1
|
McLain A, Kowalczyk A, Baran-Rachwalska P, Sutera FM, Robertson LJ, Nielsen NS, Enghild JJ, Cobice D, Bonelli F, Barbaro V, Ferrari S, Patterson B, Moore L, Marshall J, Nesbit MA, Moore T. TGFBI R124H mutant allele silencing in granular corneal dystrophy type 2 using topical siRNA delivery. J Control Release 2025; 382:113681. [PMID: 40185334 DOI: 10.1016/j.jconrel.2025.113681] [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: 12/24/2024] [Revised: 03/17/2025] [Accepted: 03/31/2025] [Indexed: 04/07/2025]
Abstract
In recent years, success has been achieved in treating several eye conditions with oligonucleotide-based therapies. Herein, we outline the experimentation involved in progressing selection and development of a lead therapeutic siRNA for R124H mutation of TGFBI gene which causes Granular Corneal Dystrophy Type 2 (GCD2/Avellino CD). Firstly, a series of siRNA designs, generated by a gene walk across the R124H TGFBI mutation site, were tested and a lead siRNA identified. The lead siRNA was delivered into an immortalised human corneal epithelial cell line to assess on-target efficacy and off-target effects. The in vivo efficacy of the lead R124H TGFBI siRNA, complexed with Bio-Courier technology, silicon stabilized hybrid lipid nanoparticles (sshLNP), was assessed in a mouse model of GCD2 which expressed the human R124H TGFBI transgene. Following topical siRNA application for 5 consecutive days, expression of the R124H mutant TGFBI transgene was measured and shown to be reduced by 22.4 % (± 15.7 %, p < 0.05). We investigated gene expression in the mouse cornea and showed expression of murine Tgfbi was 20-fold lower than TGFBI in human cornea, and expression of the mutant TGFBI transgene was a further 3-fold lower. This estimated 60-fold lower mutant transgene expression may explain the low frequency of corneal deposits observed in this mouse model, limiting its usefulness to test whether siRNA silencing is capable of phenotypic improvement or regression of GCD2/Avellino corneal dystrophy. We assessed WT TGFBI silencing in human primary corneal epithelial cells (PCEC) derived from human corneal limbal biopsy material, which express TGFBI at a similar level to human corneal biopsy. We demonstrated that a single 100 nM siRNA treatment, delivered by the sshLNP to the primary human corneal epithelial cells, gave 26.6 % (± 6.6 %, p < 0.001) reduction in TGFBI mRNA and a 15.4 % (±10.5 %, p < 0.05 %) reduction in TGFBi protein after 48 h. In consideration of the mutant gene expression levels in existing models of GCD2 disease, an ex vivo model of mutation-expressing primary corneal epithelial cells generated from corneal limbal biopsies from GCD2 patients would be more suitable than existing transgenic mouse models for future pre-clinical work in the development of gene silencing therapies for corneal dystrophies.
Collapse
Affiliation(s)
- Andrew McLain
- Integrated Diagnostics Laboratory, Northland House, CDHT, Frederick Street, Ulster University, Belfast, UK
| | - Amanda Kowalczyk
- Integrated Diagnostics Laboratory, Northland House, CDHT, Frederick Street, Ulster University, Belfast, UK
| | | | | | - Louise J Robertson
- Integrated Diagnostics Laboratory, Northland House, CDHT, Frederick Street, Ulster University, Belfast, UK
| | - Nadia Sukusu Nielsen
- Department of Molecular Biology and Genetics, Aarhus University, DK-8000 Aarhus C, Denmark
| | - Jan J Enghild
- Department of Molecular Biology and Genetics, Aarhus University, DK-8000 Aarhus C, Denmark
| | - Diego Cobice
- Integrated Diagnostics Laboratory, Northland House, CDHT, Frederick Street, Ulster University, Belfast, UK
| | - Filippo Bonelli
- Fondazione Banca degli Occhi del Veneto, Via Paccagnella 11, 30174 Venice, Italy
| | - Vanessa Barbaro
- Fondazione Banca degli Occhi del Veneto, Via Paccagnella 11, 30174 Venice, Italy
| | - Stefano Ferrari
- Fondazione Banca degli Occhi del Veneto, Via Paccagnella 11, 30174 Venice, Italy
| | - Benjamin Patterson
- Integrated Diagnostics Laboratory, Northland House, CDHT, Frederick Street, Ulster University, Belfast, UK
| | - Luca Moore
- LM Lassi Ltd, 22 Great Victoria Street, Belfast, Northern Ireland BT2 7BA, UK; University of York, Heslington, York YO10 5DD, UK
| | - John Marshall
- University College London, Institute of Ophthalmology, 11-43 Bath Street, London EC1V 9EL, UK
| | - M Andrew Nesbit
- Integrated Diagnostics Laboratory, Northland House, CDHT, Frederick Street, Ulster University, Belfast, UK
| | - Tara Moore
- Integrated Diagnostics Laboratory, Northland House, CDHT, Frederick Street, Ulster University, Belfast, UK; LM Lassi Ltd, 22 Great Victoria Street, Belfast, Northern Ireland BT2 7BA, UK; Avellino USA, Menlo Park, 4300 Bohannon Drive, Menlo Park, CA 94025, USA.
| |
Collapse
|
2
|
Bokov RO, Sharlo KA, Vilchinskaya NA, Tyganov SA, Turtikova OV, Rozhkov SV, Deviatiiarov RM, Gusev OA, Tomilovskaya ES, Shenkman BS, Orlov OI. Molecular insights into human soleus muscle atrophy development: long-term dry immersion effects on the transcriptomic profile and posttranslational signaling. Physiol Genomics 2025; 57:357-382. [PMID: 40072920 DOI: 10.1152/physiolgenomics.00196.2024] [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: 12/31/2024] [Revised: 01/27/2025] [Accepted: 03/06/2025] [Indexed: 03/14/2025] Open
Abstract
Muscle disuse results in complex signaling alterations followed by structural and functional changes, such as atrophy, force decrease, and slow-to-fast fiber-type shift. Little is known about human skeletal muscle signaling alterations under long-term muscle disuse. In this study, we describe the effects of 21-day dry immersion on human postural soleus muscle. We performed both transcriptomic analysis and Western blots to describe the states of the key signaling pathways regulating soleus muscle fiber size, fiber type, and metabolism. Twenty-one-day dry immersion resulted in both slow-type and fast-type myofibers atrophy, downregulation of rRNA content, and mTOR signaling. Twenty-one-day dry immersion also leads to slow-to-fast fiber-type and gene expression shift, upregulation of p-eEF2, p-CaMKII, p-ACC content and downregulation of NFATc1 nuclear content. It also caused massive gene expression alterations associated with calcium signaling, cytoskeletal parameters, and downregulated mitochondrial signaling (including fusion, fission, and marker of mitochondrial density).NEW & NOTEWORTHY The main findings of our study are as follows: 1) The soleus slow fibers atrophy after 21-day dry immersion (DI) does not exceed that after 7-day DI; 2) The soleus ubiquitin ligases expression after 21-day DI returns to its initial level; 3) The soleus slow fibers atrophy after 21-day DI is accompanied by a mitochondrial apparatus structural markers decrease; 4) The soleus fibers signaling pathways restructuring process during 21-day DI is carried out in a complex manner.
Collapse
Affiliation(s)
- Roman O Bokov
- Institute of Biomedical Problems, Russian Academy of Sciences, Moscow, Russia
| | - Kristina A Sharlo
- Institute of Biomedical Problems, Russian Academy of Sciences, Moscow, Russia
| | | | - Sergey A Tyganov
- Institute of Biomedical Problems, Russian Academy of Sciences, Moscow, Russia
| | - Olga V Turtikova
- Institute of Biomedical Problems, Russian Academy of Sciences, Moscow, Russia
| | - Sergey V Rozhkov
- Institute of Biomedical Problems, Russian Academy of Sciences, Moscow, Russia
| | | | - Oleg A Gusev
- Life Improvement by Future Technologies Center, Moscow, Russia
| | | | - Boris S Shenkman
- Institute of Biomedical Problems, Russian Academy of Sciences, Moscow, Russia
| | - Oleg I Orlov
- Institute of Biomedical Problems, Russian Academy of Sciences, Moscow, Russia
| |
Collapse
|
3
|
Huang X, Li S, Wang H, Zhao L, Li X, Fan S, Hu W, Tong H, Guo G, Xu D, Zhang L, Jiang Z, Yu Q. Oestrogen Receptor Alpha in Myocyte Maintains Muscle Regeneration in Duchenne Muscular Dystrophy. J Cachexia Sarcopenia Muscle 2025; 16:e13807. [PMID: 40258782 PMCID: PMC12011492 DOI: 10.1002/jcsm.13807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 03/11/2025] [Accepted: 03/17/2025] [Indexed: 04/23/2025] Open
Abstract
BACKGROUND Oestrogen receptor alpha (ERα) plays an important role in maintaining mitochondrial function and regulating metabolism in skeletal muscle. However, its alterations and potential mechanisms in Duchenne muscular dystrophy (DMD) remain incompletely understood. In this study, we demonstrated the protective role of ERα in myocyte for skeletal muscle regeneration in mdx mice and explored the therapeutic effects of oestrogen receptor modulators on DMD. METHODS DMD patients' biopsies were obtained for histological analysis to explore the expression of ERα. The phenotype of muscle was analysed by histology and molecular biology. The therapeutical effect of different oestrogen receptor modulators was examined in mdx mice treated with fulvestrant (FVT, 20 mg/kg once a week) or oestradiol (E2, 1 mg/kg per day) for 4 weeks. The protective effect of ERα was performed on mdx mice after conditional knockout of ERα in skeletal muscle (ERαmKO mdx mice). Evidence of activation of ERα/oestrogen-related receptor alpha (ERRα)/myogenic differentiation 1 (MyoD) signalling pathway was inspected in the primary myoblasts isolated from mice, and C2C12 cells received intervention with E2/FVT/Esr1-siRNA/Esrra overexpression plasmid. RESULTS The ERα expression was increased in DMD patients' triceps (p < 0.05) and mdx mice muscles (p < 0.05). FVT reduced ERα levels in the mdx mice muscles (p < 0.01) but had no significant effect on skeletal muscle regeneration on mdx mice. Compared with mdx mice, E2 reduced the levels of creatine kinase (CK) and lactic dehydrogenase (LDH) (p < 0.001) in serum, enhanced skeletal muscle function, alleviated skeletal muscle atrophy and fibre loss and upregulated the expression of ERα in GAS (p < 0.001) and TA (p < 0.05). The myogenic factors such as myosin heavy chain (MyHC, p < 0.001), myogenin (MyoG, p < 0.05), MyoD (p < 0.05) and ERRα (p < 0.001) were increased in mdx mice GAS with E2. But E2 had no effect on ERαmKO mdx mice. The primary myoblasts and C2C12 were treated with E2 displayed an increased-on myocyte fusion index (p < 0.05), ERα MyoD and ERRα expressions (p < 0.05). The myocytes' fusion index (p < 0.05) and ERα, MyoD and ERRα expression (p < 0.05) were decreased in si-Esr1-transfected C2C12 cells and increased in OE-Esrra-transfected C2C12 cells. CONCLUSION We demonstrated that ERα in myocyte exerted a protective effect on skeletal muscle regeneration in DMD patients and mdx mice through the ERα-ERRα-MyoD pathway, which has potential implications for DMD therapy strategies.
Collapse
Affiliation(s)
- Xiaofei Huang
- New Drug Screening and Pharmacodynamics Evaluation CenterChina Pharmaceutical UniversityNanjingChina
| | - Sijia Li
- New Drug Screening and Pharmacodynamics Evaluation CenterChina Pharmaceutical UniversityNanjingChina
| | - Huna Wang
- New Drug Screening and Pharmacodynamics Evaluation CenterChina Pharmaceutical UniversityNanjingChina
| | - Lei Zhao
- Department of Neurology Children'sHospital of Fudan UniversityShanghaiChina
| | - Xihua Li
- Department of Neurology Children'sHospital of Fudan UniversityShanghaiChina
| | - Shusheng Fan
- New Drug Screening and Pharmacodynamics Evaluation CenterChina Pharmaceutical UniversityNanjingChina
| | - Wanting Hu
- New Drug Screening and Pharmacodynamics Evaluation CenterChina Pharmaceutical UniversityNanjingChina
| | - Haowei Tong
- New Drug Screening and Pharmacodynamics Evaluation CenterChina Pharmaceutical UniversityNanjingChina
| | - Guangyao Guo
- New Drug Screening and Pharmacodynamics Evaluation CenterChina Pharmaceutical UniversityNanjingChina
| | - Dengqiu Xu
- Department of Hepatobiliary Surgery Innovative Institute of Tumor Immunity and Medicine (ITIM), Anhui Province key Laboratory of Tumor Immune Microenvironment and ImmunotherapyThe First Affiliated Hospital of Anhui Medical UniversityHefeiChina
| | - Luyong Zhang
- New Drug Screening and Pharmacodynamics Evaluation CenterChina Pharmaceutical UniversityNanjingChina
- Center for Drug Research and DevelopmentGuangdong Pharmaceutical UniversityGuangzhouChina
| | - Zhenzhou Jiang
- New Drug Screening and Pharmacodynamics Evaluation CenterChina Pharmaceutical UniversityNanjingChina
- Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of EducationChina Pharmaceutical UniversityNanjingChina
| | - Qinwei Yu
- New Drug Screening and Pharmacodynamics Evaluation CenterChina Pharmaceutical UniversityNanjingChina
| |
Collapse
|
4
|
Fox SN, Savage CH, Amireddy NR, McMeekin LJ, Crossman DK, Detloff PJ, Gray M, Cowell RM. Estrogen-related receptor gamma is a regulator of mitochondrial, autophagy, and immediate-early gene programs in spiny projection neurons: Relevance for transcriptional changes in Huntington disease. Neurobiol Dis 2025; 206:106818. [PMID: 39884587 DOI: 10.1016/j.nbd.2025.106818] [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: 10/25/2024] [Revised: 01/16/2025] [Accepted: 01/27/2025] [Indexed: 02/01/2025] Open
Abstract
Mitochondrial dysfunction, transcriptional dysregulation, and protein aggregation are hallmarks of multiple neurodegenerative disorders, including Huntington's disease (HD). Strategies are needed to counteract these processes to restore neuronal health and function in HD. Recent evidence indicates that the transcription factor estrogen-related receptor gamma (ERRγ/Esrrg) is required for normal expression of mitochondrial, synaptic, and autophagy genes in neurons. Further, overexpression of Esrrg in dopaminergic neurons reduces synuclein load in the pre-formed fibril model of synucleinopathy. For these reasons, we sought to understand ERRγ's role in transcriptional regulation in spiny projection neurons (SPNs), one of the neuronal populations vulnerable to transcriptional dysregulation, mitochondrial dysfunction, and protein aggregation in HD. Here, we demonstrate that developmental deletion of Esrrg selectively in SPNs causes a transcriptional pattern consistent with a reduction of Drd1 and Drd2-positive neurons in the mouse dorsolateral striatum. To avoid effects of developmental deletion and explore Esrrg's role within adult SPN populations, we deleted or overexpressed Esrrg in adult SPNs. While overexpression was sufficient to increase the expression of mitochondrial and lysosome-related transcripts, Esrrg deletion surprisingly caused increased expression of immediate-early genes and genes with enrichment of binding sites for transcriptional repressors. In contrast, these genes were downregulated by Esrrg overexpression. Concordantly, Esrrg-deficient mice exhibited lack of amphetamine-induced hyperactivity and further upregulation of immediate-early genes. To determine whether the alterations observed with ERRγ modulation have any relevance for understanding transcriptional changes in SPNs in neurodegeneration, we measured Esrrg and its responsive genes in two mouse models of HD. We found an increase in Esrrg expression in HD models, accompanied by a transcriptional profile with similarities to that observed with Esrrg overexpression, suggesting the existence of an ERRγ-dependent, stress-related response. Altogether, these studies suggest that ERRγ is a key activator of mitochondrial and lysosomal transcripts in SPNs with a potential bi-functional role as a mediator of immediate-early gene repression. Ongoing studies are investigating mechanisms underlying ERRγ's roles in transcriptional activation and repression in SPNs to inform strategies to promote neuroprotective actions of ERRγ in SPNs in HD.
Collapse
Affiliation(s)
- Stephanie N Fox
- Department of Neurology and Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, Birmingham, AL 35294, USA; Southern Research, Birmingham, AL 35205, USA.
| | - Cody H Savage
- Department of Neurology and Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, Birmingham, AL 35294, USA; Southern Research, Birmingham, AL 35205, USA
| | - Narcy R Amireddy
- Department of Neurology and Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| | | | - David K Crossman
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| | - Peter J Detloff
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Michelle Gray
- Department of Neurology and Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| | - Rita M Cowell
- Department of Neurology and Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, Birmingham, AL 35294, USA; Southern Research, Birmingham, AL 35205, USA.
| |
Collapse
|
5
|
Ai Z, Yuan D, Dong R, Zhou S, Cao J. Rostellularia procumbens (L) Nees. extract attenuates adriamycin-induced nephropathy by maintaining mitochondrial dynamics balance via SIRT1/PGC-1α signaling pathway activation. JOURNAL OF ETHNOPHARMACOLOGY 2025; 340:119297. [PMID: 39733803 DOI: 10.1016/j.jep.2024.119297] [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: 07/28/2024] [Revised: 12/03/2024] [Accepted: 12/26/2024] [Indexed: 12/31/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Rostellularia procumbens (L) Nees. (R. procumbens) is a classical Chinese herbal medicine that has been used for effective treatment of kidney disease for nearly a thousand years in China. Recently, significant progress has been achieved in understanding the abnormal mitochondrial structure and function from chronic kidney disease (CKD). However, the regulatory mechanisms underlying R. procumbens treatment for CKD and its association with dysfunctional mitochondrial function remain elusive. AIM OF THE STUDY To study the protective effect of N-butanol extract from R. procumbens (J-NE) on chronic glomerulonephritis (CGN) mice using a mice model and mitochondrial function-related experiments. MATERIALS AND METHODS A renal injury mouse model was developed using a single tail vein injection of adriamycin (9 mg/kg). Renal pathology was analyzed through hematoxylin-eosin (HE) staining and transmission electron microscopy (TEM). Cell apoptosis in kidney tissues was analyzed using TUNEL staining. Protein levels were measured via immunohistochemistry (HIF-1α, FN, α-SMA, and Collagen I) and Western blot (Mn-SOD, p-Drp-S637, MFN1, MFN2, OPA1, TFAM, Nrf1, ATP6, SIRT1, and PGC-1α) analysis. UHPLC-MS/MS was used to analyze the presence of bioactive phytocompounds in J-NE. RESULTS The results reported that the levels of kidney injury markers (urinary protein, glomerular atrophy, and renal cell apoptosis), mitochondrial dysfunction markers (mitochondrial ultrastructure, Mn-SOD, HIF-1α, FN and α-SMA),mitochondrial dynamic imbalance markers (p-Drp-S637, MFN1, MFN2 and OPA1) and SIRT1/PGC-1α signaling pathway markers (TFAM, Nrf1, ATP6, SIRT1, and PGC-1α) were settled to a significant improvement by the oral administration of J-NE. CONCLUSIONS In conclusion, R. procumbens could be able to protect the kidneys from podocyte injury caused mitochondrial dynamics and energy metabolism dysregulation by modulating the SIRT1/PGC-1α signaling pathway.
Collapse
Affiliation(s)
- Zhongzhu Ai
- Faculty of Pharmacy, Hubei University of Chinese Medicine, Wuhan, 430065, China
| | - Dongfeng Yuan
- Research Center for Computer-aided Drug Discovery, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Ruotong Dong
- Faculty of Pharmacy, Hubei University of Chinese Medicine, Wuhan, 430065, China
| | - Shanshan Zhou
- The First Clinical Medical School, Hubei University of Chinese Medicine, Wuhan, 430065, China.
| | - Jigang Cao
- School of Basic Medical Science, Hubei University of Chinese Medicine, Wuhan, 430065, China.
| |
Collapse
|
6
|
Sreedevi K, James A, Do S, Yedla S, Arowa S, Oka SI, Wende AR, Zaitsev AV, Warren JS. PERM1 regulates mitochondrial energetics through O-GlcNAcylation in the heart. J Mol Cell Cardiol 2025; 198:1-12. [PMID: 39581161 DOI: 10.1016/j.yjmcc.2024.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 11/07/2024] [Accepted: 11/14/2024] [Indexed: 11/26/2024]
Abstract
PERM1 was initially identified as a new downstream target of PGC-1α and ERRs that regulates mitochondrial bioenergetics in skeletal muscle. Subsequently, we and other groups demonstrated that PERM1 is also a positive regulator of mitochondrial bioenergetics in the heart. However, the exact mechanisms of regulatory functions of PERM1 remain poorly understood. O-GlcNAcylation is a post-translational modification of proteins that are regulated by two enzymes: O-GlcNAc transferase (OGT) that adds O-GlcNAc to proteins; O-GlcNAcase (OGA) that removes O-GlcNAc from proteins. O-GlcNAcylation is a powerful signaling mechanism mediating cellular responses to stressors and nutrient availability, which, among other targets, may influence cardiac metabolism. We hypothesized that PERM1 regulates mitochondrial energetics in cardiomyocytes through modulation of O-GlcNAcylation. We found that overexpression of PERM1 decreased the total levels of O-GlcNAcylated proteins, concomitant with decreased OGT and increased OGA expression levels. Luciferase gene reporter assay showed that PERM1 significantly decreases the promoter activity of Ogt without changing the promoter activity of Oga. The downregulation of OGT by PERM1 overexpression was mediated through its interaction with E2F1, a known transcription repressor of Ogt. A deliberate increase of O-GlcNAcylation through Oga silencing in cardiomyocytes decreased the basal and maximal mitochondrial respiration and ATP production rates, all of which were completely restored by PERM1 overexpression. Furthermore, excessive O-GlcNAcylation caused by the loss of PERM1 led to the increase of O-GlcNAcylated PGC-1α, a master regulator of mitochondrial bioenergetics, concurrent with the dissociation of PGC-1α from PPARα, a well-known transcription factor that regulates fatty acid β-oxidation. We conclude that PERM1 positively regulates mitochondrial energetics, in part, via suppressing O-GlcNAcylation in cardiac myocytes.
Collapse
Affiliation(s)
- Karthi Sreedevi
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, VA, USA
| | - Amina James
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, VA, USA
| | - Sara Do
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, VA, USA
| | - Shreya Yedla
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, VA, USA
| | - Sumaita Arowa
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, VA, USA
| | - Shin-Ichi Oka
- Departiment of Cell and Molecular Biology, Rutgers New Jersey Medical School, Newark, NJ, USA
| | - Adam R Wende
- Division of Molecular and Cellular Pathology, Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Alexey V Zaitsev
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, VA, USA
| | - Junco S Warren
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, VA, USA; Center for Vascular and Heart Research, Fralin Biomedical Research Institute, Virginia Tech, Roanoke, VA, USA; Department of Human Nutrition, Food and Exercise, Virginia Tech, Blacksburg, VA, USA.
| |
Collapse
|
7
|
Chen X, Schubert SL, Müller A, Pishnamaz M, Hildebrand F, Nourbakhsh M. Metabolic Activity in Human Intermuscular Adipose Tissue Directs the Response of Resident PPARγ + Macrophages to Fatty Acids. Biomedicines 2024; 13:10. [PMID: 39857594 PMCID: PMC11759838 DOI: 10.3390/biomedicines13010010] [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: 11/22/2024] [Revised: 12/22/2024] [Accepted: 12/23/2024] [Indexed: 01/27/2025] Open
Abstract
Background/Objectives: Peroxisome proliferator-activated receptor gamma (PPARγ) is a fatty acid-binding transcription activator of the adipokine chemerin. The key role of PPARγ in adipogenesis was established by reports on adipose tissue-resident macrophages that express PPARγ. The present study examined PPARγ+ macrophages in human skeletal muscle tissues, their response to fatty acid (FA) species, and their correlations with age, obesity, adipokine expression, and an abundance of other macrophage phenotypes. Methods: An ex vivo human skeletal muscle model with surgical specimens that were maintained without or with FAs for up to 11 days was utilized. Immunofluorescence analysis was used to detect macrophage phenotypes and mitochondrial activity. Preconfigured arrays were used to detect the expression of 34 different adipokines and chemokines. Results: Data from 14 adults revealed that PPARγ+ macrophages exclusively reside in intermuscular adipose tissue (IMAT), and their abundance correlates with the metabolic status of surrounding adipocytes during tissue maintenance in vitro for 9-11 days. Elevated fatty acid levels lead to significant increases in PPARγ+ populations, which are correlated with the donor's body mass index (BMI). Conclusions: PPARγ+ macrophages represent a distinctly specialized population of regulatory cells that reside within human IMATs in accordance with their metabolic status. Thus, future in-depth studies on IMAT-resident PPARγ+ macrophage action mechanisms will elucidate the role of skeletal muscle in the pathogenesis of human metabolic dysfunction.
Collapse
Affiliation(s)
- Xiaoying Chen
- Institute of Pathology, RWTH Aachen University Hospital, 52074 Aachen, Germany; (X.C.); (A.M.)
| | - Sebastian Ludger Schubert
- Clinic for Orthopedics, Trauma, and Reconstructive Surgery, RWTH Aachen University Hospital, 52074 Aachen, Germany; (S.L.S.); (M.P.); (F.H.)
| | - Aline Müller
- Institute of Pathology, RWTH Aachen University Hospital, 52074 Aachen, Germany; (X.C.); (A.M.)
| | - Miguel Pishnamaz
- Clinic for Orthopedics, Trauma, and Reconstructive Surgery, RWTH Aachen University Hospital, 52074 Aachen, Germany; (S.L.S.); (M.P.); (F.H.)
| | - Frank Hildebrand
- Clinic for Orthopedics, Trauma, and Reconstructive Surgery, RWTH Aachen University Hospital, 52074 Aachen, Germany; (S.L.S.); (M.P.); (F.H.)
| | - Mahtab Nourbakhsh
- Institute of Pathology, RWTH Aachen University Hospital, 52074 Aachen, Germany; (X.C.); (A.M.)
| |
Collapse
|
8
|
Soares Menezes E, Wu Z, Renwick JRM, Moran-MacDonald A, Gurd BJ. PERM1-An Emerging Transcriptional Regulator of Mitochondrial Biogenesis: A Systematic Review. Genes (Basel) 2024; 15:1305. [PMID: 39457429 PMCID: PMC11508041 DOI: 10.3390/genes15101305] [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: 09/07/2024] [Revised: 09/30/2024] [Accepted: 10/02/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND/OBJECTIVES This systematic review aims to explore the role of PERM1 across different organisms, tissues, and cellular functions, with a particular focus on its involvement in regulating skeletal muscle mitochondrial biogenesis. METHODS This systematic review follows The PRISMA 2020 Statement. We used the Covidence systematic review software for abstract/title screening, full-text review, and data extraction. The review included studies that examined PERM1 expression or activity in skeletal muscle, heart, and adipose tissue and/or cells, from mice, rats, and humans, and involved exercise or disease models. Risk of bias was assessed using the Cochrane Collaboration tool, and the data were extracted and synthesized qualitatively, with bioinformatic analyses performed using the MetaMEx database. RESULTS Twenty-one studies were included in our data extraction process, where 10 studies involved humans, 21 involved mice, four involved rats, and 11 involved cells. CONCLUSIONS PERM1 in skeletal muscle increases with endurance exercise, affecting muscle function and oxidative metabolism, but its role in humans is not well understood. In cardiac tissue, PERM1 is vital for function and mitochondrial biogenesis purposes, but decreases with disease and pressure overload. Our review synthesizes the current understanding of PERM1's function, raises awareness of its role in mitochondrial regulation, and identifies key areas for future research in the field.
Collapse
Affiliation(s)
- Eveline Soares Menezes
- School of Kinesiology and Health Studies, Queen’s University, Kingston, ON K7L3N6, Canada; (E.S.M.); (Z.W.); (A.M.-M.)
| | - Zeyu Wu
- School of Kinesiology and Health Studies, Queen’s University, Kingston, ON K7L3N6, Canada; (E.S.M.); (Z.W.); (A.M.-M.)
| | - John R. M. Renwick
- Department of Kinesiology, McMaster University, Hamilton, ON L8S 4K1, Canada;
| | - Andres Moran-MacDonald
- School of Kinesiology and Health Studies, Queen’s University, Kingston, ON K7L3N6, Canada; (E.S.M.); (Z.W.); (A.M.-M.)
| | - Brendon J. Gurd
- School of Kinesiology and Health Studies, Queen’s University, Kingston, ON K7L3N6, Canada; (E.S.M.); (Z.W.); (A.M.-M.)
| |
Collapse
|
9
|
Sreedevi K, Zaitsev AV, Doku A, Thomas R, James A, Do S, Zhang M, Sedovy MW, Leng X, Dennison CL, Johnstone SR, Kirk JA, Yan Z, Warren JS. Adeno-associated virus-mediated gene delivery of Perm1 enhances cardiac contractility in mice. Am J Physiol Heart Circ Physiol 2024; 327:H1112-H1118. [PMID: 39269449 PMCID: PMC11482265 DOI: 10.1152/ajpheart.00545.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 09/03/2024] [Accepted: 09/11/2024] [Indexed: 09/15/2024]
Abstract
Reduced muscle contractility and mitochondrial bioenergetics are the hallmarks of systolic heart failure. There is currently no therapy targeting both. Here, we show that gene delivery of Perm1 via adeno-associated virus (AAV) simultaneously enhances cardiac contractility and mitochondrial biogenesis in C57BL6 mice. Moreover, we found that PERM1 interacts with troponin C (TnC), a key contractile protein in striated muscle, and that AAV-Perm1 led to the upregulation of TnC. This study suggests that gene delivery of Perm1 may be a novel therapeutic approach to treat systolic heart failure by simultaneously restoring cardiac contractility and mitochondrial bioenergetics.NEW & NOTEWORTHY Perm1 gene delivered with AAV9 enhances cardiac contractility in mice, and it is concomitant with the increase of mitochondrial bioenergetics and upregulation of TnC. This is the first study showing that PERM1, previously known as a striated muscle-specific mitochondrial regulator, also positively regulates cardiac contractility.
Collapse
Affiliation(s)
- Karthi Sreedevi
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, Virginia, United States
- Center for Vascular and Heart Research, Fralin Biomedical Research Institute, Virginia Tech, Roanoke, Virginia, United States
| | - Alexey V Zaitsev
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, Virginia, United States
- Center for Vascular and Heart Research, Fralin Biomedical Research Institute, Virginia Tech, Roanoke, Virginia, United States
| | - Abigail Doku
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, Virginia, United States
- Translational Biology Medicine and Health Graduate Program, Virginia Tech, Blacksburg, Virginia, United States
- Center for Vascular and Heart Research, Fralin Biomedical Research Institute, Virginia Tech, Roanoke, Virginia, United States
| | - Rebekah Thomas
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, Virginia, United States
- Translational Biology Medicine and Health Graduate Program, Virginia Tech, Blacksburg, Virginia, United States
- Center for Vascular and Heart Research, Fralin Biomedical Research Institute, Virginia Tech, Roanoke, Virginia, United States
| | - Amina James
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, Virginia, United States
- Department of Human Nutrition, Food and Exercise, Virginia Tech, Blacksburg, Virginia, United States
- Center for Vascular and Heart Research, Fralin Biomedical Research Institute, Virginia Tech, Roanoke, Virginia, United States
| | - Sara Do
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, Virginia, United States
- Center for Vascular and Heart Research, Fralin Biomedical Research Institute, Virginia Tech, Roanoke, Virginia, United States
| | - Mei Zhang
- Fralin Biomedical Research Institute, Center for Exercise Medicine Research at Virginia Tech Carilion, Roanoke, Virginia, United States
| | - Meghan W Sedovy
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, Virginia, United States
- Translational Biology Medicine and Health Graduate Program, Virginia Tech, Blacksburg, Virginia, United States
- Center for Vascular and Heart Research, Fralin Biomedical Research Institute, Virginia Tech, Roanoke, Virginia, United States
| | - Xinyan Leng
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, Virginia, United States
- Center for Vascular and Heart Research, Fralin Biomedical Research Institute, Virginia Tech, Roanoke, Virginia, United States
| | - Clare L Dennison
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, Virginia, United States
- Center for Vascular and Heart Research, Fralin Biomedical Research Institute, Virginia Tech, Roanoke, Virginia, United States
- Tissue Processing Core at Fralin Biomedical Research Institute, Virginia Tech, Roanoke, Virginia, United States
| | - Scott R Johnstone
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, Virginia, United States
- Center for Vascular and Heart Research, Fralin Biomedical Research Institute, Virginia Tech, Roanoke, Virginia, United States
- Department of Biological Sciences, College of Science, Virginia Tech, Roanoke, Virginia, United States
- Department of Surgery, Virginia Tech Carilion School of Medicine, Roanoke, Virginia, United States
| | - Jonathan A Kirk
- Department of Cell and Molecular Physiology, Loyola University Chicago, Chicago, Illinois, United States
| | - Zhen Yan
- Department of Human Nutrition, Food and Exercise, Virginia Tech, Blacksburg, Virginia, United States
- Fralin Biomedical Research Institute, Center for Exercise Medicine Research at Virginia Tech Carilion, Roanoke, Virginia, United States
| | - Junco S Warren
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, Virginia, United States
- Department of Human Nutrition, Food and Exercise, Virginia Tech, Blacksburg, Virginia, United States
- Center for Vascular and Heart Research, Fralin Biomedical Research Institute, Virginia Tech, Roanoke, Virginia, United States
| |
Collapse
|
10
|
Rojas-Torres M, Beltrán-Camacho L, Martínez-Val A, Sánchez-Gomar I, Eslava-Alcón S, Rosal-Vela A, Jiménez-Palomares M, Doiz-Artázcoz E, Martínez-Torija M, Moreno-Luna R, Olsen JV, Duran-Ruiz MC. Unraveling the differential mechanisms of revascularization promoted by MSCs & ECFCs from adipose tissue or umbilical cord in a murine model of critical limb-threatening ischemia. J Biomed Sci 2024; 31:71. [PMID: 39004727 PMCID: PMC11247736 DOI: 10.1186/s12929-024-01059-w] [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: 01/08/2024] [Accepted: 06/25/2024] [Indexed: 07/16/2024] Open
Abstract
BACKGROUND Critical limb-threatening ischemia (CLTI) constitutes the most severe manifestation of peripheral artery disease, usually induced by atherosclerosis. CLTI patients suffer from high risk of amputation of the lower extremities and elevated mortality rates, while they have low options for surgical revascularization due to associated comorbidities. Alternatively, cell-based therapeutic strategies represent an effective and safe approach to promote revascularization. However, the variability seen in several factors such as cell combinations or doses applied, have limited their success in clinical trials, being necessary to reach a consensus regarding the optimal "cellular-cocktail" prior further application into the clinic. To achieve so, it is essential to understand the mechanisms by which these cells exert their regenerative properties. Herein, we have evaluated, for the first time, the regenerative and vasculogenic potential of a combination of endothelial colony forming cells (ECFCs) and mesenchymal stem cells (MSCs) isolated from adipose-tissue (AT), compared with ECFCs from umbilical cord blood (CB-ECFCs) and AT-MSCs, in a murine model of CLTI. METHODS Balb-c nude mice (n:32) were distributed in four different groups (n:8/group): control shams, and ischemic mice (after femoral ligation) that received 50 µl of physiological serum alone or a cellular combination of AT-MSCs with either CB-ECFCs or AT-ECFCs. Follow-up of blood flow reperfusion and ischemic symptoms was carried out for 21 days, when mice were sacrificed to evaluate vascular density formation. Moreover, the long-term molecular changes in response to CLTI and both cell combinations were analyzed in a proteomic quantitative approach. RESULTS AT-MSCs with either AT- or CB-ECFCs, promoted a significant recovery of blood flow in CLTI mice 21 days post-ischemia. Besides, they modulated the inflammatory and necrotic related processes, although the CB group presented the slowest ischemic progression along the assay. Moreover, many proteins involved in the repairing mechanisms promoted by cell treatments were identified. CONCLUSIONS The combination of AT-MSCs with AT-ECFCs or with CB-ECFCs promoted similar revascularization in CLTI mice, by restoring blood flow levels, together with the modulation of the inflammatory and necrotic processes, and reduction of muscle damage. The protein changes identified are representative of the molecular mechanisms involved in ECFCs and MSCs-induced revascularization (immune response, vascular repair, muscle regeneration, etc.).
Collapse
Affiliation(s)
- Marta Rojas-Torres
- Biomedicine, Biotechnology and Public Health Department, University of Cadiz, Cadiz, 11002, Spain
- Biomedical Research and Innovation Institute of Cadiz (INiBICA), Cadiz, 11002, Spain
| | - Lucía Beltrán-Camacho
- Cell Biology, Physiology and Immunology Department, University of Cordoba, Cordoba, 14004, Spain
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Cordoba, 14004, Spain
| | - Ana Martínez-Val
- National Center of Cardiovascular Research Carlos III (CNIC), Madrid, 28029, Spain
| | - Ismael Sánchez-Gomar
- Biomedicine, Biotechnology and Public Health Department, University of Cadiz, Cadiz, 11002, Spain
- Biomedical Research and Innovation Institute of Cadiz (INiBICA), Cadiz, 11002, Spain
| | - Sara Eslava-Alcón
- Biomedicine, Biotechnology and Public Health Department, University of Cadiz, Cadiz, 11002, Spain
- Biomedical Research and Innovation Institute of Cadiz (INiBICA), Cadiz, 11002, Spain
| | - Antonio Rosal-Vela
- Biomedicine, Biotechnology and Public Health Department, University of Cadiz, Cadiz, 11002, Spain
- Biomedical Research and Innovation Institute of Cadiz (INiBICA), Cadiz, 11002, Spain
| | - Margarita Jiménez-Palomares
- Biomedicine, Biotechnology and Public Health Department, University of Cadiz, Cadiz, 11002, Spain
- Biomedical Research and Innovation Institute of Cadiz (INiBICA), Cadiz, 11002, Spain
| | - Esther Doiz-Artázcoz
- Angiology & Vascular Surgery Unit, Hospital Universitario Puerta del Mar, Cadiz, Spain
| | - Mario Martínez-Torija
- Pathophysiology and Regenerative Medicine Group, Hospital Nacional de Parapléjicos (SESCAM), Toledo, 45071, Spain
- Nursing department, Hospital Universitario de Toledo (SESCAM), Toledo, 45071, Spain
| | - Rafael Moreno-Luna
- Pathophysiology and Regenerative Medicine Group, Hospital Nacional de Parapléjicos (SESCAM), Toledo, 45071, Spain.
- Cooperative Research Network Orientated to Health Results, Vascular Brain Diseases, RICORS-ICTUS, SESCAM, Toledo, Spain.
| | - Jesper V Olsen
- Novo Nordisk Foundation Center for Protein Research, Copenhagen, Denmark
| | - Ma Carmen Duran-Ruiz
- Biomedicine, Biotechnology and Public Health Department, University of Cadiz, Cadiz, 11002, Spain.
- Biomedical Research and Innovation Institute of Cadiz (INiBICA), Cadiz, 11002, Spain.
- Biomedicine, Biotechnology and Public Health Department, Science Faculty, Cádiz University. Torre Sur. Avda. República Saharaui S/N, Polígono Río San Pedro, Puerto Real, Cádiz, 11519, Spain.
| |
Collapse
|
11
|
Rubio-Tomás T, Soler-Botija C, Martínez-Estrada O, Villena JA. Transcriptional control of cardiac energy metabolism in health and disease: Lessons from animal models. Biochem Pharmacol 2024; 224:116185. [PMID: 38561091 DOI: 10.1016/j.bcp.2024.116185] [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: 12/11/2023] [Revised: 02/27/2024] [Accepted: 03/29/2024] [Indexed: 04/04/2024]
Abstract
Cardiac ATP production is tightly regulated in order to satisfy the evolving energetic requirements imposed by different cues during health and pathological conditions. In order to sustain high ATP production rates, cardiac cells are endowed with a vast mitochondrial network that is essentially acquired during the perinatal period. Nevertheless, adult cardiac cells also adapt their mitochondrial mass and oxidative function to changes in energy demand and substrate availability by fine-tuning the pathways and mitochondrial machinery involved in energy production. The reliance of cardiac cells on mitochondrial metabolism makes them particularly sensitive to alterations in proper mitochondrial function, so that deficiency in energy production underlies or precipitates the development of heart diseases. Mitochondrial biogenesis is a complex process fundamentally controlled at the transcriptional level by a network of transcription factors and co-regulators, sometimes with partially redundant functions, that ensure adequate energy supply to the working heart. Novel uncovered regulators, such as RIP140, PERM1, MED1 or BRD4 have been recently shown to modulate or facilitate the transcriptional activity of the PGC-1s/ERRs/PPARs regulatory axis, allowing cardiomyocytes to adapt to a variety of physiological or pathological situations requiring different energy provision. In this review, we summarize the current knowledge on the mechanisms that regulate cardiac mitochondrial biogenesis, highlighting the recent discoveries of new transcriptional regulators and describing the experimental models that have provided solid evidence of the relevant contribution of these factors to cardiac function in health and disease.
Collapse
Affiliation(s)
- Teresa Rubio-Tomás
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology - Hellas, Heraklion GR-70013, Crete, Greece
| | - Carolina Soler-Botija
- ICREC (Heart Failure and Cardiac Regeneration) Research Program, Health Science Research Institute Germans Trias i Pujol (IGTP), Can Ruti Campus, Badalona, Spain; CIBER on Cardiovascular Diseases (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
| | | | - Josep A Villena
- Laboratory of Metabolism and Obesity, Vall d'Hebron-Institut de Recerca, Universitat Autònoma de Barcelona, 08035 Barcelona, Spain; CIBER on Diabetes and Associated Metabolic Diseases (CIBERDEM), 28029 Madrid, Spain.
| |
Collapse
|
12
|
Liu BH, Xu CZ, Liu Y, Lu ZL, Fu TL, Li GR, Deng Y, Luo GQ, Ding S, Li N, Geng Q. Mitochondrial quality control in human health and disease. Mil Med Res 2024; 11:32. [PMID: 38812059 PMCID: PMC11134732 DOI: 10.1186/s40779-024-00536-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 05/07/2024] [Indexed: 05/31/2024] Open
Abstract
Mitochondria, the most crucial energy-generating organelles in eukaryotic cells, play a pivotal role in regulating energy metabolism. However, their significance extends beyond this, as they are also indispensable in vital life processes such as cell proliferation, differentiation, immune responses, and redox balance. In response to various physiological signals or external stimuli, a sophisticated mitochondrial quality control (MQC) mechanism has evolved, encompassing key processes like mitochondrial biogenesis, mitochondrial dynamics, and mitophagy, which have garnered increasing attention from researchers to unveil their specific molecular mechanisms. In this review, we present a comprehensive summary of the primary mechanisms and functions of key regulators involved in major components of MQC. Furthermore, the critical physiological functions regulated by MQC and its diverse roles in the progression of various systemic diseases have been described in detail. We also discuss agonists or antagonists targeting MQC, aiming to explore potential therapeutic and research prospects by enhancing MQC to stabilize mitochondrial function.
Collapse
Affiliation(s)
- Bo-Hao Liu
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
- Department of Thoracic Surgery, First Hospital of Jilin University, Changchun, 130021, China
| | - Chen-Zhen Xu
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Yi Liu
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Zi-Long Lu
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Ting-Lv Fu
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Guo-Rui Li
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Yu Deng
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Guo-Qing Luo
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Song Ding
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Ning Li
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| | - Qing Geng
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| |
Collapse
|
13
|
Habib M, Lalagkas PN, Melamed RD. Mapping drug biology to disease genetics to discover drug impacts on the human phenome. BIOINFORMATICS ADVANCES 2024; 4:vbae038. [PMID: 38736684 PMCID: PMC11087821 DOI: 10.1093/bioadv/vbae038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 01/18/2024] [Accepted: 03/07/2024] [Indexed: 05/14/2024]
Abstract
Motivation Medications can have unexpected effects on disease, including not only harmful drug side effects, but also beneficial drug repurposing. These effects on disease may result from hidden influences of drugs on disease gene networks. Then, discovering how biological effects of drugs relate to disease biology can both provide insight into the mechanism of latent drug effects, and can help predict new effects. Results Here, we develop Draphnet, a model that integrates molecular data on 429 drugs and gene associations of nearly 200 common phenotypes to learn a network that explains drug effects on disease in terms of these molecular signals. We present evidence that our method can both predict drug effects, and can provide insight into the biology of unexpected drug effects on disease. Using Draphnet to map a drug's known molecular effects to downstream effects on the disease genome, we put forward disease genes impacted by drugs, and we suggest a new grouping of drugs based on shared effects on the disease genome. Our approach has multiple applications, including predicting drug uses and learning drug biology, with implications for personalized medicine. Availability and implementation Code to reproduce the analysis is available at https://github.com/RDMelamed/drug-phenome.
Collapse
Affiliation(s)
- Mamoon Habib
- Department of Computer Science, University of Massachusetts Lowell, Lowell, MA 01854, United States
| | | | - Rachel D Melamed
- Department of Biological Science, University of Massachusetts Lowell, Lowell, MA 01854, United States
| |
Collapse
|
14
|
Sakamoto T, Kelly DP. Cardiac maturation. J Mol Cell Cardiol 2024; 187:38-50. [PMID: 38160640 PMCID: PMC10923079 DOI: 10.1016/j.yjmcc.2023.12.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 12/12/2023] [Accepted: 12/21/2023] [Indexed: 01/03/2024]
Abstract
The heart undergoes a dynamic maturation process following birth, in response to a wide range of stimuli, including both physiological and pathological cues. This process entails substantial re-programming of mitochondrial energy metabolism coincident with the emergence of specialized structural and contractile machinery to meet the demands of the adult heart. Many components of this program revert to a more "fetal" format during development of pathological cardiac hypertrophy and heart failure. In this review, emphasis is placed on recent progress in our understanding of the transcriptional control of cardiac maturation, encompassing the results of studies spanning from in vivo models to cardiomyocytes derived from human stem cells. The potential applications of this current state of knowledge to new translational avenues aimed at the treatment of heart failure is also addressed.
Collapse
Affiliation(s)
- Tomoya Sakamoto
- Cardiovascular Institute, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Daniel P Kelly
- Cardiovascular Institute, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
15
|
Kumar A, Narkar VA. Nuclear receptors as potential therapeutic targets in peripheral arterial disease and related myopathy. FEBS J 2023; 290:4596-4613. [PMID: 35942640 PMCID: PMC9908775 DOI: 10.1111/febs.16593] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 07/28/2022] [Accepted: 08/08/2022] [Indexed: 12/31/2022]
Abstract
Peripheral arterial disease (PAD) is a prevalent cardiovascular complication of limb vascular insufficiency, causing ischemic injury, mitochondrial metabolic damage and functional impairment in the skeletal muscle, and ultimately leading to immobility and mortality. While potential therapies have been mostly focussed on revascularization, none of the currently available pharmacological treatments are fully effective in PAD, often leading to amputations, particularly in chronic metabolic diseases. One major limitation of focussed angiogenesis and revascularization as a therapeutic strategy is a limited effect on metabolic restoration and muscle regeneration in the affected limb. Therefore, additional preclinical investigations are needed to discover novel treatment options for PAD preferably targeting multiple aspects of muscle recovery. In this review, we propose nuclear receptors expressed in the skeletal muscle as potential candidates for ischemic muscle repair in PAD. We review classic steroid and orphan receptors that have been reported to be involved in the regulation of paracrine muscle angiogenesis, oxidative metabolism, mitochondrial biogenesis and muscle regeneration, and discuss how these receptors could be critical for recovery from ischemic muscle damage. Furthermore, we identify existing gaps in our understanding of nuclear receptor signalling in the skeletal muscle and propose future areas of research that could be instrumental in exploring nuclear receptors as therapeutic candidates for treating PAD.
Collapse
Affiliation(s)
- Ashok Kumar
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX 77204
| | - Vihang A. Narkar
- Brown Foundation Institute of Molecular Medicine, UTHealth McGovern Medical School, Houston, TX, 77030
- University of Texas MD Anderson and UTHealth Graduate School of Biomedical Sciences, Houston, TX, 77030
| |
Collapse
|
16
|
Di Pierro E, Perrone M, Franco M, Granata F, Duca L, Lattuada D, De Luca G, Graziadei G. Mitochondrial DNA Copy Number Drives the Penetrance of Acute Intermittent Porphyria. Life (Basel) 2023; 13:1923. [PMID: 37763326 PMCID: PMC10532762 DOI: 10.3390/life13091923] [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: 08/07/2023] [Revised: 09/07/2023] [Accepted: 09/11/2023] [Indexed: 09/29/2023] Open
Abstract
No published study has investigated the mitochondrial count in patients suffering from acute intermittent porphyria (AIP). In order to determine whether mitochondrial content can influence the pathogenesis of porphyria, we measured the mitochondrial DNA (mtDNA) copy number in the peripheral blood cells of 34 patients and 37 healthy individuals. We found that all AIP patients had a low number of mitochondria, likely as a result of a protective mechanism against an inherited heme synthesis deficiency. Furthermore, we identified a close correlation between disease penetrance and decreases in the mitochondrial content and serum levels of PERM1, a marker of mitochondrial biogenesis. In a healthy individual, mitochondrial count is usually modulated to fit its ability to respond to various environmental stressors and bioenergetic demands. In AIP patients, coincidentally, the phenotype only manifests in response to endogenous and exogenous triggers factors. Therefore, these new findings suggest that a deficiency in mitochondrial proliferation could affect the individual responsiveness to stimuli, providing a new explanation for the variability in the clinical manifestations of porphyria. However, the metabolic and/or genetic factors responsible for this impairment remain to be identified. In conclusion, both mtDNA copy number per cell and mitochondrial biogenesis seem to play a role in either inhibiting or promoting disease expression. They could serve as two novel biomarkers for porphyria.
Collapse
Affiliation(s)
- Elena Di Pierro
- Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (M.P.); (F.G.); (L.D.); (D.L.); (G.G.)
| | - Miriana Perrone
- Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (M.P.); (F.G.); (L.D.); (D.L.); (G.G.)
| | - Milena Franco
- Department of Molecular Medicine, University of Pavia, 27100 Pavia, Italy;
| | - Francesca Granata
- Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (M.P.); (F.G.); (L.D.); (D.L.); (G.G.)
| | - Lorena Duca
- Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (M.P.); (F.G.); (L.D.); (D.L.); (G.G.)
| | - Debora Lattuada
- Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (M.P.); (F.G.); (L.D.); (D.L.); (G.G.)
| | - Giacomo De Luca
- School of Internal Medicine, University of Milan, 20122 Milan, Italy;
| | - Giovanna Graziadei
- Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (M.P.); (F.G.); (L.D.); (D.L.); (G.G.)
| |
Collapse
|
17
|
Aboouf MA, Gorr TA, Hamdy NM, Gassmann M, Thiersch M. Myoglobin in Brown Adipose Tissue: A Multifaceted Player in Thermogenesis. Cells 2023; 12:2240. [PMID: 37759463 PMCID: PMC10526770 DOI: 10.3390/cells12182240] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 09/01/2023] [Accepted: 09/04/2023] [Indexed: 09/29/2023] Open
Abstract
Brown adipose tissue (BAT) plays an important role in energy homeostasis by generating heat from chemical energy via uncoupled oxidative phosphorylation. Besides its high mitochondrial content and its exclusive expression of the uncoupling protein 1, another key feature of BAT is the high expression of myoglobin (MB), a heme-containing protein that typically binds oxygen, thereby facilitating the diffusion of the gas from cell membranes to mitochondria of muscle cells. In addition, MB also modulates nitric oxide (NO•) pools and can bind C16 and C18 fatty acids, which indicates a role in lipid metabolism. Recent studies in humans and mice implicated MB present in BAT in the regulation of lipid droplet morphology and fatty acid shuttling and composition, as well as mitochondrial oxidative metabolism. These functions suggest that MB plays an essential role in BAT energy metabolism and thermogenesis. In this review, we will discuss in detail the possible physiological roles played by MB in BAT thermogenesis along with the potential underlying molecular mechanisms and focus on the question of how BAT-MB expression is regulated and, in turn, how this globin regulates mitochondrial, lipid, and NO• metabolism. Finally, we present potential MB-mediated approaches to augment energy metabolism, which ultimately could help tackle different metabolic disorders.
Collapse
Affiliation(s)
- Mostafa A. Aboouf
- Institute of Veterinary Physiology, University of Zurich, 8057 Zurich, Switzerland
- Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, 8057 Zurich, Switzerland
- Department of Biochemistry, Faculty of Pharmacy, Ain Shams University, Cairo 11566, Egypt
| | - Thomas A. Gorr
- Institute of Veterinary Physiology, University of Zurich, 8057 Zurich, Switzerland
| | - Nadia M. Hamdy
- Department of Biochemistry, Faculty of Pharmacy, Ain Shams University, Cairo 11566, Egypt
| | - Max Gassmann
- Institute of Veterinary Physiology, University of Zurich, 8057 Zurich, Switzerland
- Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, 8057 Zurich, Switzerland
| | - Markus Thiersch
- Institute of Veterinary Physiology, University of Zurich, 8057 Zurich, Switzerland
- Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, 8057 Zurich, Switzerland
| |
Collapse
|
18
|
Yang YN, Zhang MQ, Yu FL, Han B, Bao MY, Yan-He, Li X, Zhang Y. Peroxisom proliferator-activated receptor-γ coactivator-1α in neurodegenerative disorders: A promising therapeutic target. Biochem Pharmacol 2023; 215:115717. [PMID: 37516277 DOI: 10.1016/j.bcp.2023.115717] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Revised: 07/26/2023] [Accepted: 07/26/2023] [Indexed: 07/31/2023]
Abstract
Neurodegenerative disorders (NDDs) are characterized by progressive loss of selectively vulnerable neuronal populations and myelin sheath, leading to behavioral and cognitive dysfunction that adversely affect the quality of life. Identifying novel therapies that attenuate the progression of NDDs would be of significance. Peroxisome proliferator-activated receptor-γ coactivator-1α (PGC-1α), a widely expressed transcriptional regulator, modulates the expression of genes engaged in mitochondrial biosynthesis, metabolic regulation, and oxidative stress (OS). Emerging evidences point to the strong connection between PGC-1α and NDDs, suggesting its positive impaction on the progression of NDDs. Therefore, it is urgent to gain a deeper and broader understanding between PGC-1α and NDDs. To this end, this review presents a comprehensive overview of PGC-1α, including its basic characteristics, the post-translational modulations, as well as the interacting transcription factors. Secondly, the pathogenesis of PGC-1α in various NDDs, such as Alzheimer's (AD), Parkinson's (PD), and Huntington's disease (HD) is briefly discussed. Additionally, this study summarizes the underlying mechanisms that PGC-1α is neuroprotective in NDDs via regulating neuroinflammation, OS, and mitochondrial dysfunction. Finally, we briefly outline the shortcomings of current NDDs drug therapy, and summarize the functions and potential applications of currently available PGC-1α modulators (activator or inhibitors). Generally, this review updates our insight of the important role of PGC-1α on the development of NDDs, and provides a promising therapeutic target/ drug for the treatment of NDDs.
Collapse
Affiliation(s)
- Ya-Na Yang
- Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry (Shaanxi Normal University), The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi 710119, China
| | - Mao-Qing Zhang
- Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry (Shaanxi Normal University), The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi 710119, China
| | - Feng-Lin Yu
- Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry (Shaanxi Normal University), The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi 710119, China
| | - Bing Han
- Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry (Shaanxi Normal University), The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi 710119, China
| | - Ming-Yue Bao
- Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry (Shaanxi Normal University), The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi 710119, China
| | - Yan-He
- Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry (Shaanxi Normal University), The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi 710119, China
| | - Xing Li
- Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry (Shaanxi Normal University), The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi 710119, China
| | - Yuan Zhang
- Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry (Shaanxi Normal University), The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi 710119, China.
| |
Collapse
|
19
|
He Y, Wang Q, Wu H, Dong Y, Peng Z, Guo X, Jiang N. The role of IGF-1 in exercise to improve obesity-related cognitive dysfunction. Front Neurosci 2023; 17:1229165. [PMID: 37638322 PMCID: PMC10447980 DOI: 10.3389/fnins.2023.1229165] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 07/31/2023] [Indexed: 08/29/2023] Open
Abstract
Obesity is an important factor that threatens human health. The occurrence of many chronic diseases is related to obesity, and cognitive function decline often occurs with the onset of obesity. With the further prevalence of obesity, it is bound to lead to a wider range of cognitive dysfunction (ORCD). Therefore, it is crucial to suppress ORCD through intervention. In this regard, exercise has been shown to be effective in preventing obesity and improving cognitive function as a non-drug treatment. There is sufficient evidence that exercise has a regulatory effect on a growth factor closely related to cognitive function-insulin-like growth factor 1 (IGF-1). IGF-1 may be an important mediator in improving ORCD through exercise. This article reviews the effects of obesity and IGF-1 on cognitive function and the regulation of exercise on IGF-1. It analyzes the mechanism by which exercise can improve ORCD by regulating IGF-1. Overall, this review provides evidence from relevant animal studies and human studies, showing that exercise plays a role in improving ORCD. It emphasizes the importance of IGF-1, which helps to understand the health effects of exercise and promotes research on the treatment of ORCD.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Ning Jiang
- Tianjin Key Laboratory of Exercise Physiology and Sports Medicine, Institute of Sport, Exercise and Health, Tianjin University of Sport, Tianjin, China
| |
Collapse
|
20
|
Paez HG, Ferrandi PJ, Pitzer CR, Mohamed JS, Alway SE. Loss of NOR-1 represses muscle metabolism through mTORC1-mediated signaling and mitochondrial gene expression in C2C12 myotubes. FASEB J 2023; 37:e23050. [PMID: 37389860 DOI: 10.1096/fj.202202029r] [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: 12/03/2022] [Revised: 05/26/2023] [Accepted: 06/08/2023] [Indexed: 07/01/2023]
Abstract
Gene expression of the NR4A nuclear orphan receptor NOR-1 is reduced in obesity and in human skeletal muscle during disuse. It has been well established that NOR-1 is highly responsive to both aerobic and resistance exercise and NOR-1 overexpression is coincident with a plethora of metabolic benefits. However, it is unclear whether loss of NOR-1 contributes to inappropriate metabolic signaling in skeletal muscle that could lead to insulin resistance. The purpose of this study was to elucidate the impact of NOR-1 deficiency on C2C12 metabolic signaling. Changes in gene expression after siRNA-mediated NOR-1 knockdown in C2C12 myotubes were determined by qPCR and bioinformatic analysis of RNA-Seq data. Our RNA-Seq data identified several metabolic targets regulated by NOR-1 and implicates NOR-1 as a modulator of mTORC1 signaling via Akt-independent mechanisms. Furthermore, pathway analysis revealed NOR-1 knockdown perturbs the insulin resistance and insulin sensitivity pathways. Taken together, these data suggest skeletal muscle NOR-1 deficiency may contribute to altered metabolic signaling that is consistent with metabolic disease. We postulate that strategies that improve NOR-1 may be important to offset the negative impact that inactivity, obesity, and type 2 diabetes have on mitochondria and muscle metabolism.
Collapse
Affiliation(s)
- Hector G Paez
- Department of Physiology, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, USA
- Integrated Biomedical Sciences Graduate Program, College of Graduate Health Sciences, University of Tennessee Health Science Center, Memphis, Tennessee, USA
- Laboratory of Muscle Biology and Sarcopenia, Department of Physical Therapy, College of Health Professions, University of Tennessee Health Science Center, Memphis, Tennessee, USA
- Division of Regenerative and Rehabilitation Sciences, Center for Muscle, Metabolism and Neuropathology, College of Health Professions, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Peter J Ferrandi
- Integrated Biomedical Sciences Graduate Program, College of Graduate Health Sciences, University of Tennessee Health Science Center, Memphis, Tennessee, USA
- Division of Regenerative and Rehabilitation Sciences, Center for Muscle, Metabolism and Neuropathology, College of Health Professions, University of Tennessee Health Science Center, Memphis, Tennessee, USA
- Laboratory of Muscle and Nerve, Department of Diagnostic and Health Sciences, College of Health Professions, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Christopher R Pitzer
- Department of Physiology, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, USA
- Integrated Biomedical Sciences Graduate Program, College of Graduate Health Sciences, University of Tennessee Health Science Center, Memphis, Tennessee, USA
- Laboratory of Muscle Biology and Sarcopenia, Department of Physical Therapy, College of Health Professions, University of Tennessee Health Science Center, Memphis, Tennessee, USA
- Division of Regenerative and Rehabilitation Sciences, Center for Muscle, Metabolism and Neuropathology, College of Health Professions, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Junaith S Mohamed
- Integrated Biomedical Sciences Graduate Program, College of Graduate Health Sciences, University of Tennessee Health Science Center, Memphis, Tennessee, USA
- Division of Regenerative and Rehabilitation Sciences, Center for Muscle, Metabolism and Neuropathology, College of Health Professions, University of Tennessee Health Science Center, Memphis, Tennessee, USA
- Laboratory of Muscle and Nerve, Department of Diagnostic and Health Sciences, College of Health Professions, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Stephen E Alway
- Department of Physiology, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, USA
- Integrated Biomedical Sciences Graduate Program, College of Graduate Health Sciences, University of Tennessee Health Science Center, Memphis, Tennessee, USA
- Laboratory of Muscle Biology and Sarcopenia, Department of Physical Therapy, College of Health Professions, University of Tennessee Health Science Center, Memphis, Tennessee, USA
- Division of Regenerative and Rehabilitation Sciences, Center for Muscle, Metabolism and Neuropathology, College of Health Professions, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| |
Collapse
|
21
|
Sopariwala DH, Hao NTT, Narkar VA. Estrogen-related Receptor Signaling in Skeletal Muscle Fitness. Int J Sports Med 2023; 44:609-617. [PMID: 36787804 PMCID: PMC11168301 DOI: 10.1055/a-2035-8192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/16/2023]
Abstract
Skeletal muscle is a highly plastic tissue that can alter its metabolic and contractile features, as well as regenerative potential in response to exercise and other conditions. Multiple signaling factors including metabolites, kinases, receptors, and transcriptional factors have been studied in the regulation of skeletal muscle plasticity. Recently, estrogen-related receptors (ERRs) have emerged as a critical transcriptional hub in control of skeletal muscle homeostasis. ERRα and ERRγ - the two highly expressed ERR sub-types in the muscle respond to various extracellular cues such as exercise, hypoxia, fasting and dietary factors, in turn regulating gene expression in the skeletal muscle. On the other hand, conditions such as diabetes and muscular dystrophy suppress expression of ERRs in the skeletal muscle, likely contributing to disease progression. We highlight key functions of ERRs in the skeletal muscle including the regulation of fiber type, mitochondrial metabolism, vascularization, and regeneration. We also describe how ERRs are regulated in the skeletal muscle, and their interaction with important muscle regulators (e. g. AMPK and PGCs). Finally, we identify critical gaps in our understanding of ERR signaling in the skeletal muscle, and suggest future areas of investigation to advance ERRs as potential targets for function promoting therapeutics in muscle diseases.
Collapse
Affiliation(s)
- Danesh H. Sopariwala
- Brown Foundation Institute of Molecular Medicine, McGovern Medical School at The University of Texas Health Science Center (UTHealth), Houston, TX, USA
| | - Nguyen Thi Thu Hao
- Brown Foundation Institute of Molecular Medicine, McGovern Medical School at The University of Texas Health Science Center (UTHealth), Houston, TX, USA
| | - Vihang A. Narkar
- Brown Foundation Institute of Molecular Medicine, McGovern Medical School at The University of Texas Health Science Center (UTHealth), Houston, TX, USA
| |
Collapse
|
22
|
Ulaganathan T, Perales S, Mani S, Baskhairoun BA, Rajasingh J. Pathological implications of cellular stress in cardiovascular diseases. Int J Biochem Cell Biol 2023; 158:106397. [PMID: 36931385 PMCID: PMC10124590 DOI: 10.1016/j.biocel.2023.106397] [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: 09/26/2022] [Revised: 03/10/2023] [Accepted: 03/14/2023] [Indexed: 03/17/2023]
Abstract
Cellular stress has been a key factor in the development of cardiovascular diseases. Major types of cellular stress such as mitochondrial stress, endoplasmic reticulum stress, hypoxia, and replicative stress have been implicated in clinical complications of cardiac patients. The heart is the central regulator of the body by supplying oxygenated blood throughout the system. Impairment of cellular function could lead to heart failure, myocardial infarction, ischemia, and even stroke. Understanding the effect of these distinct types of cellular stress on cardiac function is crucial for the scientific community to understand and develop novel therapeutic approaches. This review will comprehensively explain the different mechanisms of cellular stress and the most recent findings related to stress-induced cardiac dysfunction.
Collapse
Affiliation(s)
- Thennavan Ulaganathan
- Department of Bioscience Research, University of Tennessee Health Science Center, Memphis, TN, USA; Department of Biotechnology, SRM Institute of Science and Technology, kattankulathur, Tamilnadu, 603203, India
| | - Selene Perales
- Department of Bioscience Research, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Saiprahalad Mani
- Department of Bioscience Research, University of Tennessee Health Science Center, Memphis, TN, USA; Department of Biotechnology, SRM Institute of Science and Technology, kattankulathur, Tamilnadu, 603203, India
| | - Boula A Baskhairoun
- Department of Bioscience Research, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Johnson Rajasingh
- Department of Bioscience Research, University of Tennessee Health Science Center, Memphis, TN, USA; Department of Medicine-Cardiology, University of Tennessee Health Science Center, Memphis, TN, USA; Department of Microbiology, Immunology, and Biochemistry, University of Tennessee Health Science Center, Memphis, TN, USA.
| |
Collapse
|
23
|
Chatzinikita E, Maridaki M, Palikaras K, Koutsilieris M, Philippou A. The Role of Mitophagy in Skeletal Muscle Damage and Regeneration. Cells 2023; 12:716. [PMID: 36899852 PMCID: PMC10000750 DOI: 10.3390/cells12050716] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 02/18/2023] [Accepted: 02/22/2023] [Indexed: 02/26/2023] Open
Abstract
Mitochondria are cellular organelles that play an essential role in generating the chemical energy needed for the biochemical reactions in cells. Mitochondrial biogenesis, i.e., de novo mitochondria formation, results in enhanced cellular respiration, metabolic processes, and ATP generation, while autophagic clearance of mitochondria (mitophagy) is required to remove damaged or useless mitochondria. The balance between the opposing processes of mitochondrial biogenesis and mitophagy is highly regulated and crucial for the maintenance of the number and function of mitochondria as well as for the cellular homeostasis and adaptations to metabolic demands and extracellular stimuli. In skeletal muscle, mitochondria are essential for maintaining energy homeostasis, and the mitochondrial network exhibits complex behaviors and undergoes dynamic remodeling in response to various conditions and pathologies characterized by changes in muscle cell structure and metabolism, such as exercise, muscle damage, and myopathies. In particular, the involvement of mitochondrial remodeling in mediating skeletal muscle regeneration following damage has received increased attention, as modifications in mitophagy-related signals arise from exercise, while variations in mitochondrial restructuring pathways can lead to partial regeneration and impaired muscle function. Muscle regeneration (through myogenesis) following exercise-induced damage is characterized by a highly regulated, rapid turnover of poor-functioning mitochondria, permitting the synthesis of better-functioning mitochondria to occur. Nevertheless, essential aspects of mitochondrial remodeling during muscle regeneration remain poorly understood and warrant further characterization. In this review, we focus on the critical role of mitophagy for proper muscle cell regeneration following damage, highlighting the molecular mechanisms of the mitophagy-associated mitochondrial dynamics and network reformation.
Collapse
Affiliation(s)
- Eirini Chatzinikita
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, 115 27 Athens, Greece
| | - Maria Maridaki
- Faculty of Physical Education and Sport Science, National and Kapodistrian University of Athens, 172 37 Athens, Greece
| | - Konstantinos Palikaras
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, 115 27 Athens, Greece
| | - Michael Koutsilieris
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, 115 27 Athens, Greece
| | - Anastassios Philippou
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, 115 27 Athens, Greece
| |
Collapse
|
24
|
Sopariwala DH, Rios AS, Pei G, Roy A, Tomaz da Silva M, Thi Thu Nguyen H, Saley A, Van Drunen R, Kralli A, Mahan K, Zhao Z, Kumar A, Narkar VA. Innately expressed estrogen-related receptors in the skeletal muscle are indispensable for exercise fitness. FASEB J 2023; 37:e22727. [PMID: 36583689 DOI: 10.1096/fj.202201518r] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 12/01/2022] [Accepted: 12/12/2022] [Indexed: 12/31/2022]
Abstract
Transcriptional determinants in the skeletal muscle that govern exercise capacity, while poorly defined, could provide molecular insights into how exercise improves fitness. Here, we have elucidated the role of nuclear receptors, estrogen-related receptor alpha and gamma (ERRα/γ) in regulating myofibrillar composition, contractility, and exercise capacity in skeletal muscle. We used muscle-specific single or double (DKO) ERRα/γ knockout mice to investigate the effect of ERRα/γ deletion on muscle and exercise parameters. Individual knockout of ERRα/γ did not have a significant impact on the skeletal muscle. On the other hand, DKO mice exhibit pale muscles compared to wild-type (WT) littermates. RNA-seq analysis revealed a predominant decrease in expression of genes linked to mitochondrial and oxidative metabolism in DKO versus WT muscles. DKO muscles exhibit marked repression of oxidative enzymatic capacity, as well as mitochondrial number and size compared to WT muscles. Mitochondrial function is also impaired in single myofibers isolated from DKO versus WT muscles. In addition, mutant muscles exhibit reduced angiogenic gene expression and decreased capillarity. Consequently, DKO mice have a significantly reduced exercise capacity, further reflected in poor fatigue resistance of DKO mice in in vivo contraction assays. These results show that ERRα and ERRγ together are a critical link between muscle aerobic capacity and exercise tolerance. The ERRα/γ mutant mice could be valuable for understanding the long-term impact of impaired mitochondria and vascular supply on the pathogenesis of muscle-linked disorders.
Collapse
Affiliation(s)
- Danesh H Sopariwala
- Brown Foundation Institute of Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center, Houston, Texas, USA
| | - Andrea S Rios
- Brown Foundation Institute of Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center, Houston, Texas, USA
| | - Guangsheng Pei
- Center for Precision Medicine, School of Biomedical Informatics, The University of Texas Health Science Center, Houston, Texas, USA
| | - Anirban Roy
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston, Houston, Texas, USA
| | - Meiricris Tomaz da Silva
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston, Houston, Texas, USA
| | - Hao Thi Thu Nguyen
- Brown Foundation Institute of Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center, Houston, Texas, USA
| | - Addison Saley
- Brown Foundation Institute of Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center, Houston, Texas, USA.,Department of Biosciences, Rice University, Houston, Texas, USA
| | - Rachel Van Drunen
- Brown Foundation Institute of Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center, Houston, Texas, USA
| | - Anastasia Kralli
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Kristin Mahan
- Brown Foundation Institute of Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center, Houston, Texas, USA.,Department of Integrative Biology and Pharmacology, The University of Texas Health Science Center, Houston, Texas, USA
| | - Zhongming Zhao
- Center for Precision Medicine, School of Biomedical Informatics, The University of Texas Health Science Center, Houston, Texas, USA.,Human Genetics Center, School of Public Health, The University of Texas Health Science Center, Houston, Texas, USA
| | - Ashok Kumar
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston, Houston, Texas, USA
| | - Vihang A Narkar
- Brown Foundation Institute of Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center, Houston, Texas, USA.,Graduate School of Biomedical Sciences at UTHealth, Houston, Texas, USA
| |
Collapse
|
25
|
Araki H, Hino S, Anan K, Kuribayashi K, Etoh K, Seko D, Takase R, Kohrogi K, Hino Y, Ono Y, Araki E, Nakao M. LSD1 defines the fiber type-selective responsiveness to environmental stress in skeletal muscle. eLife 2023; 12:84618. [PMID: 36695573 PMCID: PMC9876571 DOI: 10.7554/elife.84618] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 01/12/2023] [Indexed: 01/26/2023] Open
Abstract
Skeletal muscle exhibits remarkable plasticity in response to environmental cues, with stress-dependent effects on the fast-twitch and slow-twitch fibers. Although stress-induced gene expression underlies environmental adaptation, it is unclear how transcriptional and epigenetic factors regulate fiber type-specific responses in the muscle. Here, we show that flavin-dependent lysine-specific demethylase-1 (LSD1) differentially controls responses to glucocorticoid and exercise in postnatal skeletal muscle. Using skeletal muscle-specific LSD1-knockout mice and in vitro approaches, we found that LSD1 loss exacerbated glucocorticoid-induced atrophy in the fast fiber-dominant muscles, with reduced nuclear retention of Foxk1, an anti-autophagic transcription factor. Furthermore, LSD1 depletion enhanced endurance exercise-induced hypertrophy in the slow fiber-dominant muscles, by induced expression of ERRγ, a transcription factor that promotes oxidative metabolism genes. Thus, LSD1 serves as an 'epigenetic barrier' that optimizes fiber type-specific responses and muscle mass under the stress conditions. Our results uncover that LSD1 modulators provide emerging therapeutic and preventive strategies against stress-induced myopathies such as sarcopenia, cachexia, and disuse atrophy.
Collapse
Affiliation(s)
- Hirotaka Araki
- Department of Medical Cell Biology, Institute of Molecular Embryology and Genetics, Kumamoto UniversityKumamotoJapan
- Department of Metabolic Medicine, Faculty of Life Sciences, Kumamoto UniversityKumamotoJapan
| | - Shinjiro Hino
- Department of Medical Cell Biology, Institute of Molecular Embryology and Genetics, Kumamoto UniversityKumamotoJapan
| | - Kotaro Anan
- Department of Medical Cell Biology, Institute of Molecular Embryology and Genetics, Kumamoto UniversityKumamotoJapan
| | - Kanji Kuribayashi
- Department of Medical Cell Biology, Institute of Molecular Embryology and Genetics, Kumamoto UniversityKumamotoJapan
| | - Kan Etoh
- Department of Medical Cell Biology, Institute of Molecular Embryology and Genetics, Kumamoto UniversityKumamotoJapan
| | - Daiki Seko
- Department of Muscle Development and Regeneration, Institute of Molecular Embryology and Genetics, Kumamoto UniversityKumamotoJapan
- Department of Molecular Bone Biology, Graduate School of Biomedical Sciences, Nagasaki UniversityNagasakiJapan
| | - Ryuta Takase
- Department of Medical Cell Biology, Institute of Molecular Embryology and Genetics, Kumamoto UniversityKumamotoJapan
| | - Kensaku Kohrogi
- Department of Medical Cell Biology, Institute of Molecular Embryology and Genetics, Kumamoto UniversityKumamotoJapan
| | - Yuko Hino
- Department of Medical Cell Biology, Institute of Molecular Embryology and Genetics, Kumamoto UniversityKumamotoJapan
| | - Yusuke Ono
- Department of Muscle Development and Regeneration, Institute of Molecular Embryology and Genetics, Kumamoto UniversityKumamotoJapan
| | - Eiichi Araki
- Department of Metabolic Medicine, Faculty of Life Sciences, Kumamoto UniversityKumamotoJapan
| | - Mitsuyoshi Nakao
- Department of Medical Cell Biology, Institute of Molecular Embryology and Genetics, Kumamoto UniversityKumamotoJapan
| |
Collapse
|
26
|
Shaito A, Al-Mansoob M, Ahmad SM, Haider MZ, Eid AH, Posadino AM, Pintus G, Giordo R. Resveratrol-Mediated Regulation of Mitochondria Biogenesis-associated Pathways in Neurodegenerative Diseases: Molecular Insights and Potential Therapeutic Applications. Curr Neuropharmacol 2023; 21:1184-1201. [PMID: 36237161 PMCID: PMC10286596 DOI: 10.2174/1570159x20666221012122855] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Revised: 09/22/2022] [Accepted: 10/09/2022] [Indexed: 11/22/2022] Open
Abstract
Neurodegenerative disorders include different neurological conditions that affect nerve cells, causing the progressive loss of their functions and ultimately leading to loss of mobility, coordination, and mental functioning. The molecular mechanisms underpinning neurodegenerative disease pathogenesis are still unclear. Nonetheless, there is experimental evidence to demonstrate that the perturbation of mitochondrial function and dynamics play an essential role. In this context, mitochondrial biogenesis, the growth, and division of preexisting mitochondria, by controlling mitochondria number, plays a vital role in maintaining proper mitochondrial mass and function, thus ensuring efficient synaptic activity and brain function. Mitochondrial biogenesis is tightly associated with the control of cell division and variations in energy demand in response to extracellular stimuli; therefore, it may represent a promising therapeutic target for developing new curative approaches to prevent or counteract neurodegenerative disorders. Accordingly, several inducers of mitochondrial biogenesis have been proposed as pharmacological targets for treating diverse central nervous system conditions. The naturally occurring polyphenol resveratrol has been shown to promote mitochondrial biogenesis in various tissues, including the nervous tissue, and an ever-growing number of studies highlight its neurotherapeutic potential. Besides preventing cognitive impairment and neurodegeneration through its antioxidant and anti-inflammatory properties, resveratrol has been shown to be able to enhance mitochondria biogenesis by acting on its main effectors, including PGC-1α, SIRT1, AMPK, ERRs, TERT, TFAM, NRF-1 and NRF-2. This review aims to present and discuss the current findings concerning the impact of resveratrol on the machinery and main effectors modulating mitochondrial biogenesis in the context of neurodegenerative diseases.
Collapse
Affiliation(s)
- Abdullah Shaito
- Biomedical Research Center, College of Medicine, Qatar University, Doha, 2713, Qatar
- Department of Biomedical Sciences, College of Health Sciences, Qatar University, Doha, 2713, Qatar
| | - Maryam Al-Mansoob
- Department of Biological and Environmental Sciences, College of Arts and Sciences, Qatar University, Doha, 2713, Qatar
| | - Salma M.S. Ahmad
- Department of Biological and Environmental Sciences, College of Arts and Sciences, Qatar University, Doha, 2713, Qatar
| | | | - Ali H. Eid
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, 2713, Qatar
| | - Anna Maria Posadino
- Department of Biomedical Sciences, University of Sassari, 07100, Sassari, Italy
| | - Gianfranco Pintus
- Department of Biomedical Sciences, University of Sassari, 07100, Sassari, Italy
- Department of Medical Laboratory Sciences, College of Health Sciences and Sharjah Institute for Medical Research, University of Sharjah, University City Rd, Sharjah, 27272, United Arab Emirates
| | - Roberta Giordo
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, 505055, United Arab Emirates
| |
Collapse
|
27
|
Oka SI, Sreedevi K, Shankar TS, Yedla S, Arowa S, James A, Stone KG, Olmos K, Sabry AD, Horiuchi A, Cawley KM, O’very SA, Tong M, Byun J, Xu X, Kashyap S, Mourad Y, Vehra O, Calder D, Lunde T, Liu T, Li H, Mashchek JA, Cox J, Saijoh Y, Drakos SG, Warren JS. PERM1 regulates energy metabolism in the heart via ERRα/PGC-1α axis. Front Cardiovasc Med 2022; 9:1033457. [PMID: 36419485 PMCID: PMC9676655 DOI: 10.3389/fcvm.2022.1033457] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 10/11/2022] [Indexed: 11/09/2022] Open
Abstract
Aims PERM1 is a striated muscle-specific regulator of mitochondrial bioenergetics. We previously demonstrated that PERM1 is downregulated in the failing heart and that PERM1 positively regulates metabolic genes known as targets of the transcription factor ERRα and its coactivator PGC-1α in cultured cardiomyocytes. The aims of this study were to determine the effect of loss of PERM1 on cardiac function and energetics using newly generated Perm1-knockout (Perm1 -/-) mice and to investigate the molecular mechanisms of its transcriptional control. Methods and results Echocardiography showed that ejection fraction and fractional shortening were lower in Perm1 -/- mice than in wild-type mice (both p < 0.05), and the phosphocreatine-to-ATP ratio was decreased in Perm1 -/- hearts (p < 0.05), indicating reduced contractile function and energy reserves of the heart. Integrated proteomic and metabolomic analyses revealed downregulation of oxidative phosphorylation and upregulation of glycolysis and polyol pathways in Perm1 -/- hearts. To examine whether PERM1 regulates energy metabolism through ERRα, we performed co-immunoprecipitation assays, which showed that PERM1 bound to ERRα in cardiomyocytes and the mouse heart. DNA binding and reporter gene assays showed that PERM1 was localized to and activated the ERR target promoters partially through ERRα. Mass spectrometry-based screening in cardiomyocytes identified BAG6 and KANK2 as potential PERM1's binding partners in transcriptional regulation. Mammalian one-hybrid assay, in which PERM1 was fused to Gal4 DNA binding domain, showed that the recruitment of PERM1 to a gene promoter was sufficient to activate transcription, which was blunted by silencing of either PGC-1α, BAG6, or KANK2. Conclusion This study demonstrates that PERM1 is an essential regulator of cardiac energetics and function and that PERM1 is a novel transcriptional coactivator in the ERRα/PGC-1α axis that functionally interacts with BAG6 and KANK2.
Collapse
Affiliation(s)
- Shin-ichi Oka
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Newark, NJ, United States
| | - Karthi Sreedevi
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, VA, United States
| | - Thirupura S. Shankar
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, United States
| | - Shreya Yedla
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, VA, United States
| | - Sumaita Arowa
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, VA, United States
| | - Amina James
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, VA, United States
| | - Kathryn G. Stone
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, VA, United States
| | - Katia Olmos
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, VA, United States
| | - Amira D. Sabry
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, United States
| | - Amanda Horiuchi
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, United States
| | - Keiko M. Cawley
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, United States
| | - Sean A. O’very
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, United States
| | - Mingming Tong
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Newark, NJ, United States
| | - Jaemin Byun
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Newark, NJ, United States
| | - Xiaoyong Xu
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Newark, NJ, United States
| | - Sanchita Kashyap
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Newark, NJ, United States
| | - Youssef Mourad
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Newark, NJ, United States
| | - Omair Vehra
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Newark, NJ, United States
| | - Dallen Calder
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, United States
| | - Ty Lunde
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, United States
| | - Tong Liu
- Department of Microbiology, Biochemistry, and Molecular Genetics, Center for Advanced Proteomics Research, Rutgers New Jersey Medical School and Cancer Institute of New Jersey, Newark, NJ, United States
| | - Hong Li
- Department of Microbiology, Biochemistry, and Molecular Genetics, Center for Advanced Proteomics Research, Rutgers New Jersey Medical School and Cancer Institute of New Jersey, Newark, NJ, United States
| | - J. Alan Mashchek
- Metabolomics Core Research Facility, University of Utah, Salt Lake City, UT, United States
| | - James Cox
- Metabolomics Core Research Facility, University of Utah, Salt Lake City, UT, United States
- Department of Biochemistry, University of Utah, Salt Lake City, UT, United States
| | - Yukio Saijoh
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, United States
| | - Stavros G. Drakos
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, United States
- Division of Cardiovascular Medicine, University of Utah School of Medicine, Salt Lake City, UT, United States
| | - Junco S. Warren
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, VA, United States
- Center for Vascular and Heart Research, Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, VA, United States
- Department of Human Nutrition, Food and Exercise, Virginia Tech, Blacksburg, VA, United States
- Division of Developmental Genetics, Institute of Resource Developmental and Analysis, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
28
|
Katti P, Ajayi PT, Aponte A, Bleck CKE, Glancy B. Identification of evolutionarily conserved regulators of muscle mitochondrial network organization. Nat Commun 2022; 13:6622. [PMID: 36333356 PMCID: PMC9636386 DOI: 10.1038/s41467-022-34445-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Accepted: 10/25/2022] [Indexed: 11/06/2022] Open
Abstract
Mitochondrial networks provide coordinated energy distribution throughout muscle cells. However, pathways specifying mitochondrial networks are incompletely understood and it is unclear how they might affect contractile fiber-type. Here, we show that natural energetic demands placed on Drosophila melanogaster muscles yield native cell-types among which contractile and mitochondrial network-types are regulated differentially. Proteomic analyses of indirect flight, jump, and leg muscles, together with muscles misexpressing known fiber-type specification factor salm, identified transcription factors H15 and cut as potential mitochondrial network regulators. We demonstrate H15 operates downstream of salm regulating flight muscle contractile and mitochondrial network-type. Conversely, H15 regulates mitochondrial network configuration but not contractile type in jump and leg muscles. Further, we find that cut regulates salm expression in flight muscles and mitochondrial network configuration in leg muscles. These data indicate cell type-specific regulation of muscle mitochondrial network organization through evolutionarily conserved transcription factors cut, salm, and H15.
Collapse
Affiliation(s)
- Prasanna Katti
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Peter T Ajayi
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Angel Aponte
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Christopher K E Bleck
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Brian Glancy
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, 20892, USA.
- National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, 20892, USA.
| |
Collapse
|
29
|
Huang CY, Oka SI, Xu X, Chen CF, Tung CY, Chang YY, Mourad Y, Vehra O, Ivessa A, Yehia G, Romanienko P, Hsu CP, Sadoshima J. PERM1 regulates genes involved in fatty acid metabolism in the heart by interacting with PPARα and PGC-1α. Sci Rep 2022; 12:14576. [PMID: 36028747 PMCID: PMC9418182 DOI: 10.1038/s41598-022-18885-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 08/22/2022] [Indexed: 11/09/2022] Open
Abstract
PERM1 (PGC-1/ERR-induced regulator in muscle 1) is a muscle-specific protein induced by PGC-1 and ERRs. Previous studies have shown that PERM1 promotes mitochondrial biogenesis and metabolism in cardiomyocytes in vitro. However, the role of endogenous PERM1 in the heart remains to be investigated with loss-of-function studies in vivo. We report the generation and characterization of systemic Perm1 knockout (KO) mice. The baseline cardiac phenotype of the homozygous Perm1 KO mice appeared normal. However, RNA-sequencing and unbiased pathway analyses showed that homozygous downregulation of PERM1 leads to downregulation of genes involved in fatty acid and carbohydrate metabolism in the heart. Transcription factor binding site analyses suggested that PPARα and PGC-1α are involved in changes in the gene expression profile. Chromatin immunoprecipitation assays showed that PERM1 interacts with the proximal regions of PPAR response elements (PPREs) in endogenous promoters of genes involved in fatty acid oxidation. Co-immunoprecipitation and reporter gene assays showed that PERM1 promoted transcription via the PPRE, partly in a PPARα and PGC-1α dependent manner. These results suggest that endogenous PERM1 is involved in the transcription of genes involved in fatty acid oxidation through physical interaction with PPARα and PGC-1α in the heart in vivo.
Collapse
Affiliation(s)
- Chun-Yang Huang
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, 185 South Orange Ave., MSB G609, Newark, NJ, 07103, USA.,Division of Cardiovascular Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei, Taiwan.,Department of Medicine, School of Medicine, National Yang-Ming Chiao-Tung University, Taipei, Taiwan
| | - Shin-Ichi Oka
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, 185 South Orange Ave., MSB G609, Newark, NJ, 07103, USA
| | - Xiaoyong Xu
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, 185 South Orange Ave., MSB G609, Newark, NJ, 07103, USA.,Department of Cardiology, Ningbo Medical Center Lihuili Hospital, Ningbo, Zhejiang, China
| | - Chian-Feng Chen
- Cancer Progression Research Center, National Yang-Ming Chiao-Tung University, Taipei, Taiwan
| | - Chien-Yi Tung
- Cancer Progression Research Center, National Yang-Ming Chiao-Tung University, Taipei, Taiwan
| | - Ya-Yuan Chang
- Cancer Progression Research Center, National Yang-Ming Chiao-Tung University, Taipei, Taiwan
| | - Youssef Mourad
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, 185 South Orange Ave., MSB G609, Newark, NJ, 07103, USA
| | - Omair Vehra
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, 185 South Orange Ave., MSB G609, Newark, NJ, 07103, USA
| | - Andreas Ivessa
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, 185 South Orange Ave., MSB G609, Newark, NJ, 07103, USA
| | - Ghassan Yehia
- Genome Editing Core Facility, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, 08901, USA
| | - Peter Romanienko
- Genome Editing Core Facility, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, 08901, USA
| | - Chiao-Po Hsu
- Division of Cardiovascular Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei, Taiwan.,Department of Medicine, School of Medicine, National Yang-Ming Chiao-Tung University, Taipei, Taiwan
| | - Junichi Sadoshima
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, 185 South Orange Ave., MSB G609, Newark, NJ, 07103, USA.
| |
Collapse
|
30
|
Abbasian S, Ravasi AA, Haghighi AH, Aydin S, Delbari A, Aydın S. Preconditioning intensive training ameliorates reduction of transcription biofactors of PGC1α-pathway in paretic muscle due to cerebral ischemia. Biotech Histochem 2022; 98:46-53. [PMID: 35892280 DOI: 10.1080/10520295.2022.2098535] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022] Open
Abstract
Exercise training increases fibronectin type III domain-containing protein 5 (FNDC5/irisin) via the peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC1α)-pathway. The PGC1α pathway induced FNDC5/irisin changes in response to exercise training and ischemic stroke are not entirely understood. We investigated the relation of the PGC-1α/FNDC5/irisin pathway to exercise training and to the pathophysiology of ischemic stroke in paretic muscles of stroke-induced rat models. We induced cerebral ischemia following completion of high-intensity interval training (HIIT) to evaluate PGC1α-pathway biofactors in paretic muscles. To define the underlying molecular mechanisms for improvement in paretic muscles following cerebral ischemia, we evaluated PCG-1α-pathway factors using immunofluorescence tracking and enzyme-linked immunosorbent assay (ELISA) immunoassay. We found that HIIT for 3 weeks produced increased expression and release of PGC-1α-pathway biomarkers in both the serum and paretic muscle of stroke-induced rats. We also found a close relation between the expression of PCG-1α-pathway factors in skeletal muscle and their concentration in blood. We found that PGC-1α-pathway biomarkers cause irisin up-regulation following induction of cerebral ischemia. The reduction in neurofunctional deficits following increased PGC-1α-pathway biomarkers suggests that these factors may act as markers of improvement in paretic muscle healing following cerebral ischemia.
Collapse
Affiliation(s)
| | | | | | | | - Ahmad Delbari
- University of Social Welfare and Rehabilitation Sciences, Iran
| | | |
Collapse
|
31
|
Scholtes C, Giguère V. Transcriptional control of energy metabolism by nuclear receptors. Nat Rev Mol Cell Biol 2022; 23:750-770. [DOI: 10.1038/s41580-022-00486-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/08/2022] [Indexed: 12/11/2022]
|
32
|
Podkalicka P, Mucha O, Kaziród K, Szade K, Stępniewski J, Ivanishchuk L, Hirao H, Pośpiech E, Józkowicz A, Kupiec-Weglinski JW, Dulak J, Łoboda A. miR-378 affects metabolic disturbances in the mdx model of Duchenne muscular dystrophy. Sci Rep 2022; 12:3945. [PMID: 35273230 PMCID: PMC8913680 DOI: 10.1038/s41598-022-07868-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 02/22/2022] [Indexed: 02/08/2023] Open
Abstract
Although Duchenne muscular dystrophy (DMD) primarily affects muscle tissues, the alterations to systemic metabolism manifested in DMD patients contribute to the severe phenotype of this fatal disorder. We propose that microRNA-378a (miR-378) alters carbohydrate and lipid metabolism in dystrophic mdx mice. In our study, we utilized double knockout animals which lacked both dystrophin and miR-378 (mdx/miR-378-/-). RNA sequencing of the liver identified 561 and 194 differentially expressed genes that distinguished mdx versus wild-type (WT) and mdx/miR-378-/- versus mdx counterparts, respectively. Bioinformatics analysis predicted, among others, carbohydrate metabolism disorder in dystrophic mice, as functionally proven by impaired glucose tolerance and insulin sensitivity. The lack of miR-378 in mdx animals mitigated those effects with a faster glucose clearance in a glucose tolerance test (GTT) and normalization of liver glycogen levels. The absence of miR-378 also restored the expression of genes regulating lipid homeostasis, such as Acly, Fasn, Gpam, Pnpla3, and Scd1. In conclusion, we report for the first time that miR-378 loss results in increased systemic metabolism of mdx mice. Together with our previous finding, demonstrating alleviation of the muscle-related symptoms of DMD, we propose that the inhibition of miR-378 may represent a new strategy to attenuate the multifaceted symptoms of DMD.
Collapse
Affiliation(s)
- Paulina Podkalicka
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Krakow, 30-387, Kraków, Poland
| | - Olga Mucha
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Krakow, 30-387, Kraków, Poland
| | - Katarzyna Kaziród
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Krakow, 30-387, Kraków, Poland
| | - Krzysztof Szade
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Krakow, 30-387, Kraków, Poland
| | - Jacek Stępniewski
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Krakow, 30-387, Kraków, Poland
| | - Liudmyla Ivanishchuk
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Krakow, 30-387, Kraków, Poland
| | - Hirofumi Hirao
- The Dumont-UCLA Transplantation Center, Department of Surgery, Division of Liver and Pancreas Transplantation, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
| | - Ewelina Pośpiech
- Malopolska Centre of Biotechnology, Jagiellonian University in Krakow, 30-387, Kraków, Poland
| | - Alicja Józkowicz
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Krakow, 30-387, Kraków, Poland
| | - Jerzy W Kupiec-Weglinski
- The Dumont-UCLA Transplantation Center, Department of Surgery, Division of Liver and Pancreas Transplantation, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
| | - Józef Dulak
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Krakow, 30-387, Kraków, Poland
| | - Agnieszka Łoboda
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Krakow, 30-387, Kraków, Poland.
| |
Collapse
|
33
|
Taylor DF, Bishop DJ. Transcription Factor Movement and Exercise-Induced Mitochondrial Biogenesis in Human Skeletal Muscle: Current Knowledge and Future Perspectives. Int J Mol Sci 2022; 23:1517. [PMID: 35163441 PMCID: PMC8836245 DOI: 10.3390/ijms23031517] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 01/19/2022] [Accepted: 01/21/2022] [Indexed: 02/01/2023] Open
Abstract
In response to exercise, the oxidative capacity of mitochondria within skeletal muscle increases through the coordinated expression of mitochondrial proteins in a process termed mitochondrial biogenesis. Controlling the expression of mitochondrial proteins are transcription factors-a group of proteins that regulate messenger RNA transcription from DNA in the nucleus and mitochondria. To fulfil other functions or to limit gene expression, transcription factors are often localised away from DNA to different subcellular compartments and undergo rapid movement or accumulation only when required. Although many transcription factors involved in exercise-induced mitochondrial biogenesis have been identified, numerous conflicting findings and gaps exist within our knowledge of their subcellular movement. This review aims to summarise and provide a critical analysis of the published literature regarding the exercise-induced movement of transcription factors involved in mitochondria biogenesis in skeletal muscle.
Collapse
Affiliation(s)
| | - David J. Bishop
- Institute for Health and Sport (iHeS), Footscray Park, Victoria University, Melbourne 8001, Australia;
| |
Collapse
|
34
|
Saliu TP, Kumrungsee T, Miyata K, Tominaga H, Yazawa N, Hashimoto K, Kamesawa M, Yanaka N. Comparative study on molecular mechanism of diabetic myopathy in two different types of streptozotocin-induced diabetic models. Life Sci 2022; 288:120183. [PMID: 34848193 DOI: 10.1016/j.lfs.2021.120183] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 11/21/2021] [Accepted: 11/22/2021] [Indexed: 11/25/2022]
Abstract
AIMS Streptozotocin (STZ)-induced diabetic animal models have been widely used to study diabetic myopathy; however, non-specific cytotoxic effects of high-dose STZ have been discussed. The purpose of this study was to compare diabetic myopathy in a high-STZ model with another well-established STZ model with reduced cytotoxicity (high-fat diet (HFD) and low-dose STZ) and to identify mechanistic insights underlying diabetic myopathy in STZ models that can mimic perturbations observed in human patients with diabetic myopathy. MAIN METHODS Male C57BL6 mice were injected with a single high dose of STZ (180 mg/kg, High-STZ) or were given HFD plus low-dose STZ injection (STZ, 55 mg/kg/day, five consecutive days, HFD/STZ). We characterized diabetic myopathy by histological and immunochemical analyses and conducted gene expression analysis. KEY FINDINGS The high-STZ model showed a significant reduction in tibialis anterior myofiber size along with decreased satellite cell content and downregulation of inflammation response and collagen gene expression. Interestingly, blood corticosteroid levels were significantly increased in the high-STZ model, which was possibly related to lowered inflammation response-related gene expression. Further analyses using the HFD/STZ model showed downregulation of gene expression related to mitochondrial functions accompanied by a significant decrease in ATP levels in the muscles. SIGNIFICANCE The high-STZ model is suitable for studies regarding not only severe diabetic myopathy with excessive blood glucose but also negative impact of glucocorticoids on skeletal muscles. In contrast, the HFD/STZ model is characterized by higher immune responses and lower ATP production, which also reflects the pathologies observed in human diabetic patients.
Collapse
Affiliation(s)
- Tolulope Peter Saliu
- Graduate School of Integrated Sciences for Life, Hiroshima University, 4-4 Kagamiyama 1-chome, Higashi-Hiroshima 739-8528, Japan
| | - Thanutchaporn Kumrungsee
- Graduate School of Integrated Sciences for Life, Hiroshima University, 4-4 Kagamiyama 1-chome, Higashi-Hiroshima 739-8528, Japan.
| | - Kenshu Miyata
- Graduate School of Integrated Sciences for Life, Hiroshima University, 4-4 Kagamiyama 1-chome, Higashi-Hiroshima 739-8528, Japan
| | - Hikaru Tominaga
- Graduate School of Integrated Sciences for Life, Hiroshima University, 4-4 Kagamiyama 1-chome, Higashi-Hiroshima 739-8528, Japan
| | - Nao Yazawa
- Graduate School of Integrated Sciences for Life, Hiroshima University, 4-4 Kagamiyama 1-chome, Higashi-Hiroshima 739-8528, Japan
| | - Kotaro Hashimoto
- Graduate School of Integrated Sciences for Life, Hiroshima University, 4-4 Kagamiyama 1-chome, Higashi-Hiroshima 739-8528, Japan
| | - Mion Kamesawa
- Graduate School of Integrated Sciences for Life, Hiroshima University, 4-4 Kagamiyama 1-chome, Higashi-Hiroshima 739-8528, Japan
| | - Noriyuki Yanaka
- Graduate School of Integrated Sciences for Life, Hiroshima University, 4-4 Kagamiyama 1-chome, Higashi-Hiroshima 739-8528, Japan.
| |
Collapse
|
35
|
Nishida Y, Nawaz A, Hecht K, Tobe K. Astaxanthin as a Novel Mitochondrial Regulator: A New Aspect of Carotenoids, beyond Antioxidants. Nutrients 2021; 14:nu14010107. [PMID: 35010981 PMCID: PMC8746862 DOI: 10.3390/nu14010107] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 12/21/2021] [Accepted: 12/23/2021] [Indexed: 12/12/2022] Open
Abstract
Astaxanthin is a member of the carotenoid family that is found abundantly in marine organisms, and has been gaining attention in recent years due to its varied biological/physiological activities. It has been reported that astaxanthin functions both as a pigment, and as an antioxidant with superior free radical quenching capacity. We recently reported that astaxanthin modulated mitochondrial functions by a novel mechanism independent of its antioxidant function. In this paper, we review astaxanthin’s well-known antioxidant activity, and expand on astaxanthin’s lesser-known molecular targets, and its role in mitochondrial energy metabolism.
Collapse
Affiliation(s)
- Yasuhiro Nishida
- First Department of Internal Medicine, Faculty of Medicine, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan
- Fuji Chemical Industries, Co., Ltd., 55 Yokohoonji, Kamiich-machi, Nakaniikawa-gun, Toyama 930-0405, Japan
- Correspondence: (Y.N.); (A.N.); (K.T.)
| | - Allah Nawaz
- First Department of Internal Medicine, Faculty of Medicine, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan
- Department of Molecular and Medical Pharmacology, Faculty of Medicine, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan
- Correspondence: (Y.N.); (A.N.); (K.T.)
| | - Karen Hecht
- AstaReal, Inc., 3 Terri Lane, Unit 12, Burlington, NJ 08016, USA;
| | - Kazuyuki Tobe
- First Department of Internal Medicine, Faculty of Medicine, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan
- Correspondence: (Y.N.); (A.N.); (K.T.)
| |
Collapse
|
36
|
Effects of Metformin in Heart Failure: From Pathophysiological Rationale to Clinical Evidence. Biomolecules 2021; 11:biom11121834. [PMID: 34944478 PMCID: PMC8698925 DOI: 10.3390/biom11121834] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 11/26/2021] [Accepted: 12/01/2021] [Indexed: 12/20/2022] Open
Abstract
Type 2 diabetes mellitus (T2DM) is a worldwide major health burden and heart failure (HF) is the most common cardiovascular (CV) complication in affected patients. Therefore, identifying the best pharmacological approach for glycemic control, which is also useful to prevent and ameliorate the prognosis of HF, represents a crucial issue. Currently, the choice is between the new drugs sodium/glucose co-transporter 2 inhibitors that have consistently shown in large CV outcome trials (CVOTs) to reduce the risk of HF-related outcomes in T2DM, and metformin, an old medicament that might end up relegated to the background while exerting interesting protective effects on multiple organs among which include heart failure. When compared with other antihyperglycemic medications, metformin has been demonstrated to be safe and to lower morbidity and mortality for HF, even if these results are difficult to interpret as they emerged mainly from observational studies. Meta-analyses of randomized controlled clinical trials have not produced positive results on the risk or clinical course of HF and sadly, large CV outcome trials are lacking. The point of force of metformin with respect to new diabetic drugs is the amount of data from experimental investigations that, for more than twenty years, still continues to provide mechanistic explanations of the several favorable actions in heart failure such as, the improvement of the myocardial energy metabolic status by modulation of glucose and lipid metabolism, the attenuation of oxidative stress and inflammation, and the inhibition of myocardial cell apoptosis, leading to reduced cardiac remodeling and preserved left ventricular function. In the hope that specific large-scale trials will be carried out to definitively establish the metformin benefit in terms of HF failure outcomes, we reviewed the literature in this field, summarizing the available evidence from experimental and clinical studies reporting on effects in heart metabolism, function, and structure, and the prominent pathophysiological mechanisms involved.
Collapse
|
37
|
Bean C, Audano M, Varanita T, Favaretto F, Medaglia M, Gerdol M, Pernas L, Stasi F, Giacomello M, Herkenne S, Muniandy M, Heinonen S, Cazaly E, Ollikainen M, Milan G, Pallavicini A, Pietiläinen KH, Vettor R, Mitro N, Scorrano L. The mitochondrial protein Opa1 promotes adipocyte browning that is dependent on urea cycle metabolites. Nat Metab 2021; 3:1633-1647. [PMID: 34873337 DOI: 10.1038/s42255-021-00497-2] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 10/27/2021] [Indexed: 12/14/2022]
Abstract
White to brown/beige adipocytes conversion is a possible therapeutic strategy to tackle the current obesity epidemics. While mitochondria are key for energy dissipation in brown fat, it is unknown if they can drive adipocyte browning. Here, we show that the mitochondrial cristae biogenesis protein optic atrophy 1 (Opa1) facilitates cell-autonomous adipocyte browning. In two cohorts of patients with obesity, including weight discordant monozygotic twin pairs, adipose tissue OPA1 levels are reduced. In the mouse, Opa1 overexpression favours white adipose tissue expandability as well as browning, ultimately improving glucose tolerance and insulin sensitivity. Transcriptomics and metabolomics analyses identify the Jumanji family chromatin remodelling protein Kdm3a and urea cycle metabolites, including fumarate, as effectors of Opa1-dependent browning. Mechanistically, the higher cyclic adenosine monophosphate (cAMP) levels in Opa1 pre-adipocytes activate cAMP-responsive element binding protein (CREB), which transcribes urea cycle enzymes. Flux analyses in pre-adipocytes indicate that Opa1-dependent fumarate accumulation depends on the urea cycle. Conversely, adipocyte-specific Opa1 deletion curtails urea cycle and beige differentiation of pre-adipocytes, and is rescued by fumarate supplementation. Thus, the urea cycle links the mitochondrial dynamics protein Opa1 to white adipocyte browning.
Collapse
Affiliation(s)
- Camilla Bean
- Department of Biology, University of Padova, Padova, Italy
- Veneto Institute of Molecular Medicine, Padova, Italy
| | - Matteo Audano
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Tatiana Varanita
- Department of Biology, University of Padova, Padova, Italy
- Veneto Institute of Molecular Medicine, Padova, Italy
| | | | - Marta Medaglia
- Department of Biology, University of Padova, Padova, Italy
- Veneto Institute of Molecular Medicine, Padova, Italy
| | - Marco Gerdol
- Department of Life Science, University of Trieste, Trieste, Italy
| | - Lena Pernas
- Department of Biology, University of Padova, Padova, Italy
- Veneto Institute of Molecular Medicine, Padova, Italy
| | - Fabio Stasi
- Department of Medicine, University of Padova, Padova, Italy
| | | | - Stèphanie Herkenne
- Department of Biology, University of Padova, Padova, Italy
- Veneto Institute of Molecular Medicine, Padova, Italy
| | - Maheswary Muniandy
- Obesity Research Unit, Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Sini Heinonen
- Obesity Research Unit, Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Emma Cazaly
- Institute for Molecular Medicine Finland, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Miina Ollikainen
- Institute for Molecular Medicine Finland, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | | | | | - Kirsi H Pietiläinen
- Obesity Research Unit, Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Obesity Centre, Abdominal Centre, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Roberto Vettor
- Department of Medicine, University of Padova, Padova, Italy
| | - Nico Mitro
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Luca Scorrano
- Department of Biology, University of Padova, Padova, Italy.
- Veneto Institute of Molecular Medicine, Padova, Italy.
| |
Collapse
|
38
|
Napoli E, Flores A, Mansuri Y, Hagerman RJ, Giulivi C. Sulforaphane improves mitochondrial metabolism in fibroblasts from patients with fragile X-associated tremor and ataxia syndrome. Neurobiol Dis 2021; 157:105427. [PMID: 34153466 PMCID: PMC8475276 DOI: 10.1016/j.nbd.2021.105427] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 06/10/2021] [Accepted: 06/16/2021] [Indexed: 02/09/2023] Open
Abstract
CGG expansions between 55 and 200 in the 5'-untranslated region of the fragile-X mental retardation gene (FMR1) increase the risk of developing the late-onset debilitating neuromuscular disease Fragile X-Associated Tremor/Ataxia Syndrome (FXTAS). While the science behind this mutation, as a paradigm for RNA-mediated nucleotide triplet repeat expansion diseases, has progressed rapidly, no treatment has proven effective at delaying the onset or decreasing morbidity, especially at later stages of the disease. Here, we demonstrated the beneficial effect of the phytochemical sulforaphane (SFN), exerted through NRF2-dependent and independent manner, on pathways relevant to brain function, bioenergetics, unfolded protein response, proteosome, antioxidant defenses, and iron metabolism in fibroblasts from FXTAS-affected subjects at all disease stages. This study paves the way for future clinical studies with SFN in the treatment of FXTAS, substantiated by the established use of this agent in clinical trials of diseases with NRF2 dysregulation and in which age is the leading risk factor.
Collapse
Affiliation(s)
- Eleonora Napoli
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, CA 95616
| | - Amanda Flores
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, CA 95616;,Department of Biochemistry, Medical Sciences Campus, University of Puerto Rico, San Juan, Puerto Rico
| | - Yasmeen Mansuri
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, CA 95616
| | - Randi J. Hagerman
- Department of Pediatrics, University of California Davis Medical Center, Sacramento, CA;,Medical Investigations of Neurodevelopmental Disorders (M.I.N.D.) Institute, University of California Davis, CA 95817
| | - Cecilia Giulivi
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, CA 95616, United States of America; Medical Investigations of Neurodevelopmental Disorders (M.I.N.D.) Institute, University of California Davis, CA 95817, USA.
| |
Collapse
|
39
|
Bock T, Türk C, Aravamudhan S, Keufgens L, Bloch W, Rozsivalova DH, Romanello V, Nogara L, Blaauw B, Trifunovic A, Braun T, Krüger M. PERM1 interacts with the MICOS-MIB complex to connect the mitochondria and sarcolemma via ankyrin B. Nat Commun 2021; 12:4900. [PMID: 34385433 PMCID: PMC8361071 DOI: 10.1038/s41467-021-25185-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 07/20/2021] [Indexed: 12/16/2022] Open
Abstract
Skeletal muscle subsarcolemmal mitochondria (SSM) and intermyofibrillar mitochondria subpopulations have distinct metabolic activity and sensitivity, though the mechanisms that localize SSM to peripheral areas of muscle fibers are poorly understood. A protein interaction study and complexome profiling identifies PERM1 interacts with the MICOS-MIB complex. Ablation of Perm1 in mice reduces muscle force, decreases mitochondrial membrane potential and complex I activity, and reduces the numbers of SSM in skeletal muscle. We demonstrate PERM1 interacts with the intracellular adaptor protein ankyrin B (ANKB) that connects the cytoskeleton to the plasma membrane. Moreover, we identify a C-terminal transmembrane helix that anchors PERM1 into the outer mitochondrial membrane. We conclude PERM1 functions in the MICOS-MIB complex and acts as an adapter to connect the mitochondria with the sarcolemma via ANKB.
Collapse
Affiliation(s)
- Theresa Bock
- Institute for Genetics, Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Cologne, Germany
| | - Clara Türk
- BASF SE, Metabolomics and Proteomics, Ludwigshafen am Rhein, Germany
| | | | - Lena Keufgens
- Institute for Genetics, Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Cologne, Germany
| | - Wilhelm Bloch
- Department of Molecular and Cellular Sport Medicine, Institute of Sport Medicine and Cardiovascular Research, German Sport University Cologne, Cologne, Germany
| | - Dieu Hien Rozsivalova
- Institute for Genetics, Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Cologne, Germany
| | - Vanina Romanello
- Venetian Institute of Molecular Medicine (VIMM), Department of Biomedical Sciences Padova, University of Padova, Padova, Italy
| | - Leonardo Nogara
- Venetian Institute of Molecular Medicine (VIMM), Department of Biomedical Sciences Padova, University of Padova, Padova, Italy
| | - Bert Blaauw
- Venetian Institute of Molecular Medicine (VIMM), Department of Biomedical Sciences Padova, University of Padova, Padova, Italy
| | - Aleksandra Trifunovic
- Institute for Genetics, Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Cologne, Germany
- Center for Molecular Medicine (CMMC), University of Cologne, Cologne, Germany
| | - Thomas Braun
- Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Marcus Krüger
- Institute for Genetics, Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Cologne, Germany.
- Center for Molecular Medicine (CMMC), University of Cologne, Cologne, Germany.
| |
Collapse
|
40
|
Yang X, Chen Q, Ouyang Q, Rong P, Feng W, Quan C, Li M, Jiang Q, Liang H, Zhao TJ, Wang HY, Chen S. Tissue-Specific Splicing and Dietary Interaction of a Mutant As160 Allele Determine Muscle Metabolic Fitness in Rodents. Diabetes 2021; 70:1826-1842. [PMID: 33980689 DOI: 10.2337/db21-0039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Accepted: 05/06/2021] [Indexed: 11/13/2022]
Abstract
Ethnic groups are physiologically and genetically adapted to their diets. Inuit bear a frequent AS160R684X mutation that causes type 2 diabetes. Whether this mutation evolutionarily confers adaptation in Inuit and how it causes metabolic disorders upon dietary changes are unknown due to limitations in human studies. Here, we develop a genetically modified rat model bearing an orthologous AS160R693X mutation, which mimics human patients exhibiting postprandial hyperglycemia and hyperinsulinemia. Importantly, a sugar-rich diet aggravates metabolic abnormalities in AS160R693X rats. The AS160R693X mutation diminishes a dominant long-variant AS160 without affecting a minor short-variant AS160 in skeletal muscle, which suppresses muscle glucose utilization but induces fatty acid oxidation. This fuel switch suggests a possible adaptation in Inuit who traditionally had lipid-rich hypoglycemic diets. Finally, induction of the short-variant AS160 restores glucose utilization in rat myocytes and a mouse model. Our findings have implications for development of precision treatments for patients bearing the AS160R684X mutation.
Collapse
Affiliation(s)
- Xinyu Yang
- MOE Key Laboratory of Model Animal for Disease Study, Department of Endocrinology, Nanjing Drum Tower Hospital, and Model Animal Research Center, School of Medicine, Nanjing University, Nanjing
| | - Qiaoli Chen
- MOE Key Laboratory of Model Animal for Disease Study, Department of Endocrinology, Nanjing Drum Tower Hospital, and Model Animal Research Center, School of Medicine, Nanjing University, Nanjing
| | - Qian Ouyang
- MOE Key Laboratory of Model Animal for Disease Study, Department of Endocrinology, Nanjing Drum Tower Hospital, and Model Animal Research Center, School of Medicine, Nanjing University, Nanjing
| | - Ping Rong
- MOE Key Laboratory of Model Animal for Disease Study, Department of Endocrinology, Nanjing Drum Tower Hospital, and Model Animal Research Center, School of Medicine, Nanjing University, Nanjing
| | - Weikuan Feng
- MOE Key Laboratory of Model Animal for Disease Study, Department of Endocrinology, Nanjing Drum Tower Hospital, and Model Animal Research Center, School of Medicine, Nanjing University, Nanjing
| | - Chao Quan
- MOE Key Laboratory of Model Animal for Disease Study, Department of Endocrinology, Nanjing Drum Tower Hospital, and Model Animal Research Center, School of Medicine, Nanjing University, Nanjing
| | - Min Li
- MOE Key Laboratory of Model Animal for Disease Study, Department of Endocrinology, Nanjing Drum Tower Hospital, and Model Animal Research Center, School of Medicine, Nanjing University, Nanjing
| | - Qing Jiang
- Department of Sports Medicine and Adult Reconstructive Surgery, Drum Tower Hospital, School of Medicine, Nanjing University, China
| | - Hui Liang
- Department of General Surgery, First Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Tong-Jin Zhao
- Institute of Metabolism and Integrative Biology, Fudan University, Shanghai, China
| | - Hong Yu Wang
- MOE Key Laboratory of Model Animal for Disease Study, Department of Endocrinology, Nanjing Drum Tower Hospital, and Model Animal Research Center, School of Medicine, Nanjing University, Nanjing
| | - Shuai Chen
- MOE Key Laboratory of Model Animal for Disease Study, Department of Endocrinology, Nanjing Drum Tower Hospital, and Model Animal Research Center, School of Medicine, Nanjing University, Nanjing
| |
Collapse
|
41
|
Cho Y, Tachibana S, Lam K, Arita Y, Khosrowjerdi S, Zhang O, Liang A, Li R, Andreyev A, Murphy AN, Ross RS. Perm1 promotes cardiomyocyte mitochondrial biogenesis and protects against hypoxia/reoxygenation-induced damage in mice. J Biol Chem 2021; 297:100825. [PMID: 34029594 PMCID: PMC8214196 DOI: 10.1016/j.jbc.2021.100825] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 04/27/2021] [Accepted: 05/20/2021] [Indexed: 11/25/2022] Open
Abstract
Normal contractile function of the heart depends on a constant and reliable production of ATP by cardiomyocytes. Dysregulation of cardiac energy metabolism can result in immature heart development and disrupt the ability of the adult myocardium to adapt to stress, potentially leading to heart failure. Further, restoration of abnormal mitochondrial function can have beneficial effects on cardiac dysfunction. Previously, we identified a novel protein termed Perm1 (PGC-1 and estrogen-related receptor (ERR)-induced regulator, muscle 1) that is enriched in skeletal and cardiac-muscle mitochondria and transcriptionally regulated by PGC-1 (peroxisome proliferator-activated receptor gamma coactivator 1) and ERR. The role of Perm1 in the heart is poorly understood and is studied here. We utilized cell culture, mouse models, and human tissue, to study its expression and transcriptional control, as well as its role in transcription of other factors. Critically, we tested Perm1's role in cardiomyocyte mitochondrial function and its ability to protect myocytes from stress-induced damage. Our studies show that Perm1 expression increases throughout mouse cardiogenesis, demonstrate that Perm1 interacts with PGC-1α and enhances activation of PGC-1 and ERR, increases mitochondrial DNA copy number, and augments oxidative capacity in cultured neonatal mouse cardiomyocytes. Moreover, we found that Perm1 reduced cellular damage produced as a result of hypoxia and reoxygenation-induced stress and mitigated cell death of cardiomyocytes. Taken together, our results show that Perm1 promotes mitochondrial biogenesis in mouse cardiomyocytes. Future studies can assess the potential of Perm1 to be used as a novel therapeutic to restore cardiac dysfunction induced by ischemic injury.
Collapse
Affiliation(s)
- Yoshitake Cho
- Division of Cardiovascular Medicine, Department of Medicine, University of California San Diego, La Jolla, California, USA; Department of Medicine/Cardiology, Veterans Administration Healthcare, San Diego, California, USA.
| | - Shizuko Tachibana
- Division of Cardiovascular Medicine, Department of Medicine, University of California San Diego, La Jolla, California, USA
| | - Kayla Lam
- Division of Cardiovascular Medicine, Department of Medicine, University of California San Diego, La Jolla, California, USA
| | - Yoh Arita
- Department of Pharmacology, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, California, USA
| | - Shamim Khosrowjerdi
- Division of Cardiovascular Medicine, Department of Medicine, University of California San Diego, La Jolla, California, USA
| | - Oliver Zhang
- Division of Cardiovascular Medicine, Department of Medicine, University of California San Diego, La Jolla, California, USA
| | - Alex Liang
- Division of Cardiovascular Medicine, Department of Medicine, University of California San Diego, La Jolla, California, USA; Department of Medicine/Cardiology, Veterans Administration Healthcare, San Diego, California, USA
| | - Ruixia Li
- Division of Cardiovascular Medicine, Department of Medicine, University of California San Diego, La Jolla, California, USA; Department of Medicine/Cardiology, Veterans Administration Healthcare, San Diego, California, USA
| | - Aleksander Andreyev
- Department of Pharmacology, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, California, USA
| | - Anne N Murphy
- Department of Pharmacology, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, California, USA
| | - Robert S Ross
- Division of Cardiovascular Medicine, Department of Medicine, University of California San Diego, La Jolla, California, USA; Department of Medicine/Cardiology, Veterans Administration Healthcare, San Diego, California, USA
| |
Collapse
|
42
|
Blackburn ML, Wankhade UD, Ono-Moore KD, Chintapalli SV, Fox R, Rutkowsky JM, Willis BJ, Tolentino T, Lloyd KCK, Adams SH. On the potential role of globins in brown adipose tissue: a novel conceptual model and studies in myoglobin knockout mice. Am J Physiol Endocrinol Metab 2021; 321:E47-E62. [PMID: 33969705 PMCID: PMC8321818 DOI: 10.1152/ajpendo.00662.2020] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Myoglobin (Mb) regulates O2 bioavailability in muscle and heart as the partial pressure of O2 (Po2) drops with increased tissue workload. Globin proteins also modulate cellular NO pools, "scavenging" NO at higher Po2 and converting NO2- to NO as Po2 falls. Myoglobin binding of fatty acids may also signal a role in fat metabolism. Interestingly, Mb is expressed in brown adipose tissue (BAT), but its function is unknown. Herein, we present a new conceptual model that proposes links between BAT thermogenic activation, concurrently reduced Po2, and NO pools regulated by deoxy/oxy-globin toggling and xanthine oxidoreductase (XOR). We describe the effect of Mb knockout (Mb-/-) on BAT phenotype [lipid droplets, mitochondrial markers uncoupling protein 1 (UCP1) and cytochrome C oxidase 4 (Cox4), transcriptomics] in male and female mice fed a high-fat diet (HFD, 45% of energy, ∼13 wk), and examine Mb expression during brown adipocyte differentiation. Interscapular BAT weights did not differ by genotype, but there was a higher prevalence of mid-large sized droplets in Mb-/-. COX4 protein expression was significantly reduced in Mb-/- BAT, and a suite of metabolic/NO/stress/hypoxia transcripts were lower. All of these Mb-/--associated differences were most apparent in females. The new conceptual model, and results derived from Mb-/- mice, suggest a role for Mb in BAT metabolic regulation, in part through sexually dimorphic systems and NO signaling. This possibility requires further validation in light of significant mouse-to-mouse variability of BAT Mb mRNA and protein abundances in wild-type mice and lower expression relative to muscle and heart.NEW & NOTEWORTHY Myoglobin confers the distinct red color to muscle and heart, serving as an oxygen-binding protein in oxidative fibers. Less attention has been paid to brown fat, a thermogenic tissue that also expresses myoglobin. In a mouse knockout model lacking myoglobin, brown fat had larger fat droplets and lower markers of mitochondrial oxidative metabolism, especially in females. Gene expression patterns suggest a role for myoglobin as an oxygen/nitric oxide-sensor that regulates cellular metabolic and signaling pathways.
Collapse
Affiliation(s)
- Michael L Blackburn
- Arkansas Children's Nutrition Center, Little Rock, Arkansas
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Umesh D Wankhade
- Arkansas Children's Nutrition Center, Little Rock, Arkansas
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | | | - Sree V Chintapalli
- Arkansas Children's Nutrition Center, Little Rock, Arkansas
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Renee Fox
- Arkansas Children's Nutrition Center, Little Rock, Arkansas
| | - Jennifer M Rutkowsky
- Department of Molecular Biosciences, UC Davis School of Veterinary Medicine, University of California, Davis, California
- Mouse Metabolic Phenotyping Center, University of California, Davis, California
| | - Brandon J Willis
- Mouse Biology Program, University of California, Davis, California
| | - Todd Tolentino
- Mouse Metabolic Phenotyping Center, University of California, Davis, California
- Mouse Biology Program, University of California, Davis, California
| | - K C Kent Lloyd
- Mouse Metabolic Phenotyping Center, University of California, Davis, California
- Mouse Biology Program, University of California, Davis, California
- Department of Surgery, University of California Davis School of Medicine, Sacramento, California
| | - Sean H Adams
- Arkansas Children's Nutrition Center, Little Rock, Arkansas
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, Arkansas
- Department of Surgery, University of California Davis School of Medicine, Sacramento, California
- Center for Alimentary and Metabolic Science, University of California Davis School of Medicine, Sacramento, California
| |
Collapse
|
43
|
Skeletal Muscle Metabolism: Origin or Prognostic Factor for Amyotrophic Lateral Sclerosis (ALS) Development? Cells 2021; 10:cells10061449. [PMID: 34207859 PMCID: PMC8226541 DOI: 10.3390/cells10061449] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Revised: 06/03/2021] [Accepted: 06/07/2021] [Indexed: 12/26/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease characterized by progressive and selective loss of motor neurons, amyotrophy and skeletal muscle paralysis usually leading to death due to respiratory failure. While generally considered an intrinsic motor neuron disease, data obtained in recent years, including our own, suggest that motor neuron protection is not sufficient to counter the disease. The dismantling of the neuromuscular junction is closely linked to chronic energy deficit found throughout the body. Metabolic (hypermetabolism and dyslipidemia) and mitochondrial alterations described in patients and murine models of ALS are associated with the development and progression of disease pathology and they appear long before motor neurons die. It is clear that these metabolic changes participate in the pathology of the disease. In this review, we summarize these changes seen throughout the course of the disease, and the subsequent impact of glucose–fatty acid oxidation imbalance on disease progression. We also highlight studies that show that correcting this loss of metabolic flexibility should now be considered a major goal for the treatment of ALS.
Collapse
|
44
|
Random errors in protein synthesis activate an age-dependent program of muscle atrophy in mice. Commun Biol 2021; 4:703. [PMID: 34103648 PMCID: PMC8187632 DOI: 10.1038/s42003-021-02204-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 05/12/2021] [Indexed: 12/13/2022] Open
Abstract
Random errors in protein synthesis are prevalent and ubiquitous, yet their effect on organismal health has remained enigmatic for over five decades. Here, we studied whether mice carrying the ribosomal ambiguity (ram) mutation Rps2-A226Y, recently shown to increase the inborn error rate of mammalian translation, if at all viable, present any specific, possibly aging-related, phenotype. We introduced Rps2-A226Y using a Cre/loxP strategy. Resulting transgenic mice were mosaic and showed a muscle-related phenotype with reduced grip strength. Analysis of gene expression in skeletal muscle using RNA-Seq revealed transcriptomic changes occurring in an age-dependent manner, involving an interplay of PGC1α, FOXO3, mTOR, and glucocorticoids as key signaling pathways, and finally resulting in activation of a muscle atrophy program. Our results highlight the relevance of translation accuracy, and show how disturbances thereof may contribute to age-related pathologies. By introducing a ribosomal ambiguity mutation into mice, Moore et al. establish an in-vivo model to investigate how age-related diseases are related to decreasing accuracy in protein synthesis. Their findings potentially offer new insights into the pathological changes observed in age-related diseases, such as muscle atrophy
Collapse
|
45
|
Carmona-Mora P, Ander BP, Jickling GC, Dykstra-Aiello C, Zhan X, Ferino E, Hamade F, Amini H, Hull H, Sharp FR, Stamova B. Distinct peripheral blood monocyte and neutrophil transcriptional programs following intracerebral hemorrhage and different etiologies of ischemic stroke. J Cereb Blood Flow Metab 2021; 41:1398-1416. [PMID: 32960689 PMCID: PMC8142129 DOI: 10.1177/0271678x20953912] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 07/07/2020] [Accepted: 07/29/2020] [Indexed: 12/25/2022]
Abstract
Understanding cell-specific transcriptome responses following intracerebral hemorrhage (ICH) and ischemic stroke (IS) will improve knowledge of the immune response to brain injury. Transcriptomic profiles of 141 samples from 48 subjects with ICH, different IS etiologies, and vascular risk factor controls were characterized using RNA-seq in isolated neutrophils, monocytes and whole blood. In both IS and ICH, monocyte genes were down-regulated, whereas neutrophil gene expression changes were generally up-regulated. The monocyte down-regulated response to ICH included innate, adaptive immune, dendritic, NK cell and atherosclerosis signaling. Neutrophil responses to ICH included tRNA charging, mitochondrial dysfunction, and ER stress pathways. Common monocyte and neutrophil responses to ICH included interferon signaling, neuroinflammation, death receptor signaling, and NFAT pathways. Suppressed monocyte responses to IS included interferon and dendritic cell maturation signaling, phagosome formation, and IL-15 signaling. Activated neutrophil responses to IS included oxidative phosphorylation, mTOR, BMP, growth factor signaling, and calpain proteases-mediated blood-brain barrier (BBB) dysfunction. Common monocyte and neutrophil responses to IS included JAK1, JAK3, STAT3, and thrombopoietin signaling. Cell-type and cause-specific approaches will assist the search for future IS and ICH biomarkers and treatments.
Collapse
Affiliation(s)
- Paulina Carmona-Mora
- Department of Neurology, School of Medicine, University of California, Davis, Sacramento, CA, USA
| | - Bradley P Ander
- Department of Neurology, School of Medicine, University of California, Davis, Sacramento, CA, USA
| | - Glen C Jickling
- Department of Neurology, School of Medicine, University of California, Davis, Sacramento, CA, USA
- Department of Medicine, University of Alberta, Edmonton, Canada
| | - Cheryl Dykstra-Aiello
- Department of Neurology, School of Medicine, University of California, Davis, Sacramento, CA, USA
| | - Xinhua Zhan
- Department of Neurology, School of Medicine, University of California, Davis, Sacramento, CA, USA
| | - Eva Ferino
- Department of Neurology, School of Medicine, University of California, Davis, Sacramento, CA, USA
| | - Farah Hamade
- Department of Neurology, School of Medicine, University of California, Davis, Sacramento, CA, USA
| | - Hajar Amini
- Department of Neurology, School of Medicine, University of California, Davis, Sacramento, CA, USA
| | - Heather Hull
- Department of Neurology, School of Medicine, University of California, Davis, Sacramento, CA, USA
| | - Frank R Sharp
- Department of Neurology, School of Medicine, University of California, Davis, Sacramento, CA, USA
| | - Boryana Stamova
- Department of Neurology, School of Medicine, University of California, Davis, Sacramento, CA, USA
| |
Collapse
|
46
|
Weng K, Huo W, Gu T, Bao Q, Cao Z, Zhang Y, Zhang Y, Xu Q, Chen G. Quantitative phosphoproteomic analysis unveil the effect of marketable ages on meat quality in geese. Food Chem 2021; 361:130093. [PMID: 34029893 DOI: 10.1016/j.foodchem.2021.130093] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 05/09/2021] [Accepted: 05/10/2021] [Indexed: 10/21/2022]
Abstract
The quality of poultry goose meat is closely related to its marketable ages, with meat quality varying with increasing marketable age. Geese of two marketable ages (70-day and 120-day) were selected to understand the mechanisms behind this effect. Darker and redder meat; chewier and higher water-holding capacity (WHC) as well as greater protein and intramuscular fat (IMF) content were observed in the breast muscle (BM) of 120-day-old geese as compared to 70-day-old geese. Quantitative phosphoproteomics revealed up-regulated phosphorylated myofibrillar proteins and glycolytic enzymes in 120BM contributed to chewier meat with higher WHC. Redder meat might be attributed to phosphorylated mitochondrial proteins interacting with glycolytic enzymes in energy metabolism. Additionally, phosphorylation of PLIN1 and PERM1 might positively affect IMF deposition. Taken together, these data provided a phosphoproteomics perspective for the effect of marketable ages on meat quality and a theoretical strategy for improving meat quality in geese of younger marketable age.
Collapse
Affiliation(s)
- Kaiqi Weng
- Jiangsu Key Laboratory for Animal Genetic, Breeding and Molecular Design, Yangzhou University, Yangzhou, Jiangsu, China
| | - Weiran Huo
- Jiangsu Key Laboratory for Animal Genetic, Breeding and Molecular Design, Yangzhou University, Yangzhou, Jiangsu, China
| | - Tiantian Gu
- Jiangsu Key Laboratory for Animal Genetic, Breeding and Molecular Design, Yangzhou University, Yangzhou, Jiangsu, China
| | - Qiang Bao
- Jiangsu Key Laboratory for Animal Genetic, Breeding and Molecular Design, Yangzhou University, Yangzhou, Jiangsu, China
| | - Zhengfeng Cao
- Jiangsu Key Laboratory for Animal Genetic, Breeding and Molecular Design, Yangzhou University, Yangzhou, Jiangsu, China
| | - Yu Zhang
- Jiangsu Key Laboratory for Animal Genetic, Breeding and Molecular Design, Yangzhou University, Yangzhou, Jiangsu, China
| | - Yang Zhang
- Jiangsu Key Laboratory for Animal Genetic, Breeding and Molecular Design, Yangzhou University, Yangzhou, Jiangsu, China
| | - Qi Xu
- Jiangsu Key Laboratory for Animal Genetic, Breeding and Molecular Design, Yangzhou University, Yangzhou, Jiangsu, China.
| | - Guohong Chen
- Jiangsu Key Laboratory for Animal Genetic, Breeding and Molecular Design, Yangzhou University, Yangzhou, Jiangsu, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Jiangsu, Yangzhou, China.
| |
Collapse
|
47
|
Rahman FA, Quadrilatero J. Mitochondrial network remodeling: an important feature of myogenesis and skeletal muscle regeneration. Cell Mol Life Sci 2021; 78:4653-4675. [PMID: 33751143 PMCID: PMC11072563 DOI: 10.1007/s00018-021-03807-9] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 02/23/2021] [Accepted: 03/03/2021] [Indexed: 12/13/2022]
Abstract
The remodeling of the mitochondrial network is a critical process in maintaining cellular homeostasis and is intimately related to mitochondrial function. The interplay between the formation of new mitochondria (biogenesis) and the removal of damaged mitochondria (mitophagy) provide a means for the repopulation of the mitochondrial network. Additionally, mitochondrial fission and fusion serve as a bridge between biogenesis and mitophagy. In recent years, the importance of these processes has been characterised in multiple tissue- and cell-types, and under various conditions. In skeletal muscle, the robust remodeling of the mitochondrial network is observed, particularly after injury where large portions of the tissue/cell structures are damaged. The significance of mitochondrial remodeling in regulating skeletal muscle regeneration has been widely studied, with alterations in mitochondrial remodeling processes leading to incomplete regeneration and impaired skeletal muscle function. Needless to say, important questions related to mitochondrial remodeling and skeletal muscle regeneration still remain unanswered and require further investigation. Therefore, this review will discuss the known molecular mechanisms of mitochondrial network remodeling, as well as integrate these mechanisms and discuss their relevance in myogenesis and regenerating skeletal muscle.
Collapse
Affiliation(s)
- Fasih Ahmad Rahman
- Department of Kinesiology, University of Waterloo, Waterloo, ON, N2L 3G1, Canada
| | - Joe Quadrilatero
- Department of Kinesiology, University of Waterloo, Waterloo, ON, N2L 3G1, Canada.
| |
Collapse
|
48
|
Queiroz AL, Lessard SJ, Ouchida AT, Araujo HN, Gonçalves DA, Simões Fróes Guimarães DSP, Teodoro BG, So K, Espreafico EM, Hirshman MF, Alberici LC, Kettelhut IDC, Goodyear LJ, Silveira LR. The MicroRNA miR-696 is regulated by SNARK and reduces mitochondrial activity in mouse skeletal muscle through Pgc1α inhibition. Mol Metab 2021; 51:101226. [PMID: 33812060 PMCID: PMC8121711 DOI: 10.1016/j.molmet.2021.101226] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 03/16/2021] [Accepted: 03/25/2021] [Indexed: 11/16/2022] Open
Abstract
OBJECTIVE MicroRNAs (miRNA) are known to regulate the expression of genes involved in several physiological processes including metabolism, mitochondrial biogenesis, proliferation, differentiation, and cell death. METHODS Using "in silico" analyses, we identified 219 unique miRNAs that potentially bind to the 3'UTR region of a critical mitochondrial regulator, the peroxisome proliferator-activated receptor gamma coactivator (PGC) 1 alpha (Pgc1α). Of the 219 candidate miRNAs, miR-696 had one of the highest interactions at the 3'UTR of Pgc1α, suggesting that miR-696 may be involved in the regulation of Pgc1α. RESULTS Consistent with this hypothesis, we found that miR-696 was highly expressed in the skeletal muscle of STZ-induced diabetic mice and chronic high-fat-fed mice. C2C12 muscle cells exposed to palmitic acid also exhibited a higher expression of miR-696. This increased expression corresponded with a reduced expression of oxidative metabolism genes and reduced mitochondrial respiration. Importantly, reducing miR-696 reversed decreases in mitochondrial activity in response to palmitic acid. Using C2C12 cells treated with the AMP-activated protein kinase (AMPK) activator AICAR and skeletal muscle from AMPKα2 dominant-negative (DN) mice, we found that the signaling mechanism regulating miR-696 did not involve AMPK. In contrast, overexpression of SNF1-AMPK-related kinase (SNARK) in C2C12 cells increased miR-696 transcription while knockdown of SNARK significantly decreased miR-696. Moreover, muscle-specific transgenic mice overexpressing SNARK exhibited a lower expression of Pgc1α, elevated levels of miR-696, and reduced amounts of spontaneous activity. CONCLUSIONS Our findings demonstrate that metabolic stress increases miR-696 expression in skeletal muscle cells, which in turn inhibits Pgc1α, reducing mitochondrial function. SNARK plays a role in this process as a metabolic stress signaling molecule inducing the expression of miR-696.
Collapse
Affiliation(s)
- André L Queiroz
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, USP, Ribeirão Preto, Brazil; Research Division, Joslin Diabetes Center, and Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Sarah J Lessard
- Research Division, Joslin Diabetes Center, and Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Amanda T Ouchida
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, USP, Ribeirão Preto, Brazil
| | - Hygor N Araujo
- Obesity and Comorbidities Research Center, OCRC, IB, UNICAMP, Campinas, Brazil
| | - Dawit A Gonçalves
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, USP, Ribeirão Preto, Brazil
| | | | - Bruno G Teodoro
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, USP, Ribeirão Preto, Brazil; Department of Physics and Chemistry, Faculty of Pharmaceutical Science, USP, Ribeirão Preto, Brazil
| | - Kawai So
- Research Division, Joslin Diabetes Center, and Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Enilza M Espreafico
- Department of Cell Biology, Ribeirão Preto Medical School, USP, Ribeirão Preto, Brazil
| | - Michael F Hirshman
- Research Division, Joslin Diabetes Center, and Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Luciane C Alberici
- Department of Physics and Chemistry, Faculty of Pharmaceutical Science, USP, Ribeirão Preto, Brazil
| | - Isis do Carmo Kettelhut
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, USP, Ribeirão Preto, Brazil
| | - Laurie J Goodyear
- Research Division, Joslin Diabetes Center, and Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.
| | - Leonardo R Silveira
- Obesity and Comorbidities Research Center, OCRC, IB, UNICAMP, Campinas, Brazil.
| |
Collapse
|
49
|
Núñez Y, Radović Č, Savić R, García-Casco JM, Čandek-Potokar M, Benítez R, Radojković D, Lukić M, Gogić M, Muñoz M, Fontanesi L, Óvilo C. Muscle Transcriptome Analysis Reveals Molecular Pathways Related to Oxidative Phosphorylation, Antioxidant Defense, Fatness and Growth in Mangalitsa and Moravka Pigs. Animals (Basel) 2021; 11:ani11030844. [PMID: 33809803 PMCID: PMC8002519 DOI: 10.3390/ani11030844] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 03/04/2021] [Accepted: 03/13/2021] [Indexed: 12/28/2022] Open
Abstract
This work was aimed at evaluating loin transcriptome and metabolic pathway differences between the two main Serbian local pig breeds with divergent characteristics regarding muscle growth and fatness, as well as exploring nutrigenomic effects of tannin supplementation in Mangalitsa (MA) pigs. The study comprised 24 Mangalitsa and 10 Moravka (MO) males, which were kept under identical management conditions. Mangalitsa animals were divided in two nutritional groups (n = 12) receiving a standard (control) or tannin-supplemented diet (1.5%; MAT). Moravka pigs were fed the standard mixture. All animals were slaughtered at a similar age; 120 kg of average live weight (LW) and loin tissue was used for RNA-seq analysis. Results showed 306 differentially expressed genes (DEGs) according to breed, enriched in genes involved in growth, lipid metabolism, protein metabolism and muscle development, such as PDK4, FABP4, MYOD1 and STAT3, as well as a relevant number of genes involved in mitochondrial respiratory activity (MT-NDs, NDUFAs among others). Oxidative phosphorylation was the most significantly affected pathway, activated in Mangalitsa muscle, revealing the basis of a different muscle metabolism. Also, many other relevant pathways were affected by breed and involved in oxidative stress response, fat accumulation and development of skeletal muscle. Results also allowed the identification of potential regulators and causal networks such as those controlled by FLCN, PPARGC1A or PRKAB1 with relevant regulatory roles on DEGs involved in mitochondrial and lipid metabolism, or IL3 and TRAF2 potentially controlling DEGs involved in muscle development. The Tannin effect on transcriptome was small, with only 23 DEGs, but included interesting ones involved in lipid deposition such as PPARGC1B. The results indicate a significant effect of the breed on muscle tissue gene expression, affecting relevant biological pathways and allowing the identification of strong regulatory candidate genes to underlie the gene expression and phenotypic differences between the compared groups.
Collapse
Affiliation(s)
- Yolanda Núñez
- Departamento de Mejora Genética Animal, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA), 28040 Madrid, Spain; (Y.N.); (J.M.G.-C.); (R.B.); (M.M.)
| | - Čedomir Radović
- Institute for Animal Husbandry, 11080 Belgrade, Serbia; (Č.R.); (M.L.); (M.G.)
| | - Radomir Savić
- Faculty of Agriculture, University of Belgrade, 11080 Belgrade, Serbia; (R.S.); (D.R.)
| | - Juan M. García-Casco
- Departamento de Mejora Genética Animal, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA), 28040 Madrid, Spain; (Y.N.); (J.M.G.-C.); (R.B.); (M.M.)
| | | | - Rita Benítez
- Departamento de Mejora Genética Animal, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA), 28040 Madrid, Spain; (Y.N.); (J.M.G.-C.); (R.B.); (M.M.)
| | - Dragan Radojković
- Faculty of Agriculture, University of Belgrade, 11080 Belgrade, Serbia; (R.S.); (D.R.)
| | - Miloš Lukić
- Institute for Animal Husbandry, 11080 Belgrade, Serbia; (Č.R.); (M.L.); (M.G.)
| | - Marija Gogić
- Institute for Animal Husbandry, 11080 Belgrade, Serbia; (Č.R.); (M.L.); (M.G.)
| | - María Muñoz
- Departamento de Mejora Genética Animal, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA), 28040 Madrid, Spain; (Y.N.); (J.M.G.-C.); (R.B.); (M.M.)
| | - Luca Fontanesi
- Department of Agricultural and Food Sciences, University of Bologna, 40126 Bologna, Italy;
| | - Cristina Óvilo
- Departamento de Mejora Genética Animal, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA), 28040 Madrid, Spain; (Y.N.); (J.M.G.-C.); (R.B.); (M.M.)
- Correspondence: ; Tel.: +34-913471492
| |
Collapse
|
50
|
Aravamudhan S, Türk C, Bock T, Keufgens L, Nolte H, Lang F, Krishnan RK, König T, Hammerschmidt P, Schindler N, Brodesser S, Rozsivalova DH, Rugarli E, Trifunovic A, Brüning J, Langer T, Braun T, Krüger M. Phosphoproteomics of the developing heart identifies PERM1 - An outer mitochondrial membrane protein. J Mol Cell Cardiol 2021; 154:41-59. [PMID: 33549681 DOI: 10.1016/j.yjmcc.2021.01.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 01/05/2021] [Accepted: 01/11/2021] [Indexed: 12/15/2022]
Abstract
Heart development relies on PTMs that control cardiomyocyte proliferation, differentiation and cardiac morphogenesis. We generated a map of phosphorylation sites during the early stages of cardiac postnatal development in mice; we quantified over 10,000 phosphorylation sites and 5000 proteins that were assigned to different pathways. Analysis of mitochondrial proteins led to the identification of PGC-1- and ERR-induced regulator in muscle 1 (PERM1), which is specifically expressed in skeletal muscle and heart tissue and associates with the outer mitochondrial membrane. We demonstrate PERM1 is subject to rapid changes mediated by the UPS through phosphorylation of its PEST motif by casein kinase 2. Ablation of Perm1 in mice results in reduced protein expression of lipin-1 accompanied by accumulation of specific phospholipid species. Isolation of Perm1-deficient mitochondria revealed significant downregulation of mitochondrial transport proteins for amino acids and carnitines, including SLC25A12/13/29/34 and CPT2. Consistently, we observed altered levels of various lipid species, amino acids, and acylcarnitines in Perm1-/- mitochondria. We conclude that the outer mitochondrial membrane protein PERM1 regulates homeostasis of lipid and amino acid metabolites in mitochondria.
Collapse
Affiliation(s)
| | - Clara Türk
- CECAD Research Center, Institute for Genetics, University of Cologne, Joseph-Stelzmann-Str. 26, 50931 Cologne, Germany
| | - Theresa Bock
- CECAD Research Center, Institute for Genetics, University of Cologne, Joseph-Stelzmann-Str. 26, 50931 Cologne, Germany
| | - Lena Keufgens
- CECAD Research Center, Institute for Genetics, University of Cologne, Joseph-Stelzmann-Str. 26, 50931 Cologne, Germany
| | - Hendrik Nolte
- Max Planck Institute for Biology of Ageing, 50931 Cologne, Germany
| | - Franziska Lang
- TRON - Translational Oncology at the University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany
| | - Ramesh Kumar Krishnan
- Excellence Cluster Cardio-Pulmonary Institute (CPI), Aulweg 130, 35392 Giessen, Germany
| | - Tim König
- Montreal Neurological Institute, McGill University, 3801 University Street, H3A 2B4 Montreal, QC, Canada
| | - Philipp Hammerschmidt
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931 Cologne, Germany
| | - Natalie Schindler
- Institut für Entwicklungsbiologie und Neurobiologie (IDN), Fachbereich Biologie (FB 10), Johannes Gutenberg University (JGU) Mainz, Germany c/o Institute of Molecular Biology gGmbH (IMB), Ackermannweg 4, 55128 Mainz, Germany
| | - Susanne Brodesser
- CECAD Research Center, Institute for Genetics, University of Cologne, Joseph-Stelzmann-Str. 26, 50931 Cologne, Germany
| | - Dieu Hien Rozsivalova
- CECAD Research Center, Institute for Genetics, University of Cologne, Joseph-Stelzmann-Str. 26, 50931 Cologne, Germany
| | - Elena Rugarli
- CECAD Research Center, Institute for Genetics, University of Cologne, Joseph-Stelzmann-Str. 26, 50931 Cologne, Germany
| | - Aleksandra Trifunovic
- CECAD Research Center, Institute for Genetics, University of Cologne, Joseph-Stelzmann-Str. 26, 50931 Cologne, Germany
| | - Jens Brüning
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931 Cologne, Germany
| | - Thomas Langer
- Max Planck Institute for Biology of Ageing, 50931 Cologne, Germany
| | - Thomas Braun
- Max Planck Institute for Heart and Lung Research, Ludwigstr. 43, 61231 Bad Nauheim, Germany
| | - Marcus Krüger
- CECAD Research Center, Institute for Genetics, University of Cologne, Joseph-Stelzmann-Str. 26, 50931 Cologne, Germany; Center for Molecular Medicine (CMMC), University of Cologne, 50931 Cologne, Germany.
| |
Collapse
|