotective effects of glucocorticoids on cardiomyocyte glucose metabolism, providing critical insights into chrono-pharmacological strategies for glucocorticoid therapy in cardiovascular disease.
Collapse
Affiliation(s)
- Hima Bindu Durumutla
- Molecular Cardiovascular Biology, Heart Institute, Cincinnati Children’s Hospital Medical Center and Dept. Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Ashok Daniel Prabakaran
- Molecular Cardiovascular Biology, Heart Institute, Cincinnati Children’s Hospital Medical Center and Dept. Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Fadoua El Abdellaoui Soussi
- Molecular Cardiovascular Biology, Heart Institute, Cincinnati Children’s Hospital Medical Center and Dept. Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Olukunle Akinborewa
- Molecular Cardiovascular Biology, Heart Institute, Cincinnati Children’s Hospital Medical Center and Dept. Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Department of Pharmacology, Physiology and Neurobiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Hannah Latimer
- Molecular Cardiovascular Biology, Heart Institute, Cincinnati Children’s Hospital Medical Center and Dept. Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Kevin McFarland
- Molecular Cardiovascular Biology, Heart Institute, Cincinnati Children’s Hospital Medical Center and Dept. Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Kevin Piczer
- Molecular Cardiovascular Biology, Heart Institute, Cincinnati Children’s Hospital Medical Center and Dept. Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Cole Werbrich
- Molecular Cardiovascular Biology, Heart Institute, Cincinnati Children’s Hospital Medical Center and Dept. Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Mukesh K Jain
- Dept Cell Biology and Biochemistry, Brown University, Providence, RI, USA
| | - Saptarsi M Haldar
- Amgen Research, South San Francisco, CA, USA; Gladstone Institutes, San Francisco, CA, USA and Dept Medicine, Cardiology Division, UCSF, San Francisco, CA, USA
| | - Mattia Quattrocelli
- Molecular Cardiovascular Biology, Heart Institute, Cincinnati Children’s Hospital Medical Center and Dept. Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| |
Collapse
|
6
|
Liu Q, Zhang Y. Biological Clock Perspective in Rheumatoid Arthritis. Inflammation 2024:10.1007/s10753-024-02120-4. [PMID: 39126449 DOI: 10.1007/s10753-024-02120-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Revised: 06/13/2024] [Accepted: 08/01/2024] [Indexed: 08/12/2024]
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune disease characterized by systemic polyarticular pain, and its main pathological features include inflammatory cell infiltration, synovial fibroblast proliferation, and cartilage erosion. Immune cells, synovial cells and neuroendocrine factors play pivotal roles in the pathophysiological mechanism underlying rheumatoid arthritis. Biological clock genes regulate immune cell functions, which is linked to rhythmic changes in arthritis pathology. Additionally, the interaction between biological clock genes and neuroendocrine factors is also involved in rhythmic changes in rheumatoid arthritis. This review provides an overview of the contributions of circadian rhythm genes to RA pathology, including their interaction with the immune system and their involvement in regulating the secretion and function of neuroendocrine factors. A molecular understanding of the role of the circadian rhythm in RA may offer insights for effective disease management.
Collapse
Affiliation(s)
- Qingxue Liu
- Gengjiu Clinical College of Anhui Medical University; Anhui Zhongke Gengjiu Hospital, Hefei, 230051, China
| | - Yihao Zhang
- Department of Health Inspection and Quarantine, School of Public Health, Anhui Medical University, 81 Meishan Rd, Hefei, 230032, China.
- Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, Hefei, 230032, China.
| |
Collapse
|
7
|
Prabakaran AD, McFarland K, Miz K, Durumutla HB, Piczer K, El Abdellaoui Soussi F, Latimer H, Werbrich C, Chung HJ, Blair NS, Millay DP, Morris AJ, Prideaux B, Finck BN, Quattrocelli M. Intermittent glucocorticoid treatment improves muscle metabolism via the PGC1α/Lipin1 axis in an aging-related sarcopenia model. J Clin Invest 2024; 134:e177427. [PMID: 38702076 PMCID: PMC11142738 DOI: 10.1172/jci177427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Accepted: 04/09/2024] [Indexed: 05/06/2024] Open
Abstract
Sarcopenia burdens the older population through loss of muscle energy and mass, yet treatments to functionally rescue both parameters are lacking. The glucocorticoid prednisone remodels muscle metabolism on the basis of frequency of intake, but its mechanisms in sarcopenia are unknown. We found that once-weekly intermittent prednisone administration rescued muscle quality in aged 24-month-old mice to a level comparable to that seen in young 4-month-old mice. We discovered an age- and sex-independent glucocorticoid receptor transactivation program in muscle encompassing peroxisome proliferator-activated receptor γ coactivator 1 α (PGC1α) and its cofactor Lipin1. Treatment coordinately improved mitochondrial abundance through isoform 1 and muscle mass through isoform 4 of the myocyte-specific PGC1α, which was required for the treatment-driven increase in carbon shuttling from glucose oxidation to amino acid biogenesis. We also probed myocyte-specific Lipin1 as a nonredundant factor coaxing PGC1α upregulation to the stimulation of both oxidative and anabolic effects. Our study unveils an aging-resistant druggable program in myocytes for the coordinated rescue of energy and mass in sarcopenia.
Collapse
Affiliation(s)
- Ashok D. Prabakaran
- Molecular Cardiovascular Biology, Heart Institute, Cincinnati Children’s Hospital Medical Center (CCHMC) and Department Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Kevin McFarland
- Molecular Cardiovascular Biology, Heart Institute, Cincinnati Children’s Hospital Medical Center (CCHMC) and Department Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Karen Miz
- Molecular Cardiovascular Biology, Heart Institute, Cincinnati Children’s Hospital Medical Center (CCHMC) and Department Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Hima Bindu Durumutla
- Molecular Cardiovascular Biology, Heart Institute, Cincinnati Children’s Hospital Medical Center (CCHMC) and Department Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Kevin Piczer
- Molecular Cardiovascular Biology, Heart Institute, Cincinnati Children’s Hospital Medical Center (CCHMC) and Department Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Fadoua El Abdellaoui Soussi
- Molecular Cardiovascular Biology, Heart Institute, Cincinnati Children’s Hospital Medical Center (CCHMC) and Department Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Hannah Latimer
- Molecular Cardiovascular Biology, Heart Institute, Cincinnati Children’s Hospital Medical Center (CCHMC) and Department Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Cole Werbrich
- Molecular Cardiovascular Biology, Heart Institute, Cincinnati Children’s Hospital Medical Center (CCHMC) and Department Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Hyun-Jy Chung
- Molecular Cardiovascular Biology, Heart Institute, Cincinnati Children’s Hospital Medical Center (CCHMC) and Department Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - N. Scott Blair
- Molecular Cardiovascular Biology, Heart Institute, Cincinnati Children’s Hospital Medical Center (CCHMC) and Department Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Douglas P. Millay
- Molecular Cardiovascular Biology, Heart Institute, Cincinnati Children’s Hospital Medical Center (CCHMC) and Department Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Andrew J. Morris
- Department Pharmacology and Toxicology, University of Arkansas for Medical Sciences (UAMS) College of Medicine and Central Arkansas VA Healthcare System, Little Rock, Arkansas, USA
| | - Brendan Prideaux
- Department Neuroscience, Cell Biology, and Anatomy, University of Texas Medical Branch (UTMB), Galveston, Texas, USA
| | - Brian N. Finck
- Department of Medicine, Center for Human Nutrition, Washington University in St. Louis, Missouri, USA
| | - Mattia Quattrocelli
- Molecular Cardiovascular Biology, Heart Institute, Cincinnati Children’s Hospital Medical Center (CCHMC) and Department Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| |
Collapse
|
8
|
Wang Y, Guo D, Winkler R, Lei X, Wang X, Messina J, Luo J, Lu H. Development of novel liver-targeting glucocorticoid prodrugs. MEDICINE IN DRUG DISCOVERY 2024; 21:100172. [PMID: 38390434 PMCID: PMC10883687 DOI: 10.1016/j.medidd.2023.100172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/24/2024] Open
Abstract
Background Glucocorticoids (GCs) are widely used in the treatment of inflammatory liver diseases and sepsis, but GC's various side effects on extrahepatic tissues limit their clinical benefits. Liver-targeting GC therapy may have multiple advantages over systemic GC therapy. The purpose of this study was to develop novel liver-targeting GC prodrugs as improved treatment for inflammatory liver diseases and sepsis. Methods A hydrophilic linker or an ultra-hydrophilic zwitterionic linker carboxylic betaine (CB) was used to bridge cholic acid (CA) and dexamethasone (DEX) to generate transporter-dependent liver-targeting GC prodrugs CA-DEX and the highly hydrophilic CA-CB-DEX. The efficacy of liver-targeting DEX prodrugs and DEX were determined in primary human hepatocytes (PHH), macrophages, human whole blood, and/or mice with sepsis induced by cecal ligation and puncture. Results CA-DEX was moderately water soluble, whereas CA-CB-DEX was highly water soluble. CA-CB-DEX and CA-DEX displayed highly transporter-dependent activities in reporter assays. Data mining found marked dysregulation of many GR-target genes important for lipid catabolism, cytoprotection, and inflammation in patients with severe alcoholic hepatitis. These key GR-target genes were similarly and rapidly (within 6 h) induced or down-regulated by CA-CB-DEX and DEX in PHH. CA-CB-DEX had much weaker inhibitory effects than DEX on endotoxin-induced cytokines in mouse macrophages and human whole blood. In contrast, CA-CB-DEX exerted more potent anti-inflammatory effects than DEX in livers of septic mice. Conclusions CA-CB-DEX demonstrated good hepatocyte-selectivity in vitro and better anti-inflammatory effects in vivo. Further test of CA-CB-DEX as a novel liver-targeting GC prodrug for inflammatory liver diseases and sepsis is warranted.
Collapse
Affiliation(s)
- Yazheng Wang
- Department of Pharmacology, SUNY Upstate Medical University, Syracuse, NY 13210, United States
| | - Dandan Guo
- Department of Pharmacology, SUNY Upstate Medical University, Syracuse, NY 13210, United States
| | - Rebecca Winkler
- Department of Pharmacology, SUNY Upstate Medical University, Syracuse, NY 13210, United States
| | - Xiaohong Lei
- Department of Pharmacology, SUNY Upstate Medical University, Syracuse, NY 13210, United States
| | - Xiaojing Wang
- Department of Pharmacology, SUNY Upstate Medical University, Syracuse, NY 13210, United States
| | - Jennifer Messina
- Department of Pharmacology, SUNY Upstate Medical University, Syracuse, NY 13210, United States
| | - Juntao Luo
- Department of Pharmacology, SUNY Upstate Medical University, Syracuse, NY 13210, United States
| | - Hong Lu
- Department of Pharmacology, SUNY Upstate Medical University, Syracuse, NY 13210, United States
| |
Collapse
|
9
|
Rao Y, Su R, Wu C, Yang G, Fu R, Wu J, Liang J, Liu J, Jiang Z, Xu C, Huang L. Marine fungus Aspergillus c1. sp metabolite activates the HSF1/PGC-1α axis, inducing a thermogenic program for treating obesity. Front Pharmacol 2024; 15:1320040. [PMID: 38333010 PMCID: PMC10851286 DOI: 10.3389/fphar.2024.1320040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2023] [Accepted: 01/15/2024] [Indexed: 02/10/2024] Open
Abstract
Background and aims: Obesity is one of the most prevalent diseases worldwide with less ideal approved agents in clinic. Activating the HSF1/PGC-1α axis in adipose tissues has been reported to induce thermogenesis in mice, which presents a promising therapeutic avenue for obesity treatment. The present study aimed to identified novel natural HSF1 activator and evaluated the therapeutic effects of the newly discovered compound on obesity-associated metabolic disorders and the molecular mechanisms of these effects. Methods: Our previous reported HSF1/PGC-1α activator screening system was used to identify novel natural HSF1 activator. The PGC-1α luciferase activity, immunoblot, protein nuclear-translocation, immunofluorescence, chromatin immunoprecipitation assays were used to evaluate the activity of compound HN-001 in activating HSF1. The experiments of mitochondrial number measurement, TG assay and imaging, cellular metabolic assay, gene assays, and CRISPR/Cas 9 were applied for investigating the metabolic effect of HN-001 in C3H10-T1/2 adipocytes. The in vivo anti-obesity efficacies and beneficial metabolic effects of HN-001 were evaluated by performing body and fat mass quantification, plasma chemical analysis, GTT, ITT, cold tolerance test, thermogenesis analysis. Results: HN-001 dose- and time-dependently activated HSF1 and induced HSF1 nuclear translocation, resulting in an enhancement in binding with the gene Pgc-1α. This improvement induced activation of adipose thermogenesis and enhancement of mitochondrial oxidation capacity, thus inhibiting adipocyte maturation. Deletion of HSF1 in adipocytes impaired mitochondrial oxidation and abolished the above beneficial metabolic effects of HN-001, including adipocyte browning induction, improvements in mitogenesis and oxidation capacity, and lipid-lowering ability. In mice, HN-001 treatment efficiently alleviated diet-induced obesity and metabolic disorders. These changes were associated with increased body temperature in mice and activation of the HSF1/PGC-1α axis in adipose tissues. UCP1 expression and mitochondrial biogenesis were increased in both white and brown adipose tissues of HN-001-treated mice. Conclusion: These data indicate that HN-001 may have therapeutic potential for obesity-related metabolic diseases by increasing the capacity of energy expenditure in adipose tissues through a mechanism involving the HSF1/PGC-1α axis, which shed new light on the development of novel anti-obesity agents derived from marine sources.
Collapse
Affiliation(s)
- Yong Rao
- *Correspondence: Yong Rao, ; Ling Huang,
| | | | | | | | | | | | | | | | | | | | - Ling Huang
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Hainan University, Haikou, China
| |
Collapse
|
10
|
Ninfali C, Cortés M, Martínez-Campanario MC, Domínguez V, Han L, Tobías E, Esteve-Codina A, Enrich C, Pintado B, Garrabou G, Postigo A. The adaptive antioxidant response during fasting-induced muscle atrophy is oppositely regulated by ZEB1 and ZEB2. Proc Natl Acad Sci U S A 2023; 120:e2301120120. [PMID: 37948583 PMCID: PMC10655555 DOI: 10.1073/pnas.2301120120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 09/26/2023] [Indexed: 11/12/2023] Open
Abstract
Reactive oxygen species (ROS) serve important homeostatic functions but must be constantly neutralized by an adaptive antioxidant response to prevent supraphysiological levels of ROS from causing oxidative damage to cellular components. Here, we report that the cellular plasticity transcription factors ZEB1 and ZEB2 modulate in opposing directions the adaptive antioxidant response to fasting in skeletal muscle. Using transgenic mice in which Zeb1 or Zeb2 were specifically deleted in skeletal myofibers, we show that in fasted mice, the deletion of Zeb1, but not Zeb2, increased ROS production and that the adaptive antioxidant response to fasting essentially requires ZEB1 and is inhibited by ZEB2. ZEB1 expression increased in fasted muscles and protected them from atrophy; conversely, ZEB2 expression in muscles decreased during fasting and exacerbated muscle atrophy. In fasted muscles, ZEB1 reduces mitochondrial damage and increases mitochondrial respiratory activity; meanwhile, ZEB2 did the opposite. Treatment of fasting mice with Zeb1-deficient myofibers with the antioxidant triterpenoid 1[2-cyano-3,12-dioxool-eana-1,9(11)-dien-28-oyl] trifluoro-ethylamide (CDDO-TFEA) completely reversed their altered phenotype to that observed in fasted control mice. These results set ZEB factors as potential therapeutic targets to modulate the adaptive antioxidant response in physiopathological conditions and diseases caused by redox imbalance.
Collapse
Affiliation(s)
- Chiara Ninfali
- Group of Gene Regulation in Stem Cells, Cell Plasticity, Differentiation, and Cancer, Institute of Biomedical Research August Pi Sunyer (IDIBAPS), Barcelona08036, Spain
| | - Marlies Cortés
- Group of Gene Regulation in Stem Cells, Cell Plasticity, Differentiation, and Cancer, Institute of Biomedical Research August Pi Sunyer (IDIBAPS), Barcelona08036, Spain
| | - M. C. Martínez-Campanario
- Group of Gene Regulation in Stem Cells, Cell Plasticity, Differentiation, and Cancer, Institute of Biomedical Research August Pi Sunyer (IDIBAPS), Barcelona08036, Spain
| | - Verónica Domínguez
- National Center of Biotechnology (CSIC-CNB) and Center for Molecular Biology Severo Ochoa (CSIC-CBMSO), Transgenesis Facility, High Research Council (CSIC) and Autonomous University of Madrid, Cantoblanco, Madrid28049, Spain
| | - Lu Han
- Group of Gene Regulation in Stem Cells, Cell Plasticity, Differentiation, and Cancer, Institute of Biomedical Research August Pi Sunyer (IDIBAPS), Barcelona08036, Spain
| | - Ester Tobías
- Group of Muscle Research and Mitochondrial Function, Institute of Biomedical Research August Pi Sunyer (IDIBAPS), University of Barcelona School of Medicine, Hospital Clínic of Barcelona, and Rare Diseases Networking Biomedical Research Center (CIBERer), Barcelona08036, Spain
| | | | - Carlos Enrich
- Department of Biomedicine, University of Barcelona School of Medicine, and Institute of Biomedical Research August Pi Sunyer (IDIBAPS), Barcelona08036, Spain
| | - Belén Pintado
- National Center of Biotechnology (CSIC-CNB) and Center for Molecular Biology Severo Ochoa (CSIC-CBMSO), Transgenesis Facility, High Research Council (CSIC) and Autonomous University of Madrid, Cantoblanco, Madrid28049, Spain
| | - Gloria Garrabou
- Group of Muscle Research and Mitochondrial Function, Institute of Biomedical Research August Pi Sunyer (IDIBAPS), University of Barcelona School of Medicine, Hospital Clínic of Barcelona, and Rare Diseases Networking Biomedical Research Center (CIBERer), Barcelona08036, Spain
| | - Antonio Postigo
- Group of Gene Regulation in Stem Cells, Cell Plasticity, Differentiation, and Cancer, Institute of Biomedical Research August Pi Sunyer (IDIBAPS), Barcelona08036, Spain
- Molecular Targets Program, Department of Medicine, James Graham Brown Cancer Center, Louisville, KY40202
- Catalan Institution for Research and Advanced Studies (ICREA), Barcelona08010, Spain
| |
Collapse
|
11
|
Fadel L, Dacic M, Fonda V, Sokolsky BA, Quagliarini F, Rogatsky I, Uhlenhaut NH. Modulating glucocorticoid receptor actions in physiology and pathology: Insights from coregulators. Pharmacol Ther 2023; 251:108531. [PMID: 37717739 PMCID: PMC10841922 DOI: 10.1016/j.pharmthera.2023.108531] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Revised: 09/11/2023] [Accepted: 09/13/2023] [Indexed: 09/19/2023]
Abstract
Glucocorticoids (GCs) are a class of steroid hormones that regulate key physiological processes such as metabolism, immune function, and stress responses. The effects of GCs are mediated by the glucocorticoid receptor (GR), a ligand-dependent transcription factor that activates or represses the expression of hundreds to thousands of genes in a tissue- and physiological state-specific manner. The activity of GR is modulated by numerous coregulator proteins that interact with GR in response to different stimuli assembling into a multitude of DNA-protein complexes and facilitate the integration of these signals, helping GR to communicate with basal transcriptional machinery and chromatin. Here, we provide a brief overview of the physiological and molecular functions of GR, and discuss the roles of GR coregulators in the immune system, key metabolic tissues and the central nervous system. We also present an analysis of the GR interactome in different cells and tissues, which suggests tissue-specific utilization of GR coregulators, despite widespread functions shared by some of them.
Collapse
Affiliation(s)
- Lina Fadel
- Institute for Diabetes and Endocrinology IDE, Helmholtz Munich, Ingolstaedter Landstr. 1, 857649 Neuherberg, Germany
| | - Marija Dacic
- Hospital for Special Surgery Research Institute, The David Rosenzweig Genomics Center, New York, NY, USA; Graduate Program in Physiology, Biophysics and Systems Biology, Weill Cornell Graduate School of Medical Sciences, New York, NY, USA
| | - Vlera Fonda
- Institute for Diabetes and Endocrinology IDE, Helmholtz Munich, Ingolstaedter Landstr. 1, 857649 Neuherberg, Germany
| | - Baila A Sokolsky
- Hospital for Special Surgery Research Institute, The David Rosenzweig Genomics Center, New York, NY, USA; Graduate Program in Immunology and Microbial Pathogenesis, Weill Cornell Graduate School of Medical Sciences, New York, NY, USA
| | - Fabiana Quagliarini
- Institute for Diabetes and Endocrinology IDE, Helmholtz Munich, Ingolstaedter Landstr. 1, 857649 Neuherberg, Germany
| | - Inez Rogatsky
- Hospital for Special Surgery Research Institute, The David Rosenzweig Genomics Center, New York, NY, USA; Graduate Program in Immunology and Microbial Pathogenesis, Weill Cornell Graduate School of Medical Sciences, New York, NY, USA.
| | - N Henriette Uhlenhaut
- Institute for Diabetes and Endocrinology IDE, Helmholtz Munich, Ingolstaedter Landstr. 1, 857649 Neuherberg, Germany; Metabolic Programming, TUM School of Life Sciences & ZIEL Institute for Food and Health, Gregor11 Mendel-Str. 2, 85354 Freising, Germany.
| |
Collapse
|
12
|
Prabakaran AD, McFarland K, Miz K, Durumutla HB, Piczer K, El Abdellaoui Soussi F, Latimer H, Werbrich C, Blair NS, Millay DP, Prideaux B, Finck BN, Quattrocelli M. Glucocorticoid intermittence coordinates rescue of energy and mass in aging-related sarcopenia through the myocyte-autonomous PGC1alpha-Lipin1 transactivation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.16.562573. [PMID: 37905062 PMCID: PMC10614926 DOI: 10.1101/2023.10.16.562573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/02/2023]
Abstract
Sarcopenia burdens the elderly population through loss of muscle energy and mass, yet treatments to functionally rescue both parameters are missing. The glucocorticoid prednisone remodels muscle metabolism based on frequency of intake, but its mechanisms in sarcopenia are unknown. We found that once-weekly intermittent prednisone rescued muscle quality in aged 24-month-old mice to levels comparable to young 4-month-old mice. We discovered an age- and sex-independent glucocorticoid receptor transactivation program in muscle encompassing PGC1alpha and its co-factor Lipin1. Treatment coordinately improved mitochondrial abundance through isoform 1 and muscle mass through isoform 4 of the myocyte-specific PGC1alpha, which was required for the treatment-driven increase in carbon shuttling from glucose oxidation to amino acid biogenesis. We also probed the myocyte-specific Lipin1 as non-redundant factor coaxing PGC1alpha upregulation to the stimulation of both oxidative and anabolic capacities. Our study unveils an aging-resistant druggable program in myocytes to coordinately rescue energy and mass in sarcopenia.
Collapse
Affiliation(s)
- Ashok Daniel Prabakaran
- Molecular Cardiovascular Biology, Heart Institute, Cincinnati Children’s Hospital Medical Center and Dept. Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Kevin McFarland
- Molecular Cardiovascular Biology, Heart Institute, Cincinnati Children’s Hospital Medical Center and Dept. Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Karen Miz
- Molecular Cardiovascular Biology, Heart Institute, Cincinnati Children’s Hospital Medical Center and Dept. Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Hima Bindu Durumutla
- Molecular Cardiovascular Biology, Heart Institute, Cincinnati Children’s Hospital Medical Center and Dept. Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Kevin Piczer
- Molecular Cardiovascular Biology, Heart Institute, Cincinnati Children’s Hospital Medical Center and Dept. Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Fadoua El Abdellaoui Soussi
- Molecular Cardiovascular Biology, Heart Institute, Cincinnati Children’s Hospital Medical Center and Dept. Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Hannah Latimer
- Molecular Cardiovascular Biology, Heart Institute, Cincinnati Children’s Hospital Medical Center and Dept. Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Cole Werbrich
- Molecular Cardiovascular Biology, Heart Institute, Cincinnati Children’s Hospital Medical Center and Dept. Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - N. Scott Blair
- Molecular Cardiovascular Biology, Heart Institute, Cincinnati Children’s Hospital Medical Center and Dept. Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Douglas P Millay
- Molecular Cardiovascular Biology, Heart Institute, Cincinnati Children’s Hospital Medical Center and Dept. Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Brendan Prideaux
- Department of Neuroscience, Cell Biology, and Anatomy, University of Texas Medical Branch (UTMB), Galveston, TX, USA
| | - Brian N Finck
- Department of Medicine, Center for Human Nutrition, Washington University in St Louis, MO, USA
| | - Mattia Quattrocelli
- Molecular Cardiovascular Biology, Heart Institute, Cincinnati Children’s Hospital Medical Center and Dept. Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| |
Collapse
|
13
|
Li Q, Jiang X, Zhou Y, Gu Y, Ding Y, Luo J, Pang N, Sun Y, Pei L, Pan J, Gao M, Ma S, Xiao Y, Hu D, Wu F, Yang L. Improving Mitochondrial Function in Skeletal Muscle Contributes to the Amelioration of Insulin Resistance by Nicotinamide Riboside. Int J Mol Sci 2023; 24:10015. [PMID: 37373163 DOI: 10.3390/ijms241210015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 06/01/2023] [Accepted: 06/07/2023] [Indexed: 06/29/2023] Open
Abstract
High-fat diet (HFD)-induced insulin resistance (IR) in skeletal muscle is often accompanied by mitochondrial dysfunction and oxidative stress. Boosting nicotinamide adenine dinucleotide (NAD) using nicotinamide riboside (NR) can effectively decrease oxidative stress and increase mitochondrial function. However, whether NR can ameliorate IR in skeletal muscle is still inconclusive. We fed male C57BL/6J mice with an HFD (60% fat) ± 400 mg/kg·bw NR for 24 weeks. C2C12 myotube cells were treated with 0.25 mM palmitic acid (PA) ± 0.5 mM NR for 24 h. Indicators for IR and mitochondrial dysfunction were analyzed. NR treatment alleviated IR in HFD-fed mice with regard to improved glucose tolerance and a remarkable decrease in the levels of fasting blood glucose, fasting insulin and HOMA-IR index. NR-treated HFD-fed mice also showed improved metabolic status regarding a significant reduction in body weight and lipid contents in serum and the liver. NR activated AMPK in the skeletal muscle of HFD-fed mice and PA-treated C2C12 myotube cells and upregulated the expression of mitochondria-related transcriptional factors and coactivators, thereby improving mitochondrial function and alleviating oxidative stress. Upon inhibiting AMPK using Compound C, NR lost its ability in enhancing mitochondrial function and protection against IR induced by PA. In summary, improving mitochondrial function through the activation of AMPK pathway in skeletal muscle may play an important role in the amelioration of IR using NR.
Collapse
Affiliation(s)
- Qiuyan Li
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Xuye Jiang
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
- Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, Faculty of Health and Medical Sciences, University of Copenhagen, 1172 Copenhagen, Denmark
| | - Yujia Zhou
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Yingying Gu
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Yijie Ding
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Jing Luo
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Nengzhi Pang
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Yan Sun
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Lei Pei
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Jie Pan
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Mengqi Gao
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Sixi Ma
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Ying Xiao
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - De Hu
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Feilong Wu
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Lili Yang
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| |
Collapse
|
14
|
Galasso L, Cappella A, Mulè A, Castelli L, Ciorciari A, Stacchiotti A, Montaruli A. Polyamines and Physical Activity in Musculoskeletal Diseases: A Potential Therapeutic Challenge. Int J Mol Sci 2023; 24:9798. [PMID: 37372945 DOI: 10.3390/ijms24129798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 06/02/2023] [Accepted: 06/04/2023] [Indexed: 06/29/2023] Open
Abstract
Autophagy dysregulation is commonplace in the pathogenesis of several invalidating diseases, such as musculoskeletal diseases. Polyamines, as spermidine and spermine, are small aliphatic cations essential for cell growth and differentiation, with multiple antioxidant, anti-inflammatory, and anti-apoptotic effects. Remarkably, they are emerging as natural autophagy regulators with strong anti-aging effects. Polyamine levels were significantly altered in the skeletal muscles of aged animals. Therefore, supplementation of spermine and spermidine may be important to prevent or treat muscle atrophy. Recent in vitro and in vivo experimental studies indicate that spermidine reverses dysfunctional autophagy and stimulates mitophagy in muscles and heart, preventing senescence. Physical exercise, as polyamines, regulates skeletal muscle mass inducing proper autophagy and mitophagy. This narrative review focuses on the latest evidence regarding the efficacy of polyamines and exercise as autophagy inducers, alone or coupled, in alleviating sarcopenia and aging-dependent musculoskeletal diseases. A comprehensive description of overall autophagic steps in muscle, polyamine metabolic pathways, and effects of the role of autophagy inducers played by both polyamines and exercise has been presented. Although literature shows few data in regard to this controversial topic, interesting effects on muscle atrophy in murine models have emerged when the two "autophagy-inducers" were combined. We hope these findings, with caution, can encourage researchers to continue investigating in this direction. In particular, if these novel insights could be confirmed in further in vivo and clinical studies, and the two synergic treatments could be optimized in terms of dose and duration, then polyamine supplementation and physical exercise might have a clinical potential in sarcopenia, and more importantly, implications for a healthy lifestyle in the elderly population.
Collapse
Affiliation(s)
- Letizia Galasso
- Department of Biomedical Sciences for Health, University of Milan, 20133 Milan, Italy
| | - Annalisa Cappella
- Department of Biomedical Sciences for Health, University of Milan, 20133 Milan, Italy
- U.O. Laboratorio di Morfologia Umana Applicata, I.R.C.C.S. Policlinico San Donato, San Donato Milanese, 20097 Milan, Italy
| | - Antonino Mulè
- Department of Biomedical Sciences for Health, University of Milan, 20133 Milan, Italy
| | - Lucia Castelli
- Department of Biomedical Sciences for Health, University of Milan, 20133 Milan, Italy
| | - Andrea Ciorciari
- Department of Biomedical Sciences for Health, University of Milan, 20133 Milan, Italy
| | - Alessandra Stacchiotti
- Department of Biomedical Sciences for Health, University of Milan, 20133 Milan, Italy
- U.O. Laboratorio di Morfologia Umana Applicata, I.R.C.C.S. Policlinico San Donato, San Donato Milanese, 20097 Milan, Italy
| | - Angela Montaruli
- Department of Biomedical Sciences for Health, University of Milan, 20133 Milan, Italy
- I.R.C.C.S. Ospedale Galeazzi-Sant'Ambrogio, 20157 Milan, Italy
| |
Collapse
|
15
|
Durumutla HB, Villa C, Panta M, Wintzinger M, Pragasam ADP, Miz K, Quattrocelli M. Comprehensive Analyses of Muscle Function, Lean and Muscle Mass, and Myofiber Typing in Mice. Bio Protoc 2023; 13:e4617. [PMID: 36845536 PMCID: PMC9947547 DOI: 10.21769/bioprotoc.4617] [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: 07/22/2022] [Revised: 11/15/2022] [Accepted: 01/23/2023] [Indexed: 02/17/2023] Open
Abstract
Skeletal muscle disorders commonly affect the function and integrity of muscles. Novel interventions bring new potential to rescue or alleviate the symptoms associated with these disorders. In vivo and in vitro testing in mouse models allows quantitative evaluation of the degree of muscle dysfunction, and therefore, the level of potential rescue/restoration by the target intervention. Several resources and methods are available to assess muscle function and lean and muscle mass, as well as myofiber typing as separate concepts; however, a technical resource unifying these methods is missing. Here, we provide detailed procedures for analyzing muscle function, lean and muscle mass, and myofiber typing in a comprehensive technical resource paper. Graphical abstract.
Collapse
Affiliation(s)
- Hima Bindu Durumutla
- Division of Molecular Cardiovascular Biology, Heart Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Chiara Villa
- Division of Molecular Cardiovascular Biology, Heart Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Stem Cell laboratory, Dino Ferrari Center, Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Manoj Panta
- Division of Molecular Cardiovascular Biology, Heart Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Michelle Wintzinger
- Division of Molecular Cardiovascular Biology, Heart Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Ashok Daniel Prabakaran Pragasam
- Division of Molecular Cardiovascular Biology, Heart Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Karen Miz
- Division of Molecular Cardiovascular Biology, Heart Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Mattia Quattrocelli
- Division of Molecular Cardiovascular Biology, Heart Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| |
Collapse
|
16
|
Yan Y, Li M, Lin J, Ji Y, Wang K, Yan D, Shen Y, Wang W, Huang Z, Jiang H, Sun H, Qi L. Adenosine monophosphate activated protein kinase contributes to skeletal muscle health through the control of mitochondrial function. Front Pharmacol 2022; 13:947387. [PMID: 36339617 PMCID: PMC9632297 DOI: 10.3389/fphar.2022.947387] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 10/06/2022] [Indexed: 11/26/2022] Open
Abstract
Skeletal muscle is one of the largest organs in the body and the largest protein repository. Mitochondria are the main energy-producing organelles in cells and play an important role in skeletal muscle health and function. They participate in several biological processes related to skeletal muscle metabolism, growth, and regeneration. Adenosine monophosphate-activated protein kinase (AMPK) is a metabolic sensor and regulator of systemic energy balance. AMPK is involved in the control of energy metabolism by regulating many downstream targets. In this review, we propose that AMPK directly controls several facets of mitochondrial function, which in turn controls skeletal muscle metabolism and health. This review is divided into four parts. First, we summarize the properties of AMPK signal transduction and its upstream activators. Second, we discuss the role of mitochondria in myogenesis, muscle atrophy, regeneration post-injury of skeletal muscle cells. Third, we elaborate the effects of AMPK on mitochondrial biogenesis, fusion, fission and mitochondrial autophagy, and discuss how AMPK regulates the metabolism of skeletal muscle by regulating mitochondrial function. Finally, we discuss the effects of AMPK activators on muscle disease status. This review thus represents a foundation for understanding this biological process of mitochondrial dynamics regulated by AMPK in the metabolism of skeletal muscle. A better understanding of the role of AMPK on mitochondrial dynamic is essential to improve mitochondrial function, and hence promote skeletal muscle health and function.
Collapse
Affiliation(s)
- Yan Yan
- Department of Emergency Medicine, Affiliated Hospital of Nantong University, Nantong, China
- 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, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Nantong University, Nantong, China
| | - Ming Li
- Department of Laboratory Medicine, Binhai County People’s Hospital Affiliated to Kangda College of Nanjing Medical University, Yancheng, China
| | - Jie Lin
- Department of Infectious Disease, Affiliated Hospital of Nantong University, Nantong, 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, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Nantong University, Nantong, 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, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Nantong University, Nantong, China
| | - Dajun Yan
- Department of Emergency Medicine, Affiliated Hospital of Nantong University, Nantong, 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, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Nantong University, Nantong, China
| | - Wei Wang
- Department of Emergency Medicine, Affiliated Hospital of Nantong University, Nantong, China
- Department of Pathology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China
| | - Zhongwei Huang
- Department of Emergency Medicine, Affiliated Hospital of Nantong University, Nantong, China
| | - Haiyan Jiang
- Department of Emergency Medicine, Affiliated Hospital of Nantong University, Nantong, China
- *Correspondence: Haiyan Jiang, ; Hualin Sun, ; Lei Qi,
| | - 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, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Nantong University, Nantong, China
- *Correspondence: Haiyan Jiang, ; Hualin Sun, ; Lei Qi,
| | - Lei Qi
- Department of Emergency Medicine, Affiliated Hospital of Nantong University, Nantong, China
- *Correspondence: Haiyan Jiang, ; Hualin Sun, ; Lei Qi,
| |
Collapse
|
17
|
Narrative Review: Glucocorticoids in Alcoholic Hepatitis—Benefits, Side Effects, and Mechanisms. J Xenobiot 2022; 12:266-288. [PMID: 36278756 PMCID: PMC9589945 DOI: 10.3390/jox12040019] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 09/03/2022] [Accepted: 09/05/2022] [Indexed: 11/17/2022] Open
Abstract
Alcoholic hepatitis is a major health and economic burden worldwide. Glucocorticoids (GCs) are the only first-line drugs recommended to treat severe alcoholic hepatitis (sAH), with limited short-term efficacy and significant side effects. In this review, I summarize the major benefits and side effects of GC therapy in sAH and the potential underlying mechanisms. The review of the literature and data mining clearly indicate that the hepatic signaling of glucocorticoid receptor (GR) is markedly impaired in sAH patients. The impaired GR signaling causes hepatic down-regulation of genes essential for gluconeogenesis, lipid catabolism, cytoprotection, and anti-inflammation in sAH patients. The efficacy of GCs in sAH may be compromised by GC resistance and/or GC’s extrahepatic side effects, particularly the side effects of intestinal epithelial GR on gut permeability and inflammation in AH. Prednisolone, a major GC used for sAH, activates both the GR and mineralocorticoid receptor (MR). When GC non-responsiveness occurs in sAH patients, the activation of MR by prednisolone might increase the risk of alcohol abuse, liver fibrosis, and acute kidney injury. To improve the GC therapy of sAH, the effort should be focused on developing the biomarker(s) for GC responsiveness, liver-targeting GR agonists, and strategies to overcome GC non-responsiveness and prevent alcohol relapse in sAH patients.
Collapse
|
18
|
Jang J, Koh JH, Kim Y, Kim HJ, Park S, Chang Y, Jung J, Wolfe RR, Kim IY. Balanced Free Essential Amino Acids and Resistance Exercise Training Synergistically Improve Dexamethasone-Induced Impairments in Muscle Strength, Endurance, and Insulin Sensitivity in Mice. Int J Mol Sci 2022; 23:ijms23179735. [PMID: 36077132 PMCID: PMC9456044 DOI: 10.3390/ijms23179735] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 08/23/2022] [Accepted: 08/25/2022] [Indexed: 11/16/2022] Open
Abstract
Our previous study shows that an essential amino acid (EAA)-enriched diet attenuates dexamethasone (DEX)-induced declines in muscle mass and strength, as well as insulin sensitivity, but does not affect endurance. In the present study, we hypothesized that the beneficial effects will be synergized by adding resistance exercise training (RET) to EAA, and diet-free EAA would improve endurance. To test hypotheses, mice were randomized into the following four groups: control, EAA, RET, and EAA+RET. All mice except the control were subjected to DEX treatment. We evaluated the cumulative rate of myofibrillar protein synthesis (MPS) using 2H2O labeling and mass spectrometry. Neuromuscular junction (NMJ) stability, mitochondrial contents, and molecular signaling were demonstrated in skeletal muscle. Insulin sensitivity and glucose metabolism using 13C6-glucose tracing during oral glucose tolerance tests were analyzed. We found that EAA and RET synergistically improve muscle mass and/or strength, and endurance capacity, as well as insulin sensitivity, and glucose metabolism in DEX-treated muscle. These improvements are accomplished, in part, through improvements in myofibrillar protein synthesis, NMJ, fiber type preservation, and/or mitochondrial biogenesis. In conclusion, free EAA supplementation, particularly when combined with RET, can serve as an effective means that counteracts the adverse effects on muscle of DEX that are found frequently in clinical settings.
Collapse
Affiliation(s)
- Jiwoong Jang
- Korea Mouse Metabolic Phenotyping Center, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon 21999, Korea
- Department of Internal Medicine, Gil Medical Center, Gachon University, Incheon 21565, Korea
| | - Jin-Ho Koh
- Department of Molecular Medicine, College of Medicine, Gachon University, Incheon 21565, Korea
| | - Yeongmin Kim
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon 21999, Korea
| | - Hee-Joo Kim
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon 21999, Korea
| | - Sanghee Park
- Department of Internal Medicine, Gil Medical Center, Gachon University, Incheon 21565, Korea
- Department of Molecular Medicine, College of Medicine, Gachon University, Incheon 21565, Korea
| | - Yewon Chang
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon 21999, Korea
| | - Jiyeon Jung
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon 21999, Korea
| | - Robert R. Wolfe
- Department of Geriatrics, Center for Translational Research in Aging and Longevity, Donald W. Reynolds Institute on Aging, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Il-Young Kim
- Korea Mouse Metabolic Phenotyping Center, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon 21999, Korea
- Department of Molecular Medicine, College of Medicine, Gachon University, Incheon 21565, Korea
- Correspondence: ; Tel.: +82-32-899-6685
| |
Collapse
|
19
|
Wintzinger M, Panta M, Miz K, Prabakaran AD, Durumutla HB, Sargent M, Peek CB, Bass J, Molkentin JD, Quattrocelli M. Impact of circadian time of dosing on cardiomyocyte-autonomous effects of glucocorticoids. Mol Metab 2022; 62:101528. [PMID: 35717025 PMCID: PMC9243158 DOI: 10.1016/j.molmet.2022.101528] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 05/27/2022] [Accepted: 06/11/2022] [Indexed: 11/29/2022] Open
Abstract
OBJECTIVE Mitochondrial capacity is critical to adapt the high energy demand of the heart to circadian oscillations and diseased states. Glucocorticoids regulate the circadian cycle of energy metabolism, but little is known about how circadian timing of exogenous glucocorticoid dosing directly regulates heart metabolism through cardiomyocyte-autonomous mechanisms. While chronic once-daily intake of glucocorticoids promotes metabolic stress and heart failure, we recently discovered that intermittent once-weekly dosing of exogenous glucocorticoids promoted muscle metabolism in normal and obese skeletal muscle. However, the effects of glucocorticoid intermittence on heart metabolism and heart failure remain unknown. Here we investigated the extent to which circadian time of dosing regulates the effects of the glucocorticoid prednisone in heart metabolism and function in conditions of single pulse or chronic intermittent dosing. METHODS AND RESULTS In WT mice, we found that prednisone improved cardiac content of NAD+ and ATP with light-phase dosing (ZT0), while the effects were blocked by dark-phase dosing (ZT12). The drug effects on mitochondrial function were cardiomyocyte-autonomous, as shown by inducible cardiomyocyte-restricted glucocorticoid receptor (GR) ablation, and depended on an intact cardiomyocyte clock, as shown by inducible cardiomyocyte-restricted ablation of Brain and Muscle ARNT-like 1 (BMAL1). Conjugating time-of-dosing with chronic intermittence, we found that once-weekly prednisone improved metabolism and function in heart after myocardial injury dependent on circadian time of intake, i.e. with light-phase but not dark-phase dosing. CONCLUSIONS Our study identifies cardiac-autonomous mechanisms through which circadian-specific intermittent dosing reconverts glucocorticoid drugs to metabolic boosters for the heart.
Collapse
Affiliation(s)
- Michelle Wintzinger
- Division of Molecular Cardiovascular Biology, Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Manoj Panta
- Division of Molecular Cardiovascular Biology, Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Karen Miz
- Division of Molecular Cardiovascular Biology, Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Ashok D Prabakaran
- Division of Molecular Cardiovascular Biology, Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Hima Bindu Durumutla
- Division of Molecular Cardiovascular Biology, Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA; Department of Pediatrics, University of Cincinnati College of Medicine; Cincinnati, OH, USA
| | - Michelle Sargent
- Division of Molecular Cardiovascular Biology, Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Clara Bien Peek
- Division of Endocrinology, Metabolism and Molecular Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA; Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Joseph Bass
- Division of Endocrinology, Metabolism and Molecular Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Jeffery D Molkentin
- Division of Molecular Cardiovascular Biology, Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA; Department of Pediatrics, University of Cincinnati College of Medicine; Cincinnati, OH, USA
| | - Mattia Quattrocelli
- Division of Molecular Cardiovascular Biology, Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA; Department of Pediatrics, University of Cincinnati College of Medicine; Cincinnati, OH, USA.
| |
Collapse
|