1
|
Li C, Sun J, Ling H, Qin Z, Zhu X, Fu X, Li T, Gan X. MCU regulating bone remodeling and osteogenic function through mitochondrial calcium homeostasis and oxidative stress alteration. Free Radic Biol Med 2025:S0891-5849(25)00276-X. [PMID: 40389181 DOI: 10.1016/j.freeradbiomed.2025.05.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2025] [Accepted: 05/03/2025] [Indexed: 05/21/2025]
Abstract
Mitochondria regulate cellular metabolism, signaling, immunity, and differentiation, affecting various diseases including diabetes, cancer, and osteoporosis. Mitochondrial calcium uniporter (MCU) unidirectionally transports Ca2+ across the inner mitochondrial membrane to the mitochondrial matrix. Ca2+ shuttles continuously through mitochondria and plays an essential role in the regulation of several cellular processes. However, the effects of MCU on bone metabolism-related diseases and the underlying pathophysiologic mechanisms have not been fully elucidated. Here, we found that MCU overexpression led to reduced bone volume fraction, bone trabecular thickness and number in mice in vivo. That was associated with MCU-mediated oxidative stress, overload of mitochondrial calcium, and decrease in mitochondrial membrane potential in osteoblasts. In vitro, MCU activation inhibited osteogenic differentiation capacity of osteoblasts in both early and late stages of osteogenesis. Additionally, MCU opening negatively regulated BMP/Smad signaling through ROS accumulation, thereby inhibiting osteogenic differentiation. Our results provide promising new targets for the future development of osteogenic function regulation and tissue engineering applications.
Collapse
Affiliation(s)
- Chen Li
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases,West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Jiyu Sun
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases,West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Huiling Ling
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases,West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Ziqi Qin
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases,West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Xiting Zhu
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases,West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Xinliang Fu
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases,West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Tao Li
- Laboratory of Mitochondria and Metabolism, West China Hospital of Sichuan University, Chengdu, 610041, China; Department of Anesthesiology, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital of Sichuan University, Chengdu, 610041, China; West China Hospital, Sichuan University, No 37 Wainan Guoxue Road, Chengdu, 610041, Sichuan, China.
| | - Xueqi Gan
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases,West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
2
|
Dong M, Maturana AD. Effects of aging on calcium channels in skeletal muscle. Front Mol Biosci 2025; 12:1558456. [PMID: 40177518 PMCID: PMC11961898 DOI: 10.3389/fmolb.2025.1558456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Accepted: 02/27/2025] [Indexed: 04/05/2025] Open
Abstract
In skeletal muscle, calcium is not only essential to stimulate and sustain their contractions but also for muscle embryogenesis, regeneration, energy production in mitochondria, and fusion. Different ion channels contribute to achieving the various functions of calcium in skeletal muscles. Muscle contraction is initiated by releasing calcium from the sarcoplasmic reticulum through the ryanodine receptor channels gated mechanically by four dihydropyridine receptors of T-tubules. The calcium influx through store-operated calcium channels sustains the contraction and stimulates muscle regeneration. Mitochondrial calcium uniporter allows the calcium entry into mitochondria to stimulate oxidative phosphorylation. Aging alters the expression and activity of these different calcium channels, resulting in a reduction of skeletal muscle force generation and regeneration capacity. Regular physical training and bioactive molecules from nutrients can prevent the effects of aging on calcium channels. This review focuses on the current knowledge of the effects of aging on skeletal muscles' calcium channels.
Collapse
Affiliation(s)
| | - Andrés Daniel Maturana
- Department of Applied Biosciences, Graduate School of Bioagricultural Science, Nagoya University, Nagoya, Japan
| |
Collapse
|
3
|
Umek N, Meznarič M, Šink Ž, Blagotinšek Cokan K, Prosenc Zmrzljak U, Horvat S. In situ spatial transcriptomic analysis of human skeletal muscle using the Xenium platform. Cell Tissue Res 2025; 399:291-302. [PMID: 39786556 DOI: 10.1007/s00441-024-03945-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Accepted: 12/03/2024] [Indexed: 01/12/2025]
Abstract
Traditional transcriptomic studies often overlook the complex heterogeneity of skeletal muscle, as they typically isolate RNA from mixed muscle fibre and cell populations, resulting in an averaged transcriptomic profile that obscures fibre type-specific differences. This study assessed the potential of the recently developed Xenium platform for high-resolution spatial transcriptomic analysis of human skeletal muscle histological sections. Human vastus lateralis muscle samples from two individuals were analysed using the Xenium platform and Human Multi-Tissue and Cancer Panel targeting 377 genes complemented by staining of successive sections for Myosin Heavy Chain isoforms to differentiate between type 1 and type 2 muscle fibres. Manual segmentation of muscle fibres allowed accurate comparisons of transcript densities across fibre types and subcellular regions, overcoming limitations in the platform's automated segmentation. The analysis revealed higher transcript density in type 1 fibres, particularly in nuclear and perinuclear areas, and identified 191 out of 377 genes with differential expression between muscle fibres and perimysium. Genes such as PROX1, S100A1, LGR5, ACTA2, and LPL exhibited higher expression in type 1 fibres, whereas PEBP4, CAVIN1, GATM, and PVALB in type 2 fibres. We demonstrated that the Xenium platform is capable of high-resolution spatial in situ transcriptomic analysis of skeletal muscle histological sections. This study demonstrates that, with manual segmentation, the Xenium platform effectively performs fibre type-specific transcriptomic analysis, providing new insights into skeletal muscle biology.
Collapse
Affiliation(s)
- Nejc Umek
- Institute of Anatomy, Faculty of Medicine, University of Ljubljana, Vrazov Trg 2, 1000, Ljubljana, Slovenia.
| | - Marija Meznarič
- Institute of Anatomy, Faculty of Medicine, University of Ljubljana, Vrazov Trg 2, 1000, Ljubljana, Slovenia
| | - Žiga Šink
- Institute of Anatomy, Faculty of Medicine, University of Ljubljana, Vrazov Trg 2, 1000, Ljubljana, Slovenia
| | | | | | - Simon Horvat
- Department of Animal Science, Biotechnical Faculty, University of Ljubljana, Ljubljana, Slovenia
| |
Collapse
|
4
|
Delgado de la Herran H, Vecellio Reane D, Cheng Y, Katona M, Hosp F, Greotti E, Wettmarshausen J, Patron M, Mohr H, Prudente de Mello N, Chudenkova M, Gorza M, Walia S, Feng MSF, Leimpek A, Mielenz D, Pellegata NS, Langer T, Hajnóczky G, Mann M, Murgia M, Perocchi F. Systematic mapping of mitochondrial calcium uniporter channel (MCUC)-mediated calcium signaling networks. EMBO J 2024; 43:5288-5326. [PMID: 39261663 PMCID: PMC11535509 DOI: 10.1038/s44318-024-00219-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: 02/20/2024] [Revised: 08/08/2024] [Accepted: 08/15/2024] [Indexed: 09/13/2024] Open
Abstract
The mitochondrial calcium uniporter channel (MCUC) mediates mitochondrial calcium entry, regulating energy metabolism and cell death. Although several MCUC components have been identified, the molecular basis of mitochondrial calcium signaling networks and their remodeling upon changes in uniporter activity have not been assessed. Here, we map the MCUC interactome under resting conditions and upon chronic loss or gain of mitochondrial calcium uptake. We identify 89 high-confidence interactors that link MCUC to several mitochondrial complexes and pathways, half of which are associated with human disease. As a proof-of-concept, we validate the mitochondrial intermembrane space protein EFHD1 as a binding partner of the MCUC subunits MCU, EMRE, and MCUB. We further show a MICU1-dependent inhibitory effect of EFHD1 on calcium uptake. Next, we systematically survey compensatory mechanisms and functional consequences of mitochondrial calcium dyshomeostasis by analyzing the MCU interactome upon EMRE, MCUB, MICU1, or MICU2 knockdown. While silencing EMRE reduces MCU interconnectivity, MCUB loss-of-function leads to a wider interaction network. Our study provides a comprehensive and high-confidence resource to gain insights into players and mechanisms regulating mitochondrial calcium signaling and their relevance in human diseases.
Collapse
Affiliation(s)
- Hilda Delgado de la Herran
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum Munich, Munich, Germany
| | - Denis Vecellio Reane
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum Munich, Munich, Germany
| | - Yiming Cheng
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum Munich, Munich, Germany
| | - Máté Katona
- Department of Pathology, Anatomy, and Cell Biology, MitoCare Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Fabian Hosp
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, Martinsried, Germany
- Roche Pharma Research and Early Development, Large Molecule Research, Mass Spectrometry, Penzberg, Germany
| | - Elisa Greotti
- Neuroscience Institute, National Research Council of Italy, Padua, Italy
- Department of Biomedical Sciences, University of Padova, Padua, Italy
- Padova Neuroscience Center, University of Padova, Padua, Italy
| | - Jennifer Wettmarshausen
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum Munich, Munich, Germany
| | - Maria Patron
- Institute for Genetics, Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, Center for Molecular Medicine, University of Cologne, Cologne, Germany
- Max Planck Institute for Biology of Aging, Cologne, Germany
| | - Hermine Mohr
- Institute of Diabetes and Cancer, Helmholtz Center Munich, Munich, Germany
| | - Natalia Prudente de Mello
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum Munich, Munich, Germany
| | - Margarita Chudenkova
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum Munich, Munich, Germany
| | - Matteo Gorza
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum Munich, Munich, Germany
| | - Safal Walia
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum Munich, Munich, Germany
| | - Michael Sheng-Fu Feng
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum Munich, Munich, Germany
| | - Anja Leimpek
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum Munich, Munich, Germany
| | - Dirk Mielenz
- Division of Molecular Immunology, University of Erlangen, Nikolaus-Fiebiger-Zentrum, FAU Erlangen-Nürnberg, Erlangen, Germany
| | - Natalia S Pellegata
- Institute of Diabetes and Cancer, Helmholtz Center Munich, Munich, Germany
- Department of Biology and Biotechnology, University of Pavia, Pavia, Italy
| | - Thomas Langer
- Institute for Genetics, Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, Center for Molecular Medicine, University of Cologne, Cologne, Germany
- Max Planck Institute for Biology of Aging, Cologne, Germany
| | - György Hajnóczky
- Department of Pathology, Anatomy, and Cell Biology, MitoCare Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Matthias Mann
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, Martinsried, Germany
- Faculty of Health Sciences, Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Copenhagen, Denmark
| | - Marta Murgia
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, Martinsried, Germany.
- Department of Biomedical Sciences, University of Padova, Padua, Italy.
| | - Fabiana Perocchi
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum Munich, Munich, Germany.
- Institute of Neuronal Cell Biology, Technical University of Munich, Munich, Germany.
- Munich Cluster for Systems Neurology, Munich, Germany.
| |
Collapse
|
5
|
Musliha A, Dermawan D, Rahayu P, Tjandrawinata RR. Unraveling modulation effects on albumin synthesis and inflammation by Striatin, a bioactive protein fraction isolated from Channa striata: In silico proteomics and in vitro approaches. Heliyon 2024; 10:e38386. [PMID: 39398063 PMCID: PMC11467539 DOI: 10.1016/j.heliyon.2024.e38386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 09/23/2024] [Accepted: 09/23/2024] [Indexed: 10/15/2024] Open
Abstract
Hypoalbuminemia, associated with inflammation in severely ill patients, can emerge due to decreased albumin production. Transforming growth factor-beta (TGF-β) and nuclear factor-kappa B (NF-κB) are critical signaling pathways responsible for decreased albumin expression. This study explores the protein content and modulation effects of Striatin on albumin synthesis and inflammation, employing in silico proteomics and in vitro investigations. In the in silico proteomics realm, LC/MS-MS protein sequencing, 3D modeling, protein-protein docking simulations, 100 ns molecular dynamics (MD) simulations, and MM/PBSA binding free energy calculations were carried out. Complementing this, in vitro studies examined Albumin gene expression and extracellular secretion in HepG2 cells subjected to lipopolysaccharides-induced hypoalbuminemia. Furthermore, the study probed Striatin's influence on the NF-ᴋB expression, given albumin's role as a negative acute-phase protein. The results unveiled nucleoside diphosphate kinase (NdK) and parvalbumin (PV) as the prominent constituents within Striatin. Notably, NdK and PV exhibited the ability to disrupt hydrogen bonds with specific residues in both TGF-β and NF-κB complexes, thereby enhancing their flexibility, akin to the action of the FKBP12 complex (antagonist complex). In the in vitro experiments, Striatin demonstrated a dose and time-dependent inhibition of hypoalbuminemia, with peak efficacy observed at a concentration of 20 μg/mL. At this concentration, Striatin also suppressed NF-κB expression when co-incubated with lipopolysaccharides. While these findings suggest potential inhibitory effects of Striatin on TGF-β and NF-κB activities, they are preliminary and warrant further investigation. This study highlights Striatin's potential as a therapeutic agent for inflammation-related hypoalbuminemia, though additional research is needed to fully validate these results.
Collapse
Affiliation(s)
- Affina Musliha
- Dexa Laboratories of Biomolecular Sciences, PT Dexa Medica, Jababeka Industrial Estate II, Jl. Industri Selatan V Blok PP No. 7 Cikarang, 17550, Indonesia
| | - Doni Dermawan
- Dexa Laboratories of Biomolecular Sciences, PT Dexa Medica, Jababeka Industrial Estate II, Jl. Industri Selatan V Blok PP No. 7 Cikarang, 17550, Indonesia
| | - Puji Rahayu
- Dexa Laboratories of Biomolecular Sciences, PT Dexa Medica, Jababeka Industrial Estate II, Jl. Industri Selatan V Blok PP No. 7 Cikarang, 17550, Indonesia
| | - Raymond R. Tjandrawinata
- Dexa Laboratories of Biomolecular Sciences, PT Dexa Medica, Jababeka Industrial Estate II, Jl. Industri Selatan V Blok PP No. 7 Cikarang, 17550, Indonesia
- Faculty of Biotechnology, Atma Jaya Catholic University of Indonesia, South Jakarta 12930, Indonesia
| |
Collapse
|
6
|
Mittal N, Ataman M, Tintignac L, Ham DJ, Jörin L, Schmidt A, Sinnreich M, Ruegg MA, Zavolan M. Calorie restriction and rapamycin distinctly restore non-canonical ORF translation in the muscles of aging mice. NPJ Regen Med 2024; 9:23. [PMID: 39300171 DOI: 10.1038/s41536-024-00369-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 09/10/2024] [Indexed: 09/22/2024] Open
Abstract
Loss of protein homeostasis is one of the hallmarks of aging. As such, interventions that restore proteostasis should slow down the aging process and improve healthspan. Two of the most broadly used anti-aging interventions that are effective in organisms from yeast to mammals are calorie restriction (CR) and rapamycin (RM) treatment. To identify the regulatory mechanisms by which these interventions improve the protein homeostasis, we carried out ribosome footprinting in the muscle of mice aged under standard conditions, or under long-term treatment with CR or RM. We found that the treatments distinctly impact the non-canonical translation, RM primarily remodeling the translation of upstream open reading frames (uORFs), while CR restores stop codon readthrough and the translation of downstream ORFs. Proteomics analysis revealed the expression of numerous non-canonical ORFs at the protein level. The corresponding peptides may provide entry points for therapies aiming to maintain muscle function and extend health span.
Collapse
Affiliation(s)
- Nitish Mittal
- Biozentrum, University of Basel, Basel, Switzerland.
| | - Meric Ataman
- Biozentrum, University of Basel, Basel, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Lionel Tintignac
- Biozentrum, University of Basel, Basel, Switzerland
- Departments of Neurology and Biomedicine, University of Basel, Basel, Switzerland
- University Hospital Basel, Basel, Switzerland
| | - Daniel J Ham
- Biozentrum, University of Basel, Basel, Switzerland
| | - Lena Jörin
- Biozentrum, University of Basel, Basel, Switzerland
| | | | - Michael Sinnreich
- Departments of Neurology and Biomedicine, University of Basel, Basel, Switzerland
- University Hospital Basel, Basel, Switzerland
| | | | - Mihaela Zavolan
- Biozentrum, University of Basel, Basel, Switzerland.
- Swiss Institute of Bioinformatics, Lausanne, Switzerland.
| |
Collapse
|
7
|
Marcucci L, Nogara L, Canato M, Germinario E, Raffaello A, Carraro M, Bernardi P, Pietrangelo L, Boncompagni S, Protasi F, Paolocci N, Reggiani C. Mitochondria can substitute for parvalbumin to lower cytosolic calcium levels in the murine fast skeletal muscle. Acta Physiol (Oxf) 2024; 240:e14208. [PMID: 39077881 DOI: 10.1111/apha.14208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 07/09/2024] [Accepted: 07/15/2024] [Indexed: 07/31/2024]
Abstract
AIM Parvalbumin (PV) is a primary calcium buffer in mouse fast skeletal muscle fibers. Previous work showed that PV ablation has a limited impact on cytosolic Ca2+ ([Ca2+]cyto) transients and contractile response, while it enhances mitochondrial density and mitochondrial matrix-free calcium concentration ([Ca2+]mito). Here, we aimed to quantitatively test the hypothesis that mitochondria act to compensate for PV deficiency. METHODS We determined the free Ca2+ redistribution during a 2 s 60 Hz tetanic stimulation in the sarcoplasmic reticulum, cytosol, and mitochondria. Via a reaction-diffusion Ca2+ model, we quantitatively evaluated mitochondrial uptake and storage capacity requirements to compensate for PV lack and analyzed possible extracellular export. RESULTS [Ca2+]mito during tetanic stimulation is greater in knock-out (KO) (1362 ± 392 nM) than in wild-type (WT) (855 ± 392 nM), p < 0.05. Under the assumption of a non-linear intramitochondrial buffering, the model predicts an accumulation of 725 μmoles/L fiber (buffering ratio 1:11 000) in KO, much higher than in WT (137 μmoles/L fiber, ratio 1:4500). The required transport rate via mitochondrial calcium uniporter (MCU) reaches 3 mM/s, compatible with available literature. TEM images of calcium entry units and Mn2+ quenching showed a greater capacity of store-operated calcium entry in KO compared to WT. However, levels of [Ca2+]cyto during tetanic stimulation were not modulated to variations of extracellular calcium. CONCLUSIONS The model-based analysis of experimentally determined calcium distribution during tetanic stimulation showed that mitochondria can act as a buffer to compensate for the lack of PV. This result contributes to a better understanding of mitochondria's role in modulating [Ca2+]cyto in skeletal muscle fibers.
Collapse
Affiliation(s)
- Lorenzo Marcucci
- Department of Biomedical Sciences, University of Padova, Padova, Italy
- Center for Biosystems Dynamics Research, RIKEN, Suita, Japan
| | - Leonardo Nogara
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Marta Canato
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Elena Germinario
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Anna Raffaello
- Department of Biomedical Sciences, University of Padova, Padova, Italy
- Myology Center, University of Padova, Padova, Italy
| | - Michela Carraro
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Paolo Bernardi
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Laura Pietrangelo
- CAST, Center for Advanced Studies and Technology, University G. d'Annunzio of Chieti-Pescara, Chieti, Italy
- DMSI, Department of Medicine and Aging Sciences, University G. d'Annunzio of Chieti-Pescara, Chieti, Italy
| | - Simona Boncompagni
- CAST, Center for Advanced Studies and Technology, University G. d'Annunzio of Chieti-Pescara, Chieti, Italy
- DNICS, Department of Neuroscience and Clinical Sciences, University G. d'Annunzio of Chieti-Pescara, Chieti, Italy
| | - Feliciano Protasi
- CAST, Center for Advanced Studies and Technology, University G. d'Annunzio of Chieti-Pescara, Chieti, Italy
- DMSI, Department of Medicine and Aging Sciences, University G. d'Annunzio of Chieti-Pescara, Chieti, Italy
| | - Nazareno Paolocci
- Department of Biomedical Sciences, University of Padova, Padova, Italy
- Division of Cardiology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Carlo Reggiani
- Department of Biomedical Sciences, University of Padova, Padova, Italy
- Science and Research Center Koper, Institute for Kinesiology Research, Koper, Slovenia
| |
Collapse
|
8
|
Vecellio Reane D, Serna JDC, Raffaello A. Unravelling the complexity of the mitochondrial Ca 2+ uniporter: regulation, tissue specificity, and physiological implications. Cell Calcium 2024; 121:102907. [PMID: 38788256 DOI: 10.1016/j.ceca.2024.102907] [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: 04/23/2024] [Revised: 05/10/2024] [Accepted: 05/13/2024] [Indexed: 05/26/2024]
Abstract
Calcium (Ca2+) signalling acts a pleiotropic message within the cell that is decoded by the mitochondria through a sophisticated ion channel known as the Mitochondrial Ca2+ Uniporter (MCU) complex. Under physiological conditions, mitochondrial Ca2+ signalling is crucial for coordinating cell activation with energy production. Conversely, in pathological scenarios, it can determine the fine balance between cell survival and death. Over the last decade, significant progress has been made in understanding the molecular bases of mitochondrial Ca2+ signalling. This began with the elucidation of the MCU channel components and extended to the elucidation of the mechanisms that regulate its activity. Additionally, increasing evidence suggests molecular mechanisms allowing tissue-specific modulation of the MCU complex, tailoring channel activity to the specific needs of different tissues or cell types. This review aims to explore the latest evidence elucidating the regulation of the MCU complex, the molecular factors controlling the tissue-specific properties of the channel, and the physiological and pathological implications of mitochondrial Ca2+ signalling in different tissues.
Collapse
Affiliation(s)
- Denis Vecellio Reane
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center (HDC), Helmholtz Zentrum Munich, Germany.
| | - Julian D C Serna
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| | - Anna Raffaello
- Department of Biomedical Sciences, University of Padova, Italy.
| |
Collapse
|
9
|
Yadav A, Dabur R. Skeletal muscle atrophy after sciatic nerve damage: Mechanistic insights. Eur J Pharmacol 2024; 970:176506. [PMID: 38492879 DOI: 10.1016/j.ejphar.2024.176506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 03/13/2024] [Accepted: 03/14/2024] [Indexed: 03/18/2024]
Abstract
Sciatic nerve injury leads to molecular events that cause muscular dysfunction advancement in atrophic conditions. Nerve damage renders muscles permanently relaxed which elevates intracellular resting Ca2+ levels. Increased Ca2+ levels are associated with several cellular signaling pathways including AMPK, cGMP, PLC-β, CERB, and calcineurin. Also, multiple enzymes involved in the tricarboxylic acid cycle and oxidative phosphorylation are activated by Ca2+ influx into mitochondria during muscle contraction, to meet increased ATP demand. Nerve damage induces mitophagy and skeletal muscle atrophy through increased sensitivity to Ca2+-induced opening of the permeability transition pore (PTP) in mitochondria attributed to Ca2+, ROS, and AMPK overload in muscle. Activated AMPK interacts negatively with Akt/mTOR is a highly prevalent and well-described central pathway for anabolic processes. Over the decade several reports indicate abnormal behavior of signaling machinery involved in denervation-induced muscle loss but end up with some controversial outcomes. Therefore, understanding how the synthesis and inhibitory stimuli interact with cellular signaling to control muscle mass and morphology may lead to new pharmacological insights toward understanding the underlying mechanism of muscle loss after sciatic nerve damage. Hence, the present review summarizes the existing literature on denervation-induced muscle atrophy to evaluate the regulation and expression of differential regulators during sciatic damage.
Collapse
Affiliation(s)
- Aarti Yadav
- Clinical Biochemistry Laboratory, Department of Biochemistry, Maharshi Dayanand University, Rohtak, 124001, Haryana, India
| | - Rajesh Dabur
- Clinical Biochemistry Laboratory, Department of Biochemistry, Maharshi Dayanand University, Rohtak, 124001, Haryana, India.
| |
Collapse
|
10
|
Ibrahim Z, Khan NA, Qaisar R, Saleh MA, Siddiqui R, Al-Hroub HM, Giddey AD, Semreen MH, Soares NC, Elmoselhi AB. Serum multi-omics analysis in hindlimb unloading mice model: Insights into systemic molecular changes and potential diagnostic and therapeutic biomarkers. Heliyon 2024; 10:e23592. [PMID: 38187258 PMCID: PMC10770503 DOI: 10.1016/j.heliyon.2023.e23592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Revised: 12/06/2023] [Accepted: 12/07/2023] [Indexed: 01/09/2024] Open
Abstract
Microgravity, in space travel and prolonged bed rest conditions, induces cardiovascular deconditioning along with skeletal muscle mass loss and weakness. The findings of microgravity research may also aid in the understanding and treatment of human health conditions on Earth such as muscle atrophy, and cardiovascular diseases. Due to the paucity of biomarkers and the unknown underlying mechanisms of cardiovascular and skeletal muscle deconditioning in these environments, there are insufficient diagnostic and preventative measures. In this study, we employed hindlimb unloading (HU) mouse model, which mimics astronauts in space and bedridden patients, to first evaluate cardiovascular and skeletal muscle function, followed by proteomics and metabolomics LC-MS/MS-based analysis using serum samples. Three weeks of unloading caused changes in the function of the cardiovascular system in c57/Bl6 mice, as seen by a decrease in mean arterial pressure and heart weight. Unloading for three weeks also changed skeletal muscle function, causing a loss in grip strength in HU mice and atrophy of skeletal muscle indicated by a reduction in muscle mass. These modifications were partially reversed by a two-week recovery period of reloading condition, emphasizing the significance of the recovery process. Proteomics analysis revealed 12 dysregulated proteins among the groups, such as phospholipid transfer protein, Carbonic anhydrase 3, Parvalbumin alpha, Major urinary protein 20 (Mup20), Thrombospondin-1, and Apolipoprotein C-IV. On the other hand, metabolomics analysis showed altered metabolites among the groups such as inosine, hypoxanthine, xanthosine, sphinganine, l-valine, 3,4-Dihydroxyphenylglycol, and l-Glutamic acid. The joint data analysis revealed that HU conditions mainly impacted pathways such as ABC transporters, complement and coagulation cascades, nitrogen metabolism, and purine metabolism. Overall, our results indicate that microgravity environment induces significant alterations in the function, proteins, and metabolites of these mice. These observations suggest the potential utilization of these proteins and metabolites as novel biomarkers for assessing and mitigating cardiovascular and skeletal muscle deconditioning associated with such conditions.
Collapse
Affiliation(s)
- Zeinab Ibrahim
- Research Institute of Medical & Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
- Basic Medical Sciences Department, College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Naveed A. Khan
- Research Institute of Medical & Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
- Clinical Sciences Department, College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
- Microbiota Research Center, Istinye University, Istanbul, 34010, Turkey
| | - Rizwan Qaisar
- Research Institute of Medical & Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
- Basic Medical Sciences Department, College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Mohamed A. Saleh
- Research Institute of Medical & Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
- Clinical Sciences Department, College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
| | - Ruqaiyyah Siddiqui
- Institute of Biological Chemistry, Biophysics and Bioengineering, Heriot-Watt University Edinburgh, EH14 4AS UK
- Microbiota Research Center, Istinye University, Istanbul, 34010, Turkey
| | - Hamza M. Al-Hroub
- Research Institute of Medical & Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Alexander D. Giddey
- Center for Applied and Translational Genomics, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, United Arab Emirates
| | - Mohammad Harb Semreen
- Research Institute of Medical & Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
- Department of Medicinal Chemistry, College of Pharmacy, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Nelson C. Soares
- Research Institute of Medical & Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
- Department of Medicinal Chemistry, College of Pharmacy, University of Sharjah, Sharjah 27272, United Arab Emirates
- Laboratory of Proteomics, Department of Human Genetics, National Institute of Health Doutor Ricardo Jorge (INSA), Av. Padre Cruz, Lisbon, 1649-016, Portugal
- Centre for Toxicogenomics and Human Health (ToxOmics), NOVA School/ Faculdade de Lisboa, Lisbon, Portugal
| | - Adel B. Elmoselhi
- Research Institute of Medical & Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
- Basic Medical Sciences Department, College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
| |
Collapse
|
11
|
Li X, Sun B, Li J, Ye W, Li M, Guan F, Wu S, Luo X, Feng J, Jia J, Liu X, Li T, Liu L. SEPSIS LEADS TO IMPAIRED MITOCHONDRIAL CALCIUM UPTAKE AND SKELETAL MUSCLE WEAKNESS BY REDUCING THE MICU1:MCU PROTEIN RATIO. Shock 2023; 60:698-706. [PMID: 37695737 PMCID: PMC10662578 DOI: 10.1097/shk.0000000000002221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 05/23/2023] [Accepted: 08/29/2023] [Indexed: 09/13/2023]
Abstract
Purpose: Intensive care unit-acquired weakness (ICUAW) is a severe neuromuscular complication that frequently occurs in patients with sepsis. The precise molecular pathophysiology of mitochondrial calcium uptake 1 (MICU1) and mitochondrial calcium uniporter (MCU) in ICUAW has not been fully elucidated. Here, we speculate that ICUAW is associated with MICU1:MCU protein ratio-mediated mitochondrial calcium ([Ca 2+ ] m ) uptake dysfunction. Methods: Cecal ligation and perforation (CLP) was performed on C57BL/6J mice to induce sepsis. Sham-operated animals were used as controls. Lipopolysaccharide (LPS) (5 μg/mL) was used to induce inflammation in differentiated C2C12 myoblasts. Compound muscle action potential (CMAP) was detected using a biological signal acquisition system. Grip strength was measured using a grip-strength meter. Skeletal muscle inflammatory factors were detected using ELISA kits. The cross-sectional area (CSA) of the tibialis anterior (TA) muscle was detected by hematoxylin and eosin staining. Cytosolic calcium ([Ca 2+ ] c ) levels were measured using Fluo-4 AM. Adeno-associated virus (AAV) was injected into TA muscles for 4 weeks to overexpress MICU1 prophylactically. A lentivirus was used to infect C2C12 cells to increase MICU1 expression prophylactically. Findings: The results suggest that sepsis induces [Ca 2+ ] m uptake disorder by reducing the MICU1:MCU protein ratio, resulting in skeletal muscle weakness and muscle fiber atrophy. However, MICU1 prophylactic overexpression reversed these effects by increasing the MICU1:MCU protein ratio. Conclusions: ICUAW is associated with impaired [Ca 2+ ] m uptake caused by a decreased MICU1:MCU protein ratio. MICU1 overexpression improves sepsis-induced skeletal muscle weakness and atrophy by ameliorating the [Ca 2+ ] m uptake disorder.
Collapse
Affiliation(s)
- Xuexin Li
- Department of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province, China
- Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, Southwest Medical University, Luzhou, Sichuan Province, China
| | - Bowen Sun
- Department of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province, China
- Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, Southwest Medical University, Luzhou, Sichuan Province, China
| | - Jie Li
- Department of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province, China
- Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, Southwest Medical University, Luzhou, Sichuan Province, China
| | - Wanlin Ye
- Laboratory of Mitochondria and Metabolism, West China Hospital of Sichuan University, Chengdu, China
| | - Mingjuan Li
- Department of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province, China
- Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, Southwest Medical University, Luzhou, Sichuan Province, China
| | - Fasheng Guan
- Department of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province, China
- Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, Southwest Medical University, Luzhou, Sichuan Province, China
| | - Songlin Wu
- Department of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province, China
- Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, Southwest Medical University, Luzhou, Sichuan Province, China
| | - Xuerong Luo
- Department of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province, China
- Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, Southwest Medical University, Luzhou, Sichuan Province, China
| | - Jianguo Feng
- Department of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province, China
- Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, Southwest Medical University, Luzhou, Sichuan Province, China
| | - Jing Jia
- Department of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province, China
- Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, Southwest Medical University, Luzhou, Sichuan Province, China
| | - Xueru Liu
- Department of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province, China
- Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, Southwest Medical University, Luzhou, Sichuan Province, China
| | - Tao Li
- Laboratory of Mitochondria and Metabolism, West China Hospital of Sichuan University, Chengdu, China
| | - Li Liu
- Department of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province, China
- Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, Southwest Medical University, Luzhou, Sichuan Province, China
| |
Collapse
|
12
|
O’Connor TN, Zhao N, Orciuoli HM, Brasile A, Pietrangelo L, He M, Groom L, Leigh J, Mahamed Z, Liang C, Malik S, Protasi F, Dirksen RT. Voluntary wheel running mitigates disease in an Orai1 gain-of-function mouse model of tubular aggregate myopathy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.29.559036. [PMID: 37808709 PMCID: PMC10557777 DOI: 10.1101/2023.09.29.559036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/10/2023]
Abstract
Tubular aggregate myopathy (TAM) is an inherited skeletal muscle disease associated with progressive muscle weakness, cramps, and myalgia. Tubular aggregates (TAs) are regular arrays of highly ordered and densely packed SR straight-tubes in muscle biopsies; the extensive presence of TAs represent a key histopathological hallmark of this disease in TAM patients. TAM is caused by gain-of-function mutations in proteins that coordinate store-operated Ca2+ entry (SOCE): STIM1 Ca2+ sensor proteins in the sarcoplasmic reticulum (SR) and Ca2+-permeable ORAI1 channels in the surface membrane. We have previously shown that voluntary wheel running (VWR) prevents formation of TAs in aging mice. Here, we assessed the therapeutic potential of endurance exercise (in the form of VWR) in mitigating the functional and structural alterations in a knock-in mouse model of TAM (Orai1G100S/+ or GS mice) based on a gain-of-function mutation in the ORAI1 pore. WT and GS mice were singly-housed for six months (from two to eight months of age) with either free-spinning or locked low profile wheels. Six months of VWR exercise significantly increased soleus peak tetanic specific force production, normalized FDB fiber Ca2+ store content, and markedly reduced TAs in EDL muscle from GS mice. Six months of VWR exercise normalized the expression of mitochondrial proteins found to be altered in soleus muscle of sedentary GS mice in conjunction with a signature of increased protein translation and biosynthetic processes. Parallel proteomic analyses of EDL muscles from sedentary WT and GS mice revealed changes in a tight network of pathways involved in formation of supramolecular complexes, which were also normalized following six months of VWR. In summary, sustained voluntary endurance exercise improved slow twitch muscle function, reduced the presence of TAs in fast twitch muscle, and normalized the muscle proteome of GS mice consistent with protective adaptions in proteostasis, mitochondrial structure/function, and formation of supramolecular complexes.
Collapse
Affiliation(s)
- Thomas N. O’Connor
- Department of Biomedical Genetics, Genetics and Genomics Graduate Program, University of Rochester Medical Center, Rochester, NY, USA
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY, USA
| | - Nan Zhao
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY, USA
| | - Haley M. Orciuoli
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY, USA
- Department of Biology, Biological Sciences, University of Rochester, Rochester, NY, USA
| | - Alice Brasile
- CAST, Center for Advanced Studies and Technology & DMSI, Department of Medicine and Aging Sciences, University G. d’Annunzio of Chieti-Pescara, I-66100 Chieti, Italy
| | - Laura Pietrangelo
- CAST, Center for Advanced Studies and Technology & DMSI, Department of Medicine and Aging Sciences, University G. d’Annunzio of Chieti-Pescara, I-66100 Chieti, Italy
| | - Miao He
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY, USA
| | - Linda Groom
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY, USA
| | - Jennifer Leigh
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY, USA
| | - Zahra Mahamed
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY, USA
| | - Chen Liang
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY, USA
| | - Sundeep Malik
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY, USA
| | - Feliciano Protasi
- CAST, Center for Advanced Studies and Technology & DMSI, Department of Medicine and Aging Sciences, University G. d’Annunzio of Chieti-Pescara, I-66100 Chieti, Italy
| | - Robert T. Dirksen
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY, USA
| |
Collapse
|
13
|
Ino Y, Ohira T, Kumagai K, Nakai Y, Akiyama T, Moriyama K, Takeda Y, Saito T, Ryo A, Inaba Y, Hirano H, Kimura Y. Identification of mouse soleus muscle proteins altered in response to changes in gravity loading. Sci Rep 2023; 13:15768. [PMID: 37737267 PMCID: PMC10517164 DOI: 10.1038/s41598-023-42875-8] [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: 04/03/2023] [Accepted: 09/15/2023] [Indexed: 09/23/2023] Open
Abstract
Gravity-dependent physical processes strongly affect the ability of elderly people to maintain musculoskeletal health by reducing muscle atrophy and increasing bone mineral density, thereby increasing quality of life. A need therefore exists to identify molecules in the musculoskeletal system that are responsive to gravitational loading and to establish an objective indicator for the maintenance of healthy musculoskeletal systems. Here, we performed an integrated assessment of the results of soleus muscle proteomic analyses in three model mouse experiments under different gravity environments (hypergravity, hindlimb unloading, and spaceflight). Myl6b, Gpd1, Fbp2, Pvalb, and Actn3 were shown to be gravity-responsive muscle proteins, and alterations in the levels of these proteins indicated changes in muscle fiber type to slow-twitch type due to gravity loading. In addition, immunoblotting and enzyme-linked immunosorbent assays revealed that Pvalb levels in the sera of hindlimb-unloaded mice and osteoporosis patients were higher than in control subjects, suggesting that Pvalb levels might be useful to objectively evaluate soleus muscle atrophy and bone loss.
Collapse
Affiliation(s)
- Yoko Ino
- Advanced Medical Research Center, Yokohama City University, Fukuura 3-9, Kanazawa-Ku, Yokohama, 236-0004, Japan
| | - Takashi Ohira
- Advanced Medical Research Center, Yokohama City University, Fukuura 3-9, Kanazawa-Ku, Yokohama, 236-0004, Japan.
- Department of Physiology and Regenerative Medicine, Kindai University Faculty of Medicine, Ohno-Higashi 377-2, Osaka-Sayama, Osaka, Japan.
| | - Ken Kumagai
- Department of Orthopaedic Surgery, Yokohama City University School of Medicine, Yokohama, Japan
| | - Yusuke Nakai
- Advanced Medical Research Center, Yokohama City University, Fukuura 3-9, Kanazawa-Ku, Yokohama, 236-0004, Japan
| | - Tomoko Akiyama
- Advanced Medical Research Center, Yokohama City University, Fukuura 3-9, Kanazawa-Ku, Yokohama, 236-0004, Japan
| | - Kayano Moriyama
- Advanced Medical Research Center, Yokohama City University, Fukuura 3-9, Kanazawa-Ku, Yokohama, 236-0004, Japan
| | - Yuriko Takeda
- Department of Biostatistics, Yokohama City University School of Medicine, Yokohama, Japan
| | | | - Akihide Ryo
- Advanced Medical Research Center, Yokohama City University, Fukuura 3-9, Kanazawa-Ku, Yokohama, 236-0004, Japan
| | - Yutaka Inaba
- Department of Orthopaedic Surgery, Yokohama City University School of Medicine, Yokohama, Japan
| | - Hisashi Hirano
- Advanced Medical Research Center, Yokohama City University, Fukuura 3-9, Kanazawa-Ku, Yokohama, 236-0004, Japan
| | - Yayoi Kimura
- Advanced Medical Research Center, Yokohama City University, Fukuura 3-9, Kanazawa-Ku, Yokohama, 236-0004, Japan.
| |
Collapse
|
14
|
Sharlo KA, Lvova ID, Tyganov SA, Zaripova KA, Belova SP, Kostrominova TY, Shenkman BS, Nemirovskaya TL. The Effect of SERCA Activation on Functional Characteristics and Signaling of Rat Soleus Muscle upon 7 Days of Unloading. Biomolecules 2023; 13:1354. [PMID: 37759754 PMCID: PMC10526198 DOI: 10.3390/biom13091354] [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: 07/11/2023] [Revised: 08/28/2023] [Accepted: 09/02/2023] [Indexed: 09/29/2023] Open
Abstract
Skeletal muscle abnormalities and atrophy during unloading are accompanied by the accumulation of excess calcium in the sarcoplasm. We hypothesized that calcium accumulation may occur, among other mechanisms, due to the inhibition of sarco/endoplasmic reticulum Ca2+-ATPase (SERCA) activity. Consequently, the use of the SERCA activator will reduce the level of calcium in the sarcoplasm and prevent the negative consequences of muscle unloading. Wistar rats were randomly assigned into one of three groups (eight rats per group): control rats with placebo (C), 7 days of unloading/hindlimb suspension with placebo (7HS), and 7 days of unloading treated with SERCA activator CDN1163 (7HSC). After seven days of unloading the soleus muscle, the 7HS group displayed increased fatigue in the ex vivo test, a significant increase in the level of calcium-dependent CaMK II phosphorylation and the level of tropomyosin oxidation, as well as a decrease in the content of mitochondrial DNA and protein, slow-type myosin mRNA, and the percentage of slow-type muscle fibers. All of these changes were prevented in the 7HSC group. Moreover, treatment with CDN1163 blocked a decrease in the phosphorylation of p70S6k, an increase in eEF2 phosphorylation, and an increase in MuRF-1 mRNA expression. Nevertheless, there were no differences in the degree of fast and slow muscle fiber atrophy between the 7HS and 7HSC groups. Conclusion: SERCA activation during 7 days of unloading prevented an increase in soleus fatigue, the decrease of slow-type myosin, mitochondrial markers, and markers of calcium homeostasis but had no effect on muscle atrophy.
Collapse
Affiliation(s)
- Kristina A. Sharlo
- Myology Laboratory, Institute of Biomedical Problems, RAS (Russian Academy of Sciences), Moscow 123007, Russia; (K.A.S.); (I.D.L.); (S.A.T.); (K.A.Z.); (S.P.B.); (B.S.S.)
| | - Irina D. Lvova
- Myology Laboratory, Institute of Biomedical Problems, RAS (Russian Academy of Sciences), Moscow 123007, Russia; (K.A.S.); (I.D.L.); (S.A.T.); (K.A.Z.); (S.P.B.); (B.S.S.)
| | - Sergey A. Tyganov
- Myology Laboratory, Institute of Biomedical Problems, RAS (Russian Academy of Sciences), Moscow 123007, Russia; (K.A.S.); (I.D.L.); (S.A.T.); (K.A.Z.); (S.P.B.); (B.S.S.)
| | - Ksenia A. Zaripova
- Myology Laboratory, Institute of Biomedical Problems, RAS (Russian Academy of Sciences), Moscow 123007, Russia; (K.A.S.); (I.D.L.); (S.A.T.); (K.A.Z.); (S.P.B.); (B.S.S.)
| | - Svetlana P. Belova
- Myology Laboratory, Institute of Biomedical Problems, RAS (Russian Academy of Sciences), Moscow 123007, Russia; (K.A.S.); (I.D.L.); (S.A.T.); (K.A.Z.); (S.P.B.); (B.S.S.)
| | - Tatiana Y. Kostrominova
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine-Northwest, Gary, IN 46202, USA;
| | - Boris S. Shenkman
- Myology Laboratory, Institute of Biomedical Problems, RAS (Russian Academy of Sciences), Moscow 123007, Russia; (K.A.S.); (I.D.L.); (S.A.T.); (K.A.Z.); (S.P.B.); (B.S.S.)
| | - Tatiana L. Nemirovskaya
- Myology Laboratory, Institute of Biomedical Problems, RAS (Russian Academy of Sciences), Moscow 123007, Russia; (K.A.S.); (I.D.L.); (S.A.T.); (K.A.Z.); (S.P.B.); (B.S.S.)
| |
Collapse
|
15
|
Li C, Sun J, Zhang X, Zhou M, Gan X. Implications of MCU complex in metabolic diseases. FASEB J 2023; 37:e23046. [PMID: 37389546 DOI: 10.1096/fj.202300218r] [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: 02/07/2023] [Revised: 05/17/2023] [Accepted: 06/07/2023] [Indexed: 07/01/2023]
Abstract
Metabolic diseases are considered the primary culprit for physical and mental health of individuals. Although the diagnosis of these diseases is relatively easy, more effective and convenient potent drugs are still being explored. Ca2+ across the inner mitochondrial membrane is a vital intracellular messenger that regulates energy metabolism and cellular Ca2+ homeostasis and is involved in cell death. Mitochondria rely on a selective mitochondrial Ca2+ unidirectional transport complex (MCU complex) in their inner membrane for Ca2+ uptake. We found that the channel contains several subunits and undergoes dramatic transformations in various pathological processes, especially in metabolic diseases. In this way, we believe that the MCU complex becomes a target with significant potential for these diseases. However, there is no review linking the two factors, thus hindering the possibility of new drug production. Here, we highlight the connection between MCU complex-related Ca2+ transport and the pathophysiology of metabolic diseases, adding understanding and insight at the molecular level to provide new insights for targeting MCU to reverse metabolism-related diseases.
Collapse
Affiliation(s)
- Chen Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, School of Chemical Engineering, Sichuan University, Chengdu, China
| | - Jiyu Sun
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, School of Chemical Engineering, Sichuan University, Chengdu, China
| | - Xidan Zhang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, School of Chemical Engineering, Sichuan University, Chengdu, China
| | - Min Zhou
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, School of Chemical Engineering, Sichuan University, Chengdu, China
| | - Xueqi Gan
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, School of Chemical Engineering, Sichuan University, Chengdu, China
| |
Collapse
|
16
|
Klawitter F, Ehler J, Bajorat R, Patejdl R. Mitochondrial Dysfunction in Intensive Care Unit-Acquired Weakness and Critical Illness Myopathy: A Narrative Review. Int J Mol Sci 2023; 24:5516. [PMID: 36982590 PMCID: PMC10052131 DOI: 10.3390/ijms24065516] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 03/03/2023] [Accepted: 03/09/2023] [Indexed: 03/16/2023] Open
Abstract
Mitochondria are key structures providing most of the energy needed to maintain homeostasis. They are the main source of adenosine triphosphate (ATP), participate in glucose, lipid and amino acid metabolism, store calcium and are integral components in various intracellular signaling cascades. However, due to their crucial role in cellular integrity, mitochondrial damage and dysregulation in the context of critical illness can severely impair organ function, leading to energetic crisis and organ failure. Skeletal muscle tissue is rich in mitochondria and, therefore, particularly vulnerable to mitochondrial dysfunction. Intensive care unit-acquired weakness (ICUAW) and critical illness myopathy (CIM) are phenomena of generalized weakness and atrophying skeletal muscle wasting, including preferential myosin breakdown in critical illness, which has also been linked to mitochondrial failure. Hence, imbalanced mitochondrial dynamics, dysregulation of the respiratory chain complexes, alterations in gene expression, disturbed signal transduction as well as impaired nutrient utilization have been proposed as underlying mechanisms. This narrative review aims to highlight the current known molecular mechanisms immanent in mitochondrial dysfunction of patients suffering from ICUAW and CIM, as well as to discuss possible implications for muscle phenotype, function and therapeutic approaches.
Collapse
Affiliation(s)
- Felix Klawitter
- Department of Anesthesiology, Intensive Care Medicine and Pain Therapy, Rostock University Medical Center, 18057 Rostock, Germany
| | - Johannes Ehler
- Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, 07747 Jena, Germany
| | - Rika Bajorat
- Department of Anesthesiology, Intensive Care Medicine and Pain Therapy, Rostock University Medical Center, 18057 Rostock, Germany
| | - Robert Patejdl
- Oscar Langendorff Institute of Physiology, Rostock University Medical Center, 18057 Rostock, Germany
| |
Collapse
|
17
|
Marchioretti C, Zanetti G, Pirazzini M, Gherardi G, Nogara L, Andreotti R, Martini P, Marcucci L, Canato M, Nath SR, Zuccaro E, Chivet M, Mammucari C, Pacifici M, Raffaello A, Rizzuto R, Mattarei A, Desbats MA, Salviati L, Megighian A, Sorarù G, Pegoraro E, Belluzzi E, Pozzuoli A, Biz C, Ruggieri P, Romualdi C, Lieberman AP, Babu GJ, Sandri M, Blaauw B, Basso M, Pennuto M. Defective excitation-contraction coupling and mitochondrial respiration precede mitochondrial Ca 2+ accumulation in spinobulbar muscular atrophy skeletal muscle. Nat Commun 2023; 14:602. [PMID: 36746942 PMCID: PMC9902403 DOI: 10.1038/s41467-023-36185-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 01/19/2023] [Indexed: 02/08/2023] Open
Abstract
Polyglutamine expansion in the androgen receptor (AR) causes spinobulbar muscular atrophy (SBMA). Skeletal muscle is a primary site of toxicity; however, the current understanding of the early pathological processes that occur and how they unfold during disease progression remains limited. Using transgenic and knock-in mice and patient-derived muscle biopsies, we show that SBMA mice in the presymptomatic stage develop a respiratory defect matching defective expression of genes involved in excitation-contraction coupling (ECC), altered contraction dynamics, and increased fatigue. These processes are followed by stimulus-dependent accumulation of calcium into mitochondria and structural disorganization of the muscle triads. Deregulation of expression of ECC genes is concomitant with sexual maturity and androgen raise in the serum. Consistent with the androgen-dependent nature of these alterations, surgical castration and AR silencing alleviate the early and late pathological processes. These observations show that ECC deregulation and defective mitochondrial respiration are early but reversible events followed by altered muscle force, calcium dyshomeostasis, and dismantling of triad structure.
Collapse
Affiliation(s)
- Caterina Marchioretti
- Department of Biomedical Sciences (DBS), University of Padova, 35131, Padova, Italy
- Veneto Institute of Molecular Medicine (VIMM), Padova, 35100, Italy
- Padova Neuroscience Center (PNC), Padova, 35100, Italy
- Dulbecco Telethon Institute (DTI) at the Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, 38123, Trento, Italy
| | - Giulia Zanetti
- Department of Biomedical Sciences (DBS), University of Padova, 35131, Padova, Italy
| | - Marco Pirazzini
- Department of Biomedical Sciences (DBS), University of Padova, 35131, Padova, Italy
- CIR-Myo, Centro Interdipartimentale di Ricerca di Miologia, University of Padova, 35131, Padova, Italy
| | - Gaia Gherardi
- Department of Biomedical Sciences (DBS), University of Padova, 35131, Padova, Italy
| | - Leonardo Nogara
- Department of Biomedical Sciences (DBS), University of Padova, 35131, Padova, Italy
- Veneto Institute of Molecular Medicine (VIMM), Padova, 35100, Italy
| | - Roberta Andreotti
- Department of Biomedical Sciences (DBS), University of Padova, 35131, Padova, Italy
- Veneto Institute of Molecular Medicine (VIMM), Padova, 35100, Italy
- Padova Neuroscience Center (PNC), Padova, 35100, Italy
| | - Paolo Martini
- Department of Molecular and Translational Medicine, University of Brescia, 25121, Brescia, Italy
| | - Lorenzo Marcucci
- Department of Biomedical Sciences (DBS), University of Padova, 35131, Padova, Italy
| | - Marta Canato
- Department of Biomedical Sciences (DBS), University of Padova, 35131, Padova, Italy
| | - Samir R Nath
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Emanuela Zuccaro
- Department of Biomedical Sciences (DBS), University of Padova, 35131, Padova, Italy
- Veneto Institute of Molecular Medicine (VIMM), Padova, 35100, Italy
- Padova Neuroscience Center (PNC), Padova, 35100, Italy
| | - Mathilde Chivet
- Dulbecco Telethon Institute (DTI) at the Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, 38123, Trento, Italy
| | - Cristina Mammucari
- Department of Biomedical Sciences (DBS), University of Padova, 35131, Padova, Italy
- CIR-Myo, Centro Interdipartimentale di Ricerca di Miologia, University of Padova, 35131, Padova, Italy
| | - Marco Pacifici
- Department of Biomedical Sciences (DBS), University of Padova, 35131, Padova, Italy
| | - Anna Raffaello
- Department of Biomedical Sciences (DBS), University of Padova, 35131, Padova, Italy
- CIR-Myo, Centro Interdipartimentale di Ricerca di Miologia, University of Padova, 35131, Padova, Italy
| | - Rosario Rizzuto
- Department of Biomedical Sciences (DBS), University of Padova, 35131, Padova, Italy
| | - Andrea Mattarei
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35131, Padova, Italy
| | - Maria A Desbats
- Clinical Genetics Unit, Department of Women and Children's Health, University of Padova, and Fondazione Istituto di Ricerca Pediatrica Città della Speranza, Padova, Italy
| | - Leonardo Salviati
- CIR-Myo, Centro Interdipartimentale di Ricerca di Miologia, University of Padova, 35131, Padova, Italy
- Clinical Genetics Unit, Department of Women and Children's Health, University of Padova, and Fondazione Istituto di Ricerca Pediatrica Città della Speranza, Padova, Italy
| | - Aram Megighian
- Department of Biomedical Sciences (DBS), University of Padova, 35131, Padova, Italy
- Padova Neuroscience Center (PNC), Padova, 35100, Italy
| | - Gianni Sorarù
- Padova Neuroscience Center (PNC), Padova, 35100, Italy
- Department of Neuroscience (DNS), University of Padova, 35128, Padova, Italy
| | - Elena Pegoraro
- Department of Neuroscience (DNS), University of Padova, 35128, Padova, Italy
| | - Elisa Belluzzi
- Orthopedics and Orthopedic Oncology, Department of Surgery, Oncology, and Gastroenterology DiSCOG, University-Hospital of Padova, 35128, Padova, Italy
- Musculoskeletal Pathology and Oncology Laboratory, Department of Surgery, Oncology and Gastroenterology (DiSCOG), University of Padova, 35128, Padova, Italy
| | - Assunta Pozzuoli
- Orthopedics and Orthopedic Oncology, Department of Surgery, Oncology, and Gastroenterology DiSCOG, University-Hospital of Padova, 35128, Padova, Italy
- Musculoskeletal Pathology and Oncology Laboratory, Department of Surgery, Oncology and Gastroenterology (DiSCOG), University of Padova, 35128, Padova, Italy
| | - Carlo Biz
- Orthopedics and Orthopedic Oncology, Department of Surgery, Oncology, and Gastroenterology DiSCOG, University-Hospital of Padova, 35128, Padova, Italy
| | - Pietro Ruggieri
- Orthopedics and Orthopedic Oncology, Department of Surgery, Oncology, and Gastroenterology DiSCOG, University-Hospital of Padova, 35128, Padova, Italy
| | - Chiara Romualdi
- Department of Biology, University of Padova, Padova, 35100, Italy
| | - Andrew P Lieberman
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Gopal J Babu
- Department of Cell Biology and Molecular Medicine, Rutgers, New Jersey Medical School, Newark, NJ, 07103, USA
| | - Marco Sandri
- Department of Biomedical Sciences (DBS), University of Padova, 35131, Padova, Italy
- Veneto Institute of Molecular Medicine (VIMM), Padova, 35100, Italy
| | - Bert Blaauw
- Department of Biomedical Sciences (DBS), University of Padova, 35131, Padova, Italy
- Veneto Institute of Molecular Medicine (VIMM), Padova, 35100, Italy
| | - Manuela Basso
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, 38123, Trento, Italy
| | - Maria Pennuto
- Department of Biomedical Sciences (DBS), University of Padova, 35131, Padova, Italy.
- Veneto Institute of Molecular Medicine (VIMM), Padova, 35100, Italy.
- Padova Neuroscience Center (PNC), Padova, 35100, Italy.
- Dulbecco Telethon Institute (DTI) at the Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, 38123, Trento, Italy.
| |
Collapse
|
18
|
Valentim M, Brahmbhatt A, Tupling A. Skeletal and cardiac muscle calcium transport regulation in health and disease. Biosci Rep 2022; 42:BSR20211997. [PMID: 36413081 PMCID: PMC9744722 DOI: 10.1042/bsr20211997] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 11/04/2022] [Accepted: 11/22/2022] [Indexed: 11/23/2022] Open
Abstract
In healthy muscle, the rapid release of calcium ions (Ca2+) with excitation-contraction (E-C) coupling, results in elevations in Ca2+ concentrations which can exceed 10-fold that of resting values. The sizable transient changes in Ca2+ concentrations are necessary for the activation of signaling pathways, which rely on Ca2+ as a second messenger, including those involved with force generation, fiber type distribution and hypertrophy. However, prolonged elevations in intracellular Ca2+ can result in the unwanted activation of Ca2+ signaling pathways that cause muscle damage, dysfunction, and disease. Muscle employs several calcium handling and calcium transport proteins that function to rapidly return Ca2+ concentrations back to resting levels following contraction. This review will detail our current understanding of calcium handling during the decay phase of intracellular calcium transients in healthy skeletal and cardiac muscle. We will also discuss how impairments in Ca2+ transport can occur and how mishandling of Ca2+ can lead to the pathogenesis and/or progression of skeletal muscle myopathies and cardiomyopathies.
Collapse
Affiliation(s)
- Mark A. Valentim
- Department of Kinesiology and Health Sciences, University of Waterloo, Waterloo, Ontario, N2L 3G1, Canada
| | - Aditya N. Brahmbhatt
- Department of Kinesiology and Health Sciences, University of Waterloo, Waterloo, Ontario, N2L 3G1, Canada
| | - A. Russell Tupling
- Department of Kinesiology and Health Sciences, University of Waterloo, Waterloo, Ontario, N2L 3G1, Canada
| |
Collapse
|
19
|
Özbek M, Beyaz F, Ergün E, Ergün L, Karaca H, Cabir A, Alesawi YAK. Identification of some calcium binding proteins and neural cell markers in rat testis and epididymis during postnatal development. Andrologia 2022; 54:e14633. [DOI: 10.1111/and.14633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 10/20/2022] [Accepted: 10/24/2022] [Indexed: 11/16/2022] Open
Affiliation(s)
- Mehmet Özbek
- Department of Histology and Embryology, Faculty of Veterinary Medicine Mehmet Akif Ersoy University Burdur Turkey
| | - Feyzullah Beyaz
- Department of Histology and Embryology, Faculty of Veterinary Medicine Erciyes University Kayseri Turkey
| | - Emel Ergün
- Department of Histology and Embryology, Faculty of Veterinary Medicine Ankara University Ankara Turkey
| | - Levent Ergün
- Department of Histology and Embryology, Faculty of Veterinary Medicine Ankara University Ankara Turkey
| | - Harun Karaca
- Department of Histology and Embryology, Faculty of Veterinary Medicine Mehmet Akif Ersoy University Burdur Turkey
| | - Ahmet Cabir
- Department of Histology and Embryology, Faculty of Veterinary Medicine Erciyes University Kayseri Turkey
| | - Yahy Abood Kareem Alesawi
- Department of Histology and Embryology, Faculty of Veterinary Medicine Ankara University Ankara Turkey
| |
Collapse
|
20
|
Reggiani C, Marcucci L. A controversial issue: Can mitochondria modulate cytosolic calcium and contraction of skeletal muscle fibers? J Gen Physiol 2022; 154:e202213167. [PMID: 35849108 PMCID: PMC9297197 DOI: 10.1085/jgp.202213167] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Mitochondria are characterized by a high capacity to accumulate calcium thanks to the electrochemical gradient created by the extrusion of protons in the respiratory chain. Thereby calcium can enter crossing the inner mitochondrial membrane via MCU complex, a high-capacity, low-affinity transport mechanism. Calcium uptake serves numerous purposes, among them the regulation of three dehydrogenases of the citric cycle, apoptosis via permeability transition, and, in some cell types, modulation of cytosolic calcium transients. This Review is focused on mitochondrial calcium uptake in skeletal muscle fibers and aims to reanalyze its functional impact. In particular, we ask whether mitochondrial calcium uptake is relevant for the control of cytosolic calcium transients and therefore of contractile performance. Recent data suggest that this may be the case, at least in particular conditions, as modified expression of MCU complex subunits or of proteins involved in mitochondrial dynamics and ablation of the main cytosolic calcium buffer, parvalbumin.
Collapse
Affiliation(s)
- Carlo Reggiani
- Department of Biomedical Sciences, University of Padova, Padova, Italy
- Science and Research Center Koper, Institute for Kinesiology Research, Koper, Slovenia
| | - Lorenzo Marcucci
- Department of Biomedical Sciences, University of Padova, Padova, Italy
- Center for Biosystems Dynamics Research, RIKEN, Suita, Japan
| |
Collapse
|
21
|
Quarato G, Llambi F, Guy CS, Min J, Actis M, Sun H, Narina S, Pruett-Miller SM, Peng J, Rankovic Z, Green DR. Ca 2+-mediated mitochondrial inner membrane permeabilization induces cell death independently of Bax and Bak. Cell Death Differ 2022; 29:1318-1334. [PMID: 35726022 DOI: 10.1038/s41418-022-01025-9] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 05/27/2022] [Accepted: 06/03/2022] [Indexed: 12/15/2022] Open
Abstract
The ability of mitochondria to buffer a rapid rise in cytosolic Ca2+ is a hallmark of proper cell homeostasis. Here, we employed m-3M3FBS, a putative phospholipase C (PLC) agonist, to explore the relationships between intracellular Ca2+ imbalance, mitochondrial physiology, and cell death. m-3M3FBS induced a potent dose-dependent Ca2+ release from the endoplasmic reticulum (ER), followed by a rise in intra-mitochondrial Ca2+. When the latter exceeded the organelle buffering capacity, an abrupt mitochondrial inner membrane permeabilization (MIMP) occurred, releasing matrix contents into the cytosol. MIMP was followed by cell death that was independent of Bcl-2 family members and inhibitable by the intracellular Ca2+ chelator BAPTA-AM. Cyclosporin A (CsA), capable of blocking the mitochondrial permeability transition (MPT), completely prevented cell death induced by m-3M3FBS. However, CsA acted upstream of mitochondria by preventing Ca2+ release from ER stores. Therefore, loss of Ca2+ intracellular balance and mitochondrial Ca2+ overload followed by MIMP induced a cell death process that is distinct from Bcl-2 family-regulated mitochondrial outer membrane permeabilization (MOMP). Further, the inhibition of cell death by CsA or its analogues can be independent of effects on the MPT.
Collapse
Affiliation(s)
- Giovanni Quarato
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA.
| | - Fabien Llambi
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA.,Relay Therapeutics, Cambridge, MA, 02139, USA
| | - Cliff S Guy
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Jaeki Min
- Department of Chemical Biology & Therapeutic, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA.,Amgen Inc., Thousand Oaks, CA, 91320, USA
| | - Marisa Actis
- Department of Chemical Biology & Therapeutic, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Huan Sun
- Department of Structural Biology, Department of Developmental Neurobiology, Center for Proteomics and Metabolomics, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Shilpa Narina
- Department of Cell and Molecular Biology and The Center for Advanced Genome Engineering, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Shondra M Pruett-Miller
- Department of Cell and Molecular Biology and The Center for Advanced Genome Engineering, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Junmin Peng
- Department of Structural Biology, Department of Developmental Neurobiology, Center for Proteomics and Metabolomics, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Zoran Rankovic
- Department of Chemical Biology & Therapeutic, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Douglas R Green
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA.
| |
Collapse
|
22
|
Chambers PJ, Juracic ES, Fajardo VA, Tupling AR. The role of SERCA and sarcolipin in adaptive muscle remodeling. Am J Physiol Cell Physiol 2022; 322:C382-C394. [PMID: 35044855 DOI: 10.1152/ajpcell.00198.2021] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Sarcolipin (SLN) is a small integral membrane protein that regulates the sarco(endo)plasmic reticulum Ca2+-ATPase (SERCA) pump. When bound to SERCA, SLN reduces the apparent Ca2+ affinity of SERCA and uncouples SERCA Ca2+ transport from its ATP consumption. As such, SLN plays a direct role in altering skeletal muscle relaxation and energy expenditure. Interestingly, the expression of SLN is dynamic during times of muscle adaptation, where large increases in SLN content are found in response to development, atrophy, overload and disease. Several groups have suggested that increases in SLN, especially in dystrophic muscle, are deleterious to muscle function and exacerbate already abhorrent intracellular Ca2+ levels. However, there is also significant evidence to show that increased SLN content is a beneficial adaptive mechanism which protects the SERCA pump and activates Ca2+ signaling and adaptive remodeling during times of cell stress. In this review, we first discuss the role for SLN in healthy muscle during both development and overload, where SLN has been shown to activate Ca2+ signaling to promote mitochondrial biogenesis, fibre type shifts and muscle hypertrophy. Then, with respect to muscle disease, we summarize the discrepancies in the literature as to whether SLN upregulation is adaptive or maladaptive in nature. This review is the first to offer the concept of SLN hormesis in muscle disease, wherein both too much and too little SLN are detrimental to muscle health. Finally, the underlying mechanisms which activate SLN upregulation are discussed, specifically acknowledging a potential positive feedback loop between SLN and Ca2+ signaling molecules.
Collapse
Affiliation(s)
- Paige J Chambers
- Department of Kinesiology and Health Sciences, University of Waterloo, Waterloo, Ontario, Canada
| | - Emma S Juracic
- Department of Kinesiology and Health Sciences, University of Waterloo, Waterloo, Ontario, Canada
| | - Val A Fajardo
- Department Department of Kinesiology, Brock University, St. Catharines, Ontario, Canada.,Centre for Bone and Muscle Health, Brock University, St. Catharines, Ontario, Canada
| | - A Russell Tupling
- Department of Kinesiology and Health Sciences, University of Waterloo, Waterloo, Ontario, Canada
| |
Collapse
|