1
|
Tabuchi A, Poole DC, Kano Y. Intracellular Ca 2+ After Eccentric Muscle Contractions: Key Role for Ryanodine Receptors. Exerc Sport Sci Rev 2025; 53:23-30. [PMID: 39262047 DOI: 10.1249/jes.0000000000000348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/13/2024]
Abstract
Eccentric contractions (ECC) induce excessive intracellular calcium ion (Ca 2+ ) accumulation and muscle structural damage in localized regions of the muscle fibers. In this investigation, we present the novel hypothesis that the ryanodine receptor (RyR) plays a central role in evoking a Ca 2+ dynamics profile that is markedly distinguishable from other muscle adaptive responses.
Collapse
Affiliation(s)
| | - David C Poole
- Departments of Anatomy and Physiology and Kinesiology, Kansas State University, Manhattan, KS
| | | |
Collapse
|
2
|
Khodabukus A, Prabhu NK, Roberts T, Buldo M, Detwiler A, Fralish ZD, Kondash ME, Truskey GA, Koves TR, Bursac N. Bioengineered Model of Human LGMD2B Skeletal Muscle Reveals Roles of Intracellular Calcium Overload in Contractile and Metabolic Dysfunction in Dysferlinopathy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2400188. [PMID: 38887849 PMCID: PMC11336985 DOI: 10.1002/advs.202400188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 03/19/2024] [Indexed: 06/20/2024]
Abstract
Dysferlin is a multi-functional protein that regulates membrane resealing, calcium homeostasis, and lipid metabolism in skeletal muscle. Genetic loss of dysferlin results in limb girdle muscular dystrophy 2B/2R (LGMD2B/2R) and other dysferlinopathies - rare untreatable muscle diseases that lead to permanent loss of ambulation in humans. The mild disease severity in dysferlin-deficient mice and diverse genotype-phenotype relationships in LGMD2B patients have prompted the development of new in vitro models for personalized studies of dysferlinopathy. Here the first 3-D tissue-engineered hiPSC-derived skeletal muscle ("myobundle") model of LGMD2B is described that exhibits compromised contractile function, calcium-handling, and membrane repair, and transcriptomic changes indicative of impaired oxidative metabolism and mitochondrial dysfunction. In response to the fatty acid (FA) challenge, LGMD2B myobundles display mitochondrial deficits and intracellular lipid droplet (LD) accumulation. Treatment with the ryanodine receptor (RyR) inhibitor dantrolene or the dissociative glucocorticoid vamorolone restores LGMD2B contractility, improves membrane repair, and reduces LD accumulation. Lastly, it is demonstrated that chemically induced chronic RyR leak in healthy myobundles phenocopies LGMD2B contractile and metabolic deficit, but not the loss of membrane repair capacity. Together, these results implicate intramyocellular Ca2+ leak as a critical driver of dysferlinopathic phenotype and validate the myobundle system as a platform to study LGMD2B pathogenesis.
Collapse
Affiliation(s)
| | - Neel K. Prabhu
- Department of Biomedical EngineeringDuke UniversityDurhamNC27708USA
| | - Taylor Roberts
- Department of Biomedical EngineeringDuke UniversityDurhamNC27708USA
| | - Meghan Buldo
- Department of Biomedical EngineeringDuke UniversityDurhamNC27708USA
| | - Amber Detwiler
- Department of Biomedical EngineeringDuke UniversityDurhamNC27708USA
| | | | - Megan E. Kondash
- Department of Biomedical EngineeringDuke UniversityDurhamNC27708USA
| | | | - Timothy R. Koves
- Duke Molecular Physiology InstituteDuke UniversityDurhamNC27708USA
| | - Nenad Bursac
- Department of Biomedical EngineeringDuke UniversityDurhamNC27708USA
| |
Collapse
|
3
|
Yumura S. Wound Repair of the Cell Membrane: Lessons from Dictyostelium Cells. Cells 2024; 13:341. [PMID: 38391954 PMCID: PMC10886852 DOI: 10.3390/cells13040341] [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: 12/20/2023] [Revised: 01/30/2024] [Accepted: 02/08/2024] [Indexed: 02/24/2024] Open
Abstract
The cell membrane is frequently subjected to damage, either through physical or chemical means. The swift restoration of the cell membrane's integrity is crucial to prevent the leakage of intracellular materials and the uncontrolled influx of extracellular ions. Consequently, wound repair plays a vital role in cell survival, akin to the importance of DNA repair. The mechanisms involved in wound repair encompass a series of events, including ion influx, membrane patch formation, endocytosis, exocytosis, recruitment of the actin cytoskeleton, and the elimination of damaged membrane sections. Despite the absence of a universally accepted general model, diverse molecular models have been proposed for wound repair in different organisms. Traditional wound methods not only damage the cell membrane but also impact intracellular structures, including the underlying cortical actin networks, microtubules, and organelles. In contrast, the more recent improved laserporation selectively targets the cell membrane. Studies on Dictyostelium cells utilizing this method have introduced a novel perspective on the wound repair mechanism. This review commences by detailing methods for inducing wounds and subsequently reviews recent developments in the field.
Collapse
Affiliation(s)
- Shigehiko Yumura
- Graduate School of Sciences and Technology for Innovation, Yamaguchi University, Yamaguchi 753-8511, Japan
| |
Collapse
|
4
|
Bittel DC, Jaiswal JK. Early Endosomes Undergo Calcium-Triggered Exocytosis and Enable Repair of Diffuse and Focal Plasma Membrane Injury. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2300245. [PMID: 37705135 PMCID: PMC10667805 DOI: 10.1002/advs.202300245] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 06/27/2023] [Indexed: 09/15/2023]
Abstract
Cells are routinely exposed to agents that cause plasma membrane (PM) injury. While pore-forming toxins (PFTs), and chemicals cause nanoscale holes dispersed throughout the PM, mechanical trauma causes focal lesions in the PM. To examine if these distinct injuries share common repair mechanism, membrane trafficking is monitored as the PM repairs from such injuries. During the course of repair, dispersed PM injury by the PFT Streptolysin O activates endocytosis, while focal mechanical injury to the PM inhibits endocytosis. Consequently, acute block of endocytosis prevents repair of diffuse, but not of focal injury. In contrast, a chronic block in endocytosis depletes cells of early endosomes and inhibits repair of focal injury. This study finds that both focal and diffuse PM injury activate Ca2+ -triggered exocytosis of early endosomes. The use of markers including endocytosed cargo, Rab5, Rab11, and VAMP3, all reveal injury-triggered exocytosis of early endosomes. Inhibiting Rab5 prevents injury-triggered early endosome exocytosis and phenocopies the failed PM repair of cells chronically depleted of early endosomes. These results identify early endosomes as a Ca2+ -regulated exocytic compartment, and uncover the requirement of their dual functions - endocytosis and regulated exocytosis, to differentially support PM repair based on the nature of the injury.
Collapse
Affiliation(s)
- Daniel C. Bittel
- Center for Genetic Medicine ResearchChildren's National Research Institute7144 13th Pl NWWashington, DC20012USA
| | - Jyoti K. Jaiswal
- Center for Genetic Medicine ResearchChildren's National Research Institute7144 13th Pl NWWashington, DC20012USA
- Department of Genomics and Precision MedicineGeorge Washington University School of Medicine and Health SciencesWashington, DC20012USA
| |
Collapse
|
5
|
Acin‐Perez R, Benincá C, Fernandez del Rio L, Shu C, Baghdasarian S, Zanette V, Gerle C, Jiko C, Khairallah R, Khan S, Rincon Fernandez Pacheco D, Shabane B, Erion K, Masand R, Dugar S, Ghenoiu C, Schreiner G, Stiles L, Liesa M, Shirihai OS. Inhibition of ATP synthase reverse activity restores energy homeostasis in mitochondrial pathologies. EMBO J 2023; 42:e111699. [PMID: 36912136 PMCID: PMC10183817 DOI: 10.15252/embj.2022111699] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 01/24/2023] [Accepted: 01/25/2023] [Indexed: 03/14/2023] Open
Abstract
The maintenance of cellular function relies on the close regulation of adenosine triphosphate (ATP) synthesis and hydrolysis. ATP hydrolysis by mitochondrial ATP Synthase (CV) is induced by loss of proton motive force and inhibited by the mitochondrial protein ATPase inhibitor (ATPIF1). The extent of CV hydrolytic activity and its impact on cellular energetics remains unknown due to the lack of selective hydrolysis inhibitors of CV. We find that CV hydrolytic activity takes place in coupled intact mitochondria and is increased by respiratory chain defects. We identified (+)-Epicatechin as a selective inhibitor of ATP hydrolysis that binds CV while preventing the binding of ATPIF1. In cells with Complex-III deficiency, we show that inhibition of CV hydrolytic activity by (+)-Epichatechin is sufficient to restore ATP content without restoring respiratory function. Inhibition of CV-ATP hydrolysis in a mouse model of Duchenne Muscular Dystrophy is sufficient to improve muscle force without any increase in mitochondrial content. We conclude that the impact of compromised mitochondrial respiration can be lessened using hydrolysis-selective inhibitors of CV.
Collapse
Affiliation(s)
- Rebeca Acin‐Perez
- Department of Medicine, Endocrinology, David Geffen School of MedicineUniversity of CaliforniaLos AngelesCAUSA
- Metabolism Theme, David Geffen School of MedicineUniversity of CaliforniaLos AngelesCAUSA
| | - Cristiane Benincá
- Department of Medicine, Endocrinology, David Geffen School of MedicineUniversity of CaliforniaLos AngelesCAUSA
- Metabolism Theme, David Geffen School of MedicineUniversity of CaliforniaLos AngelesCAUSA
| | - Lucia Fernandez del Rio
- Department of Medicine, Endocrinology, David Geffen School of MedicineUniversity of CaliforniaLos AngelesCAUSA
- Metabolism Theme, David Geffen School of MedicineUniversity of CaliforniaLos AngelesCAUSA
| | - Cynthia Shu
- Department of Medicine, Endocrinology, David Geffen School of MedicineUniversity of CaliforniaLos AngelesCAUSA
- Metabolism Theme, David Geffen School of MedicineUniversity of CaliforniaLos AngelesCAUSA
| | - Siyouneh Baghdasarian
- Department of Medicine, Endocrinology, David Geffen School of MedicineUniversity of CaliforniaLos AngelesCAUSA
- Metabolism Theme, David Geffen School of MedicineUniversity of CaliforniaLos AngelesCAUSA
| | - Vanessa Zanette
- Department of BioinformaticsUniversity Federal of ParanaCuritibaBrazil
| | - Christoph Gerle
- Institute for Protein ResearchOsaka UniversitySuitaJapan
- RIKEN SPring‐8 CenterSayo‐gunJapan
| | - Chimari Jiko
- Institute for Integrated Radiation and Nuclear ScienceKyoto UniversityKyotoJapan
| | | | | | | | - Byourak Shabane
- Department of Medicine, Endocrinology, David Geffen School of MedicineUniversity of CaliforniaLos AngelesCAUSA
- Metabolism Theme, David Geffen School of MedicineUniversity of CaliforniaLos AngelesCAUSA
| | | | | | | | | | | | - Linsey Stiles
- Department of Medicine, Endocrinology, David Geffen School of MedicineUniversity of CaliforniaLos AngelesCAUSA
- Metabolism Theme, David Geffen School of MedicineUniversity of CaliforniaLos AngelesCAUSA
- Department of Molecular and Medical PharmacologyUniversity of CaliforniaLos AngelesCAUSA
| | - Marc Liesa
- Department of Medicine, Endocrinology, David Geffen School of MedicineUniversity of CaliforniaLos AngelesCAUSA
- Metabolism Theme, David Geffen School of MedicineUniversity of CaliforniaLos AngelesCAUSA
- Department of Molecular and Medical PharmacologyUniversity of CaliforniaLos AngelesCAUSA
- Molecular Cellular Integrative PhysiologyUniversity of CaliforniaLos AngelesCAUSA
- Institut de Biologia Molecular de Barcelona, IBMB, CSICBarcelonaCataloniaSpain
| | - Orian S Shirihai
- Department of Medicine, Endocrinology, David Geffen School of MedicineUniversity of CaliforniaLos AngelesCAUSA
- Metabolism Theme, David Geffen School of MedicineUniversity of CaliforniaLos AngelesCAUSA
- Department of Molecular and Medical PharmacologyUniversity of CaliforniaLos AngelesCAUSA
- Molecular Cellular Integrative PhysiologyUniversity of CaliforniaLos AngelesCAUSA
| |
Collapse
|
6
|
The role of the dystrophin glycoprotein complex in muscle cell mechanotransduction. Commun Biol 2022; 5:1022. [PMID: 36168044 PMCID: PMC9515174 DOI: 10.1038/s42003-022-03980-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Accepted: 09/12/2022] [Indexed: 11/09/2022] Open
Abstract
Dystrophin is the central protein of the dystrophin-glycoprotein complex (DGC) in skeletal and heart muscle cells. Dystrophin connects the actin cytoskeleton to the extracellular matrix (ECM). Severing the link between the ECM and the intracellular cytoskeleton has a devastating impact on the homeostasis of skeletal muscle cells, leading to a range of muscular dystrophies. In addition, the loss of a functional DGC leads to progressive dilated cardiomyopathy and premature death. Dystrophin functions as a molecular spring and the DGC plays a critical role in maintaining the integrity of the sarcolemma. Additionally, evidence is accumulating, linking the DGC to mechanosignalling, albeit this role is still less understood. This review article aims at providing an up-to-date perspective on the DGC and its role in mechanotransduction. We first discuss the intricate relationship between muscle cell mechanics and function, before examining the recent research for a role of the dystrophin glycoprotein complex in mechanotransduction and maintaining the biomechanical integrity of muscle cells. Finally, we review the current literature to map out how DGC signalling intersects with mechanical signalling pathways to highlight potential future points of intervention, especially with a focus on cardiomyopathies. A review of the function of the Dystrophic Glycoprotein Complex (DGC) in mechanosignaling provides an overview of the various components of DGC and potential mechanopathogenic mechanisms, particularly as they relate to muscular dystrophy.
Collapse
|
7
|
Ito M, Tauscher-Wisniewski S, Smulders RA, Wojtkowski T, Yamada A, Koibuchi A, Uz T, Marek GJ, Goldwater RD. Single- and multiple-dose safety, tolerability, pharmacokinetic, and pharmacodynamic profiles of ASP0367, or bocidelpar sulfate, a novel modulator of peroxisome proliferator-activated receptor delta in healthy adults: Results from a phase 1 study. Muscle Nerve 2021; 65:110-120. [PMID: 34642949 PMCID: PMC9298414 DOI: 10.1002/mus.27436] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 10/04/2021] [Accepted: 10/09/2021] [Indexed: 01/24/2023]
Abstract
Introduction/Aims ASP0367, or bocidelpar sulfate, is an orally administered small molecule that potently and selectively modulates peroxisome proliferator–activated receptor δ (PPARδ) to address mitochondrial dysfunction occurring in diseases including primary mitochondrial myopathy and Duchenne muscular dystrophy. The objectives of this first‐in‐human trial were to evaluate the safety/tolerability, pharmacokinetics, and pharmacodynamics of ASP0367 in healthy participants. Methods In this double‐blind phase 1 study, adult participants were randomized to single or multiple ascending oral doses of ASP0367 or placebo. The study duration was 1 and 14 days, respectively. Pharmacokinetic parameters under fed conditions were also evaluated. Results A total of 64 (single‐dose cohort) and 37 (multiple‐dose cohort) participants were included in the study. After single doses of 1 to 120 mg, ASP0367 was rapidly absorbed, with median time to maximum plasma concentration (tmax) of 1.50 to 2.24 hours under fasting conditions; ASP0367 concentrations declined in a multiphasic manner after reaching maximum plasma concentration. Under fed conditions, tmax was delayed 1.7 hours. After multiple once‐daily doses, mean half‐life of ASP0367 10 to 75 mg ranged from 14.1 to 17.5 hours; steady state was reached after 4 days. Negligible accumulation was observed after repeated dosing. No participants receiving ASP0367 discontinued treatment, and all treatment‐emergent adverse events were mild to moderate in severity; none were considered drug‐related. No clinically significant changes were observed on laboratory or electrocardiographic evaluation. Treatment‐ and dose‐dependent upregulation of six PPARδ target genes was observed with single and multiple doses of ASP0367. Discussion ASP0367, or bocidelpar sulfate, was well tolerated; rapid absorption, roughly dose‐proportional bioavailability, and effects on PPARδ target genes were demonstrated in healthy adult participants.
Collapse
Affiliation(s)
- Mototsugu Ito
- Astellas Pharma Global Development, Inc., Northbrook, Illinois, USA
| | | | | | | | | | | | - Tolga Uz
- Astellas Pharma Global Development, Inc., Northbrook, Illinois, USA
| | - Gerard J Marek
- Astellas Pharma Global Development, Inc., Northbrook, Illinois, USA
| | | |
Collapse
|
8
|
Actin Cytoskeletal Dynamics in Single-Cell Wound Repair. Int J Mol Sci 2021; 22:ijms221910886. [PMID: 34639226 PMCID: PMC8509258 DOI: 10.3390/ijms221910886] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Revised: 10/04/2021] [Accepted: 10/04/2021] [Indexed: 11/17/2022] Open
Abstract
The plasma membrane protects the eukaryotic cell from its surroundings and is essential for cell viability; thus, it is crucial that membrane disruptions are repaired quickly to prevent immediate dyshomeostasis and cell death. Accordingly, cells have developed efficient repair mechanisms to rapidly reseal ruptures and reestablish membrane integrity. The cortical actin cytoskeleton plays an instrumental role in both plasma membrane resealing and restructuring in response to damage. Actin directly aids membrane repair or indirectly assists auxiliary repair mechanisms. Studies investigating single-cell wound repair have often focused on the recruitment and activation of specialized repair machinery, despite the undeniable need for rapid and dynamic cortical actin modulation; thus, the role of the cortical actin cytoskeleton during wound repair has received limited attention. This review aims to provide a comprehensive overview of membrane repair mechanisms directly or indirectly involving cortical actin cytoskeletal remodeling.
Collapse
|
9
|
Chandra G, Sreetama SC, Mázala DAG, Charton K, VanderMeulen JH, Richard I, Jaiswal JK. Endoplasmic reticulum maintains ion homeostasis required for plasma membrane repair. J Cell Biol 2021; 220:211873. [PMID: 33688936 PMCID: PMC7953257 DOI: 10.1083/jcb.202006035] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Revised: 12/11/2020] [Accepted: 01/27/2021] [Indexed: 12/15/2022] Open
Abstract
Of the many crucial functions of the ER, homeostasis of physiological calcium increase is critical for signaling. Plasma membrane (PM) injury causes a pathological calcium influx. Here, we show that the ER helps clear this surge in cytoplasmic calcium through an ER-resident calcium pump, SERCA, and a calcium-activated ion channel, Anoctamin 5 (ANO5). SERCA imports calcium into the ER, and ANO5 supports this by maintaining electroneutrality of the ER lumen through anion import. Preventing either of these transporter activities causes cytosolic calcium overload and disrupts PM repair (PMR). ANO5 deficit in limb girdle muscular dystrophy 2L (LGMD2L) patient cells compromises their cytosolic and ER calcium homeostasis. By generating a mouse model of LGMD2L, we find that PM injury causes cytosolic calcium overload and compromises the ability of ANO5-deficient myofibers to repair. Addressing calcium overload in ANO5-deficient myofibers enables them to repair, supporting the requirement of the ER in calcium homeostasis in injured cells and facilitating PMR.
Collapse
Affiliation(s)
- Goutam Chandra
- Center of Genetic Medicine Research, Children's National Health System, Washington, DC
| | - Sen Chandra Sreetama
- Center of Genetic Medicine Research, Children's National Health System, Washington, DC
| | - Davi A G Mázala
- Center of Genetic Medicine Research, Children's National Health System, Washington, DC
| | - Karine Charton
- Généthon, Institut National de la Santé et de la Recherche Médicale, U951, INTEGRARE Research Unit, University Paris-Saclay, Evry, France
| | - Jack H VanderMeulen
- Center of Genetic Medicine Research, Children's National Health System, Washington, DC
| | - Isabelle Richard
- Généthon, Institut National de la Santé et de la Recherche Médicale, U951, INTEGRARE Research Unit, University Paris-Saclay, Evry, France
| | - Jyoti K Jaiswal
- Center of Genetic Medicine Research, Children's National Health System, Washington, DC.,Department of Genomics and Precision Medicine, George Washington University School of Medicine and Health Sciences, Washington, DC
| |
Collapse
|
10
|
Splitting up to heal: mitochondrial shape regulates signaling for focal membrane repair. Biochem Soc Trans 2021; 48:1995-2002. [PMID: 32985660 DOI: 10.1042/bst20200120] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Revised: 09/01/2020] [Accepted: 09/07/2020] [Indexed: 12/19/2022]
Abstract
Mitochondria are central to the health of eukaryotic cells. While commonly known for their bioenergetic role, mitochondria also function as signaling organelles that regulate cell stress responses capable of restoring homeostasis or leading the stressed cell to eventual death. Damage to the plasma membrane is a potentially fatal stressor incurred by all cells. Repairing plasma membrane damage requires cells to mount a rapid and localized response to injury. Accumulating evidence has identified a role for mitochondria as an important facilitator of this acute and localized repair response. However, as mitochondria are organized in a cell-wide, interconnected network, it is unclear how they collectively sense and respond to a focal injury. Here we will discuss how mitochondrial shape change is an integral part of this localized repair response. Mitochondrial fragmentation spatially restricts beneficial repair signaling, enabling a localized response to focal injury. Conservation of mitochondrial fragmentation in response to cell and tissue damage across species demonstrates that this is a universal pro-survival adaptation to injury and suggests that mitochondrial fragmentation may provide cells a mechanism to facilitate localized signaling in contexts beyond repairing plasma membrane injury.
Collapse
|
11
|
Rovira Gonzalez YI, Moyer AL, LeTexier NJ, Bratti AD, Feng S, Peña V, Sun C, Pulcastro H, Liu T, Iyer SR, Lovering RM, O'Rourke B, Wagner KR. Mss51 deletion increases endurance and ameliorates histopathology in the mdx mouse model of Duchenne muscular dystrophy. FASEB J 2021; 35:e21276. [PMID: 33423297 DOI: 10.1096/fj.202002106rr] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 11/25/2020] [Accepted: 11/30/2020] [Indexed: 11/11/2022]
Abstract
Mitochondrial derangement is an important contributor to the pathophysiology of muscular dystrophies and may be among the earliest cellular deficits. We have previously shown that disruption of Mss51, a mammalian skeletal muscle protein that localizes to the mitochondria, results in enhanced muscle oxygen consumption rate, increased endurance capacity, and improved limb muscle strength in mice with wildtype background. Here, we investigate whether Mss51 deletion in the mdx murine model of Duchenne muscular dystrophy (mdx-Mss51 KO) counteracts the muscle pathology and mitochondrial irregularities observed in mdx mice. We found that mdx-Mss51 KO mice had increased myofiber oxygen consumption rates and an amelioration of muscle histopathology compared to mdx counterparts. This corresponded with greater treadmill endurance and less percent fatigue in muscle physiology, but no improvement in forelimb grip strength or limb muscle force production. These findings suggest that although Mss51 deletion ameliorates the skeletal muscle mitochondrial respiration defects in mdx and improves fatigue resistance in vivo, the lack of improvement in force production suggests that this target alone may be insufficient for a therapeutic effect.
Collapse
Affiliation(s)
- Yazmin I Rovira Gonzalez
- The Hugo W. Moser Research Institute, Kennedy Krieger Institute, Baltimore, MD, USA.,Cellular and Molecular Medicine Graduate Program, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Adam L Moyer
- The Hugo W. Moser Research Institute, Kennedy Krieger Institute, Baltimore, MD, USA.,Cellular and Molecular Medicine Graduate Program, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Nicolas J LeTexier
- The Hugo W. Moser Research Institute, Kennedy Krieger Institute, Baltimore, MD, USA
| | - August D Bratti
- The Hugo W. Moser Research Institute, Kennedy Krieger Institute, Baltimore, MD, USA
| | - Siyuan Feng
- The Hugo W. Moser Research Institute, Kennedy Krieger Institute, Baltimore, MD, USA
| | - Vanessa Peña
- The Hugo W. Moser Research Institute, Kennedy Krieger Institute, Baltimore, MD, USA
| | - Congshan Sun
- The Hugo W. Moser Research Institute, Kennedy Krieger Institute, Baltimore, MD, USA.,Departments of Neurology and Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Hannah Pulcastro
- The Hugo W. Moser Research Institute, Kennedy Krieger Institute, Baltimore, MD, USA
| | - Ting Liu
- Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Shama R Iyer
- Department of Orthopaedics, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Richard M Lovering
- Department of Orthopaedics, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Brian O'Rourke
- Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Kathryn R Wagner
- The Hugo W. Moser Research Institute, Kennedy Krieger Institute, Baltimore, MD, USA.,Departments of Neurology and Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, USA
| |
Collapse
|
12
|
Ren K, Wang L, Wang L, Du Q, Cao J, Jin Q, An G, Li N, Dang L, Tian Y, Wang Y, Sun J. Investigating Transcriptional Dynamics Changes and Time-Dependent Marker Gene Expression in the Early Period After Skeletal Muscle Injury in Rats. Front Genet 2021; 12:650874. [PMID: 34220936 PMCID: PMC8248501 DOI: 10.3389/fgene.2021.650874] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 05/07/2021] [Indexed: 12/21/2022] Open
Abstract
Following skeletal muscle injury (SMI), from post-injury reaction to repair consists of a complex series of dynamic changes. However, there is a paucity of research on detailed transcriptional dynamics and time-dependent marker gene expression in the early stages after SMI. In this study, skeletal muscle tissue in rats was taken at 4 to 48 h after injury for next-generation sequencing. We examined the transcriptional kinetics characteristics during above time periods after injury. STEM and maSigPro were used to screen time-correlated genes. Integrating 188 time-correlated genes with 161 genes in each time-related gene module by WGCNA, we finally identified 18 network-node regulatory genes after SMI. Histological staining analyses confirmed the mechanisms underlying changes in the tissue damage to repair process. Our research linked a variety of dynamic biological processes with specific time periods and provided insight into the characteristics of transcriptional dynamics, as well as screened time-related biological indicators with biological significance in the early stages after SMI.
Collapse
Affiliation(s)
- Kang Ren
- School of Forensic Medicine, Shanxi Medical University, Jinzhong, China.,Department of Basic Medicine, Changzhi Medical College, Changzhi, China
| | - Liangliang Wang
- School of Forensic Medicine, Shanxi Medical University, Jinzhong, China
| | - Liang Wang
- School of Forensic Medicine, Shanxi Medical University, Jinzhong, China
| | - Qiuxiang Du
- School of Forensic Medicine, Shanxi Medical University, Jinzhong, China
| | - Jie Cao
- School of Forensic Medicine, Shanxi Medical University, Jinzhong, China
| | - Qianqian Jin
- School of Forensic Medicine, Shanxi Medical University, Jinzhong, China
| | - Guoshuai An
- School of Forensic Medicine, Shanxi Medical University, Jinzhong, China
| | - Na Li
- School of Forensic Medicine, Shanxi Medical University, Jinzhong, China
| | - Lihong Dang
- School of Forensic Medicine, Shanxi Medical University, Jinzhong, China
| | - Yingjie Tian
- School of Forensic Medicine, Shanxi Medical University, Jinzhong, China
| | - Yingyuan Wang
- School of Forensic Medicine, Shanxi Medical University, Jinzhong, China
| | - Junhong Sun
- School of Forensic Medicine, Shanxi Medical University, Jinzhong, China
| |
Collapse
|
13
|
Annexins and Membrane Repair Dysfunctions in Muscular Dystrophies. Int J Mol Sci 2021; 22:ijms22105276. [PMID: 34067866 PMCID: PMC8155887 DOI: 10.3390/ijms22105276] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 05/11/2021] [Accepted: 05/12/2021] [Indexed: 11/16/2022] Open
Abstract
Muscular dystrophies constitute a group of genetic disorders that cause weakness and progressive loss of skeletal muscle mass. Among them, Miyoshi muscular dystrophy 1 (MMD1), limb girdle muscular dystrophy type R2 (LGMDR2/2B), and LGMDR12 (2L) are characterized by mutation in gene encoding key membrane-repair protein, which leads to severe dysfunctions in sarcolemma repair. Cell membrane disruption is a physiological event induced by mechanical stress, such as muscle contraction and stretching. Like many eukaryotic cells, muscle fibers possess a protein machinery ensuring fast resealing of damaged plasma membrane. Members of the annexins A (ANXA) family belong to this protein machinery. ANXA are small soluble proteins, twelve in number in humans, which share the property of binding to membranes exposing negatively-charged phospholipids in the presence of calcium (Ca2+). Many ANXA have been reported to participate in membrane repair of varied cell types and species, including human skeletal muscle cells in which they may play a collective role in protection and repair of the sarcolemma. Here, we discuss the participation of ANXA in membrane repair of healthy skeletal muscle cells and how dysregulation of ANXA expression may impact the clinical severity of muscular dystrophies.
Collapse
|
14
|
Horn A, Raavicharla S, Shah S, Cox D, Jaiswal JK. Mitochondrial fragmentation enables localized signaling required for cell repair. J Cell Biol 2021; 219:151605. [PMID: 32236517 PMCID: PMC7199862 DOI: 10.1083/jcb.201909154] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 02/05/2020] [Accepted: 03/09/2020] [Indexed: 01/08/2023] Open
Abstract
Plasma membrane injury can cause lethal influx of calcium, but cells survive by mounting a polarized repair response targeted to the wound site. Mitochondrial signaling within seconds after injury enables this response. However, as mitochondria are distributed throughout the cell in an interconnected network, it is unclear how they generate a spatially restricted signal to repair the plasma membrane wound. Here we show that calcium influx and Drp1-mediated, rapid mitochondrial fission at the injury site help polarize the repair response. Fission of injury-proximal mitochondria allows for greater amplitude and duration of calcium increase in these mitochondria, allowing them to generate local redox signaling required for plasma membrane repair. Drp1 knockout cells and patient cells lacking the Drp1 adaptor protein MiD49 fail to undergo injury-triggered mitochondrial fission, preventing polarized mitochondrial calcium increase and plasma membrane repair. Although mitochondrial fission is considered to be an indicator of cell damage and death, our findings identify that mitochondrial fission generates localized signaling required for cell survival.
Collapse
Affiliation(s)
- Adam Horn
- Children's National Health System, Center for Genetic Medicine Research, Washington, DC
| | - Shreya Raavicharla
- Children's National Health System, Center for Genetic Medicine Research, Washington, DC
| | - Sonna Shah
- Children's National Health System, Center for Genetic Medicine Research, Washington, DC
| | - Dan Cox
- John Walton Muscular Dystrophy Research Centre, Newcastle University and Newcastle Hospitals National Health Service Foundation Trust, Newcastle upon Tyne, UK
| | - Jyoti K Jaiswal
- Children's National Health System, Center for Genetic Medicine Research, Washington, DC.,Department of Genomics and Precision Medicine, George Washington University School of Medicine and Health Sciences, Washington, DC
| |
Collapse
|
15
|
McDonald CM, Ramirez‐Sanchez I, Oskarsson B, Joyce N, Aguilar C, Nicorici A, Dayan J, Goude E, Abresch RT, Villarreal F, Ceballos G, Perkins G, Dugar S, Schreiner G, Henricson EK. (-)-Epicatechin induces mitochondrial biogenesis and markers of muscle regeneration in adults with Becker muscular dystrophy. Muscle Nerve 2021; 63:239-249. [PMID: 33125736 PMCID: PMC7898288 DOI: 10.1002/mus.27108] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2019] [Revised: 10/22/2020] [Accepted: 10/25/2020] [Indexed: 12/31/2022]
Abstract
INTRODUCTION We conducted an open-label study to examine the effects of the flavonoid (-)-epicatechin in seven ambulatory adult patients with Becker muscular dystrophy (BMD). METHODS Seven participants received (-)-epicatechin 50 mg twice per day for 8 weeks. Pre- and postprocedures included biceps brachii biopsy to assess muscle structure and growth-relevant endpoints by western blotting, mitochondria volume measurement, and cristae abundance by electron microscopy, graded exercise testing, and muscle strength and function tests. RESULTS Western blotting showed significantly increased levels of enzymes modulating cellular bioenergetics (liver kinase B1 and 5'-adenosine monophosphate-activated protein kinase). Peroxisome proliferator-activated receptor gamma coactivator-1alpha, a transcriptional coactivator of genes involved in mitochondrial biogenesis and cristae-associated mitofilin levels, increased as did cristae abundance. Muscle and plasma follistatin increased significantly while myostatin decreased. Markers of skeletal muscle regeneration myogenin, myogenic regulatory factor-5, myoblast determination protein 1, myocyte enhancer factor-2, and structure-associated proteins, including dysferlin, utrophin, and intracellular creatine kinase, also increased. Exercise testing demonstrated decreased heart rate, maximal oxygen consumption per kilogram, and plasma lactate levels at defined workloads. Tissue saturation index improved in resting and postexercise states. DISCUSSION (-)-Epicatechin, an exercise mimetic, appears to have short-term positive effects on tissue biomarkers indicative of mitochondrial biogenesis and muscle regeneration, and produced improvements in graded exercise testing parameters in patients with BMD.
Collapse
Affiliation(s)
- Craig M. McDonald
- Department of Physical Medicine and RehabilitationUniversity of California Davis School of MedicineSacramentoCaliforniaUSA
| | - Israel Ramirez‐Sanchez
- Division of Cardiology, Department of Internal MedicineUniversity of California at San DiegoSan DiegoCaliforniaUSA
- Escuela Superior de Medicina, Seccion de Posgrado e Investigacion, del Instituto Politécnico NacionalMexico CityMexico
| | | | - Nanette Joyce
- Department of Physical Medicine and RehabilitationUniversity of California Davis School of MedicineSacramentoCaliforniaUSA
| | - Candace Aguilar
- Department of Physical Medicine and RehabilitationUniversity of California Davis School of MedicineSacramentoCaliforniaUSA
| | - Alina Nicorici
- Department of Physical Medicine and RehabilitationUniversity of California Davis School of MedicineSacramentoCaliforniaUSA
| | - Jonathan Dayan
- Department of Physical Medicine and RehabilitationUniversity of California Davis School of MedicineSacramentoCaliforniaUSA
| | - Erica Goude
- Department of Physical Medicine and RehabilitationUniversity of California Davis School of MedicineSacramentoCaliforniaUSA
| | - R. Ted Abresch
- Department of Physical Medicine and RehabilitationUniversity of California Davis School of MedicineSacramentoCaliforniaUSA
| | - Francisco Villarreal
- Division of Cardiology, Department of Internal MedicineUniversity of California at San DiegoSan DiegoCaliforniaUSA
| | - Guillermo Ceballos
- Escuela Superior de Medicina, Seccion de Posgrado e Investigacion, del Instituto Politécnico NacionalMexico CityMexico
| | - Guy Perkins
- Division of Cardiology, Department of Internal MedicineUniversity of California at San DiegoSan DiegoCaliforniaUSA
| | - Sundeep Dugar
- Epirium Bio, Inc (formerly Cardero Therapeutics, Inc)San DiegoCaliforniaUSA
| | - George Schreiner
- Epirium Bio, Inc (formerly Cardero Therapeutics, Inc)San DiegoCaliforniaUSA
| | - Erik K. Henricson
- Department of Physical Medicine and RehabilitationUniversity of California Davis School of MedicineSacramentoCaliforniaUSA
| |
Collapse
|
16
|
Jahnke VE, Peterson JM, Van Der Meulen JH, Boehler J, Uaesoontrachoon K, Johnston HK, Defour A, Phadke A, Yu Q, Jaiswal JK, Nagaraju K. Mitochondrial dysfunction and consequences in calpain-3-deficient muscle. Skelet Muscle 2020; 10:37. [PMID: 33308300 PMCID: PMC7730798 DOI: 10.1186/s13395-020-00254-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Accepted: 11/16/2020] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Nonsense or loss-of-function mutations in the non-lysosomal cysteine protease calpain-3 result in limb-girdle muscular dystrophy type 2A (LGMD2A). While calpain-3 is implicated in muscle cell differentiation, sarcomere formation, and muscle cytoskeletal remodeling, the physiological basis for LGMD2A has remained elusive. METHODS Cell growth, gene expression profiling, and mitochondrial content and function were analyzed using muscle and muscle cell cultures established from healthy and calpain-3-deficient mice. Calpain-3-deficient mice were also treated with PPAR-delta agonist (GW501516) to assess mitochondrial function and membrane repair. The unpaired t test was used to assess the significance of the differences observed between the two groups or treatments. ANOVAs were used to assess significance over time. RESULTS We find that calpain-3 deficiency causes mitochondrial dysfunction in the muscles and myoblasts. Calpain-3-deficient myoblasts showed increased proliferation, and their gene expression profile showed aberrant mitochondrial biogenesis. Myotube gene expression analysis further revealed altered lipid metabolism in calpain-3-deficient muscle. Mitochondrial defects were validated in vitro and in vivo. We used GW501516 to improve mitochondrial biogenesis in vivo in 7-month-old calpain-3-deficient mice. This treatment improved satellite cell activity as indicated by increased MyoD and Pax7 mRNA expression. It also decreased muscle fatigability and reduced serum creatine kinase levels. The decreased mitochondrial function also impaired sarcolemmal repair in the calpain-3-deficient skeletal muscle. Improving mitochondrial activity by acute pyruvate treatment improved sarcolemmal repair. CONCLUSION Our results provide evidence that calpain-3 deficiency in the skeletal muscle is associated with poor mitochondrial biogenesis and function resulting in poor sarcolemmal repair. Addressing this deficit by drugs that improve mitochondrial activity offers new therapeutic avenues for LGMD2A.
Collapse
Affiliation(s)
- Vanessa E Jahnke
- Center for Genetic Medicine Research, Children's National Research Institute, Children's National Hospital, Washington, D.C., USA
| | - Jennifer M Peterson
- School of Exercise and Rehabilitation Sciences, The University of Toledo, Toledo, OH, USA
| | - Jack H Van Der Meulen
- Center for Genetic Medicine Research, Children's National Research Institute, Children's National Hospital, Washington, D.C., USA
| | - Jessica Boehler
- Center for Genetic Medicine Research, Children's National Research Institute, Children's National Hospital, Washington, D.C., USA
| | - Kitipong Uaesoontrachoon
- Center for Genetic Medicine Research, Children's National Research Institute, Children's National Hospital, Washington, D.C., USA
| | - Helen K Johnston
- Center for Genetic Medicine Research, Children's National Research Institute, Children's National Hospital, Washington, D.C., USA
- Department of Genomics and Precision Medicine, George Washington University School of Medicine and Health Sciences, Washington, D.C., USA
| | - Aurelia Defour
- Center for Genetic Medicine Research, Children's National Research Institute, Children's National Hospital, Washington, D.C., USA
| | - Aditi Phadke
- Center for Genetic Medicine Research, Children's National Research Institute, Children's National Hospital, Washington, D.C., USA
| | - Qing Yu
- Center for Genetic Medicine Research, Children's National Research Institute, Children's National Hospital, Washington, D.C., USA
| | - Jyoti K Jaiswal
- Center for Genetic Medicine Research, Children's National Research Institute, Children's National Hospital, Washington, D.C., USA
- Department of Genomics and Precision Medicine, George Washington University School of Medicine and Health Sciences, Washington, D.C., USA
| | - Kanneboyina Nagaraju
- Center for Genetic Medicine Research, Children's National Research Institute, Children's National Hospital, Washington, D.C., USA.
- Department of Genomics and Precision Medicine, George Washington University School of Medicine and Health Sciences, Washington, D.C., USA.
- School of Pharmacy and Pharmaceutical Sciences, SUNY Binghamton University, PO Box 6000, Binghamton, NY, 13902, USA.
| |
Collapse
|
17
|
Bittel DC, Chandra G, Tirunagri LMS, Deora AB, Medikayala S, Scheffer L, Defour A, Jaiswal JK. Annexin A2 Mediates Dysferlin Accumulation and Muscle Cell Membrane Repair. Cells 2020; 9:cells9091919. [PMID: 32824910 PMCID: PMC7565960 DOI: 10.3390/cells9091919] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 08/03/2020] [Accepted: 08/11/2020] [Indexed: 01/08/2023] Open
Abstract
Muscle cell plasma membrane is frequently damaged by mechanical activity, and its repair requires the membrane protein dysferlin. We previously identified that, similar to dysferlin deficit, lack of annexin A2 (AnxA2) also impairs repair of skeletal myofibers. Here, we have studied the mechanism of AnxA2-mediated muscle cell membrane repair in cultured muscle cells. We find that injury-triggered increase in cytosolic calcium causes AnxA2 to bind dysferlin and accumulate on dysferlin-containing vesicles as well as with dysferlin at the site of membrane injury. AnxA2 accumulates on the injured plasma membrane in cholesterol-rich lipid microdomains and requires Src kinase activity and the presence of cholesterol. Lack of AnxA2 and its failure to translocate to the plasma membrane, both prevent calcium-triggered dysferlin translocation to the plasma membrane and compromise repair of the injured plasma membrane. Our studies identify that Anx2 senses calcium increase and injury-triggered change in plasma membrane cholesterol to facilitate dysferlin delivery and repair of the injured plasma membrane.
Collapse
Affiliation(s)
- Daniel C. Bittel
- Center for Genetic Medicine Research, 111 Michigan Av NW, Children’s National Hospital, Washington, DC 20010, USA; (D.C.B.); (G.C.); (S.M.); (L.S.); (A.D.)
| | - Goutam Chandra
- Center for Genetic Medicine Research, 111 Michigan Av NW, Children’s National Hospital, Washington, DC 20010, USA; (D.C.B.); (G.C.); (S.M.); (L.S.); (A.D.)
| | - Laxmi M. S. Tirunagri
- Department of Cellular Biophysics, The Rockefeller University, New York, NY 10065, USA;
| | - Arun B. Deora
- Department of Cell & Developmental Biology, Weill Cornell Medical College, New York, NY 10065, USA;
| | - Sushma Medikayala
- Center for Genetic Medicine Research, 111 Michigan Av NW, Children’s National Hospital, Washington, DC 20010, USA; (D.C.B.); (G.C.); (S.M.); (L.S.); (A.D.)
| | - Luana Scheffer
- Center for Genetic Medicine Research, 111 Michigan Av NW, Children’s National Hospital, Washington, DC 20010, USA; (D.C.B.); (G.C.); (S.M.); (L.S.); (A.D.)
| | - Aurelia Defour
- Center for Genetic Medicine Research, 111 Michigan Av NW, Children’s National Hospital, Washington, DC 20010, USA; (D.C.B.); (G.C.); (S.M.); (L.S.); (A.D.)
| | - Jyoti K. Jaiswal
- Center for Genetic Medicine Research, 111 Michigan Av NW, Children’s National Hospital, Washington, DC 20010, USA; (D.C.B.); (G.C.); (S.M.); (L.S.); (A.D.)
- Department of Genomics and Precision medicine, George Washington University School of Medicine and Health Sciences, Washington, DC 20010, USA
- Correspondence: ; Tel.: +1-(202)476-6456; Fax: +1-(202)476-6014
| |
Collapse
|
18
|
Bittel AJ, Sreetama SC, Bittel DC, Horn A, Novak JS, Yokota T, Zhang A, Maruyama R, Rowel Q. Lim K, Jaiswal JK, Chen YW. Membrane Repair Deficit in Facioscapulohumeral Muscular Dystrophy. Int J Mol Sci 2020; 21:E5575. [PMID: 32759720 PMCID: PMC7432481 DOI: 10.3390/ijms21155575] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Revised: 07/28/2020] [Accepted: 07/30/2020] [Indexed: 12/14/2022] Open
Abstract
Deficits in plasma membrane repair have been identified in dysferlinopathy and Duchenne Muscular Dystrophy, and contribute to progressive myopathy. Although Facioscapulohumeral Muscular Dystrophy (FSHD) shares clinicopathological features with these muscular dystrophies, it is unknown if FSHD is characterized by plasma membrane repair deficits. Therefore, we exposed immortalized human FSHD myoblasts, immortalized myoblasts from unaffected siblings, and myofibers from a murine model of FSHD (FLExDUX4) to focal, pulsed laser ablation of the sarcolemma. Repair kinetics and success were determined from the accumulation of intracellular FM1-43 dye post-injury. We subsequently treated FSHD myoblasts with a DUX4-targeting antisense oligonucleotide (AON) to reduce DUX4 expression, and with the antioxidant Trolox to determine the role of DUX4 expression and oxidative stress in membrane repair. Compared to unaffected myoblasts, FSHD myoblasts demonstrate poor repair and a greater percentage of cells that failed to repair, which was mitigated by AON and Trolox treatments. Similar repair deficits were identified in FLExDUX4 myofibers. This is the first study to identify plasma membrane repair deficits in myoblasts from individuals with FSHD, and in myofibers from a murine model of FSHD. Our results suggest that DUX4 expression and oxidative stress may be important targets for future membrane-repair therapies.
Collapse
Affiliation(s)
- Adam J. Bittel
- Research Center for Genetic Medicine, Children’s National Hospital, 111 Michigan Ave NW, Washington, DC 20010, USA; (A.J.B.); (S.C.S.); (D.C.B.); (A.H.); (J.S.N.); (A.Z.)
| | - Sen Chandra Sreetama
- Research Center for Genetic Medicine, Children’s National Hospital, 111 Michigan Ave NW, Washington, DC 20010, USA; (A.J.B.); (S.C.S.); (D.C.B.); (A.H.); (J.S.N.); (A.Z.)
| | - Daniel C. Bittel
- Research Center for Genetic Medicine, Children’s National Hospital, 111 Michigan Ave NW, Washington, DC 20010, USA; (A.J.B.); (S.C.S.); (D.C.B.); (A.H.); (J.S.N.); (A.Z.)
| | - Adam Horn
- Research Center for Genetic Medicine, Children’s National Hospital, 111 Michigan Ave NW, Washington, DC 20010, USA; (A.J.B.); (S.C.S.); (D.C.B.); (A.H.); (J.S.N.); (A.Z.)
| | - James S. Novak
- Research Center for Genetic Medicine, Children’s National Hospital, 111 Michigan Ave NW, Washington, DC 20010, USA; (A.J.B.); (S.C.S.); (D.C.B.); (A.H.); (J.S.N.); (A.Z.)
- Department of Genomics and Precision Medicine, The George Washington University School of Medicine and Health Science, 111 Michigan Ave NW, Washington, DC 20010, USA
| | - Toshifumi Yokota
- Department of Medical Genetics, University of Alberta, 116 St. & 85 Ave., Edmonton, AB T6G 2R3, Canada; (T.Y.); (R.M.); (K.R.Q.L.)
| | - Aiping Zhang
- Research Center for Genetic Medicine, Children’s National Hospital, 111 Michigan Ave NW, Washington, DC 20010, USA; (A.J.B.); (S.C.S.); (D.C.B.); (A.H.); (J.S.N.); (A.Z.)
| | - Rika Maruyama
- Department of Medical Genetics, University of Alberta, 116 St. & 85 Ave., Edmonton, AB T6G 2R3, Canada; (T.Y.); (R.M.); (K.R.Q.L.)
| | - Kenji Rowel Q. Lim
- Department of Medical Genetics, University of Alberta, 116 St. & 85 Ave., Edmonton, AB T6G 2R3, Canada; (T.Y.); (R.M.); (K.R.Q.L.)
| | - Jyoti K. Jaiswal
- Research Center for Genetic Medicine, Children’s National Hospital, 111 Michigan Ave NW, Washington, DC 20010, USA; (A.J.B.); (S.C.S.); (D.C.B.); (A.H.); (J.S.N.); (A.Z.)
- Department of Integrative Systems Biology, Institute for Biomedical Sciences, The George Washington University, 2121 I St. NW, Washington, DC 20052, USA
| | - Yi-Wen Chen
- Research Center for Genetic Medicine, Children’s National Hospital, 111 Michigan Ave NW, Washington, DC 20010, USA; (A.J.B.); (S.C.S.); (D.C.B.); (A.H.); (J.S.N.); (A.Z.)
- Department of Integrative Systems Biology, Institute for Biomedical Sciences, The George Washington University, 2121 I St. NW, Washington, DC 20052, USA
| |
Collapse
|
19
|
Boehler JF, Horn A, Novak JS, Li N, Ghimbovschi S, Lundberg IE, Alexanderson H, Alemo Munters L, Jaiswal JK, Nagaraju K. Mitochondrial dysfunction and role of harakiri in the pathogenesis of myositis. J Pathol 2019; 249:215-226. [PMID: 31135059 DOI: 10.1002/path.5309] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 04/16/2019] [Accepted: 05/23/2019] [Indexed: 12/17/2022]
Abstract
The etiology of myositis is unknown. Although attempts to identify viruses in myositis skeletal muscle have failed, several studies have identified the presence of a viral signature in myositis patients. Here we postulate that in individuals with susceptible genetic backgrounds, viral infection alters the epigenome to activate the pathological pathways leading to disease onset. To identify epigenetic changes, methylation profiling of Coxsackie B infected human myotubes and muscle biopsies from polymyositis (PM) and dermatomyositis (DM) patients were compared to changes in global transcript expression induced by in vitro Coxsackie B infection. Gene and protein expression analysis and live cell imaging were performed to examine the mechanisms. Analysis of methylation and gene expression changes identified that a mitochondria-localized activator of apoptosis - harakiri (HRK) - is upregulated in myositis skeletal muscle cells. Muscle cells with higher HRK expression have reduced mitochondrial potential and poor ability to repair from injury as compared to controls. In cells from myositis patient toll-like receptor 7 (TLR7) activates and sustains high HRK expression. Forced over expression of HRK in healthy muscle cells is sufficient to compromise their membrane repair ability. Endurance exercise that is associated with improved muscle and mitochondrial function in PM and DM patients decreased TLR7 and HRK expression identifying these as therapeutic targets. Increased HRK and TLR7 expression causes mitochondrial damage leading to poor myofiber repair, myofiber death and muscle weakness in myositis patients and exercise induced reduction of HRK and TLR7 expression in patients is associated with disease amelioration. © 2019 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Jessica F Boehler
- Center for Genetic Medicine Research, Children's National Health System, Washington, DC, USA.,Institute for Biomedical Sciences, George Washington University, Washington, DC, USA
| | - Adam Horn
- Center for Genetic Medicine Research, Children's National Health System, Washington, DC, USA.,Institute for Biomedical Sciences, George Washington University, Washington, DC, USA
| | - James S Novak
- Center for Genetic Medicine Research, Children's National Health System, Washington, DC, USA.,Department of Genomics and Precision Medicine, George Washington University School of Medicine, Washington, DC, USA
| | - Ning Li
- Department of Pharmaceutical Sciences School of Pharmacy and Pharmaceutical Sciences, Binghamton University, Binghamton, NY, USA
| | - Svetlana Ghimbovschi
- Center for Genetic Medicine Research, Children's National Health System, Washington, DC, USA
| | - Ingrid E Lundberg
- Division of Rheumatology, Department of Medicine, Karolinska Institutet, Solna, Sweden.,Division of Rheumatology, Karolinska Universitetssjukhuset, Stockholm, Sweden
| | - Helene Alexanderson
- Function Area Occupational Therapy and Physical Therapy, Karolinska University Hospital, Stockholm, Sweden.,Division of Physical Therapy, Department of NVS, Karolinska Institutet, Stockholm, Sweden.,Division of Rheumatology, Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Li Alemo Munters
- Function Area Occupational Therapy and Physical Therapy, Karolinska University Hospital and Swedish Rheumatism Association, Stockholm, Sweden
| | - Jyoti K Jaiswal
- Center for Genetic Medicine Research, Children's National Health System, Washington, DC, USA.,Department of Genomics and Precision Medicine, George Washington University School of Medicine, Washington, DC, USA
| | - Kanneboyina Nagaraju
- Center for Genetic Medicine Research, Children's National Health System, Washington, DC, USA.,Department of Pharmaceutical Sciences School of Pharmacy and Pharmaceutical Sciences, Binghamton University, Binghamton, NY, USA
| |
Collapse
|
20
|
Sønder SL, Boye TL, Tölle R, Dengjel J, Maeda K, Jäättelä M, Simonsen AC, Jaiswal JK, Nylandsted J. Annexin A7 is required for ESCRT III-mediated plasma membrane repair. Sci Rep 2019; 9:6726. [PMID: 31040365 PMCID: PMC6491720 DOI: 10.1038/s41598-019-43143-4] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Accepted: 04/15/2019] [Indexed: 12/21/2022] Open
Abstract
The plasma membrane of eukaryotic cells forms the essential barrier to the extracellular environment, and thus plasma membrane disruptions pose a fatal threat to cells. Here, using invasive breast cancer cells we show that the Ca2+ - and phospholipid-binding protein annexin A7 is part of the plasma membrane repair response by enabling assembly of the endosomal sorting complex required for transport (ESCRT) III. Following injury to the plasma membrane and Ca2+ flux into the cytoplasm, annexin A7 forms a complex with apoptosis linked gene-2 (ALG-2) to facilitate proper recruitment and binding of ALG-2 and ALG-2-interacting protein X (ALIX) to the damaged membrane. ALG-2 and ALIX assemble the ESCRT III complex, which helps excise and shed the damaged portion of the plasma membrane during wound healing. Our results reveal a novel function of annexin A7 – enabling plasma membrane repair by regulating ESCRT III-mediated shedding of injured plasma membrane.
Collapse
Affiliation(s)
- Stine Lauritzen Sønder
- Unit for Cell Death and Metabolism, Center for Autophagy, Recycling and Disease, Danish Cancer Society Research Center, Strandboulevarden 49, DK-2100, Copenhagen, Denmark
| | - Theresa Louise Boye
- Unit for Cell Death and Metabolism, Center for Autophagy, Recycling and Disease, Danish Cancer Society Research Center, Strandboulevarden 49, DK-2100, Copenhagen, Denmark
| | - Regine Tölle
- Department of Dermatology, Medical Center, University of Freiburg, 79104, Freiburg, Germany.,Department of Biology, University of Fribourg Chemin du Musée 10, 1700, Fribourg, Switzerland
| | - Jörn Dengjel
- Department of Dermatology, Medical Center, University of Freiburg, 79104, Freiburg, Germany.,Department of Biology, University of Fribourg Chemin du Musée 10, 1700, Fribourg, Switzerland
| | - Kenji Maeda
- Unit for Cell Death and Metabolism, Center for Autophagy, Recycling and Disease, Danish Cancer Society Research Center, Strandboulevarden 49, DK-2100, Copenhagen, Denmark
| | - Marja Jäättelä
- Unit for Cell Death and Metabolism, Center for Autophagy, Recycling and Disease, Danish Cancer Society Research Center, Strandboulevarden 49, DK-2100, Copenhagen, Denmark.,Department of Cellular and Molecular Medicine, Faculty of Health Sciences, University of Copenhagen, DK-2200, Copenhagen N, Denmark
| | - Adam Cohen Simonsen
- Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, Campusvej 55, DK-5230, Odense M, Denmark
| | - Jyoti K Jaiswal
- Children's National Health System, Center for Genetic Medicine Research, George Washington University School of Medicine and Health Sciences, 111 Michigan Avenue, NW, Washington, DC, 20010-2970, USA.,Department of Genomics and Precision Medicine, George Washington University School of Medicine and Health Sciences, 111 Michigan Avenue, NW, Washington, DC, 20010-2970, USA
| | - Jesper Nylandsted
- Unit for Cell Death and Metabolism, Center for Autophagy, Recycling and Disease, Danish Cancer Society Research Center, Strandboulevarden 49, DK-2100, Copenhagen, Denmark. .,Department of Cellular and Molecular Medicine, Faculty of Health Sciences, University of Copenhagen, DK-2200, Copenhagen N, Denmark.
| |
Collapse
|
21
|
Chao T, Burmeister DM, Corona BT, Greising SM. Oxidative pathophysiology following volumetric muscle loss injury in a porcine model. J Appl Physiol (1985) 2019; 126:1541-1549. [PMID: 30920884 DOI: 10.1152/japplphysiol.00026.2019] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Volumetric muscle loss (VML) occurs after severe orthopedic trauma and results in loss of muscle fibers and function that can leave patients permanently disabled. Although animals models of VML are useful to test possible therapeutic strategies, the pathophysiological characteristics of remaining skeletal muscle and changes in metabolism are not thoroughly understood. Herein, alterations of neuromuscular function, muscle fiber morphology, myosin heavy chain expression, and myofiber mitochondrial respiration were evaluated in an adult Yorkshire swine VML injury model. VML injured animals showed reduced peak isometric strength (P < 0.05) and a shift toward smaller muscle fibers independent of fiber type (P < 0.001). The muscle remaining after VML had a greater distribution of type I fibers and lower distribution of type II fibers (P < 0.001). Skeletal muscle mitochondrial state 2 and state 3, reflecting complex I respiration, increased after injury (P < 0.05) with a consistent trend to display higher oxygen flux per milligram of tissue. However, this was largely driven by increased mitochondrial content after VML which was associated with higher mitochondrial fission (FIS-1 protein levels). This study demonstrates an underlying perturbation of oxidative metabolism within the remaining musculature following surgical creation of an isolated, sterile VML injury in a porcine model that may be influential to the development of insidious pathophysiology and regenerative and rehabilitative therapies. NEW & NOTEWORTHY The natural injury sequela of volumetric muscle loss (VML) and associated pathophysiology of the remaining muscle is still incompletely understood. Herein we demonstrate a chronic muscle function deficit, with an increase in type I muscle fibers and parallel increase in oxidative capacity of remaining skeletal muscle. It is possible that the alteration in oxidative capacity after VML could largely be due to heightened mitochondrial activity and an increase in mitochondrial abundance.
Collapse
Affiliation(s)
- Tony Chao
- Damage Control Resuscitation Task Area, United States Army Institute of Surgical Research, Fort Sam Houston, Texas
| | - David M Burmeister
- Damage Control Resuscitation Task Area, United States Army Institute of Surgical Research, Fort Sam Houston, Texas
| | - Benjamin T Corona
- Extremity Trauma and Regenerative Medicine Task Area, United States Army Institute of Surgical Research, Fort Sam Houston, Texas.,School of Medicine, Wake Forest University , Winston-Salem, North Carolina
| | - Sarah M Greising
- Extremity Trauma and Regenerative Medicine Task Area, United States Army Institute of Surgical Research, Fort Sam Houston, Texas.,School of Kinesiology, University of Minnesota , Minneapolis, Minnesota
| |
Collapse
|
22
|
Gaglianone RB, Santos AT, Bloise FF, Ortiga-Carvalho TM, Costa ML, Quirico-Santos T, da Silva WS, Mermelstein C. Reduced mitochondrial respiration and increased calcium deposits in the EDL muscle, but not in soleus, from 12-week-old dystrophic mdx mice. Sci Rep 2019; 9:1986. [PMID: 30760802 PMCID: PMC6374364 DOI: 10.1038/s41598-019-38609-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Accepted: 01/04/2019] [Indexed: 12/12/2022] Open
Abstract
Mitochondria play an important role in providing ATP for muscle contraction. Muscle physiology is compromised in Duchenne muscular dystrophy (DMD) and several studies have shown the involvement of bioenergetics. In this work we investigated the mitochondrial physiology in fibers from fast-twitch muscle (EDL) and slow-twitch muscle (soleus) in the mdx mouse model for DMD and in control C57BL/10J mice. In our study, multiple mitochondrial respiratory parameters were investigated in permeabilized muscle fibers from 12-week-old animals, a critical age where muscle regeneration is observed in the mdx mouse. Using substrates of complex I and complex II from the electron transport chain, ADP and mitochondrial inhibitors, we found in the mdx EDL, but not in the mdx soleus, a reduction in coupled respiration suggesting that ATP synthesis is affected. In addition, the oxygen consumption after addition of complex II substrate is reduced in mdx EDL; the maximal consumption rate (measured in the presence of uncoupler) also seems to be reduced. Mitochondria are involved in calcium regulation and we observed, using alizarin stain, calcium deposits in mdx muscles but not in control muscles. Interestingly, more calcium deposits were found in mdx EDL than in mdx soleus. These data provide evidence that in 12-week-old mdx mice, calcium is accumulated and mitochondrial function is disturbed in the fast-twitch muscle EDL, but not in the slow-twitch muscle soleus.
Collapse
Affiliation(s)
- Rhayanna B Gaglianone
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Anderson Teixeira Santos
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Flavia Fonseca Bloise
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Tania Maria Ortiga-Carvalho
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Manoel Luis Costa
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | | | - Wagner Seixas da Silva
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Claudia Mermelstein
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil.
| |
Collapse
|
23
|
Thankam FG, Chandra IS, Kovilam AN, Diaz CG, Volberding BT, Dilisio MF, Radwan MM, Gross RM, Agrawal DK. Amplification of Mitochondrial Activity in the Healing Response Following Rotator Cuff Tendon Injury. Sci Rep 2018; 8:17027. [PMID: 30451947 PMCID: PMC6242817 DOI: 10.1038/s41598-018-35391-7] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Accepted: 10/31/2018] [Indexed: 02/07/2023] Open
Abstract
Mitochondrial function following rotator cuff tendon injury (RCI) influences the tendon healing. We examined the mitochondrial morphology and function under hypoxia in the shoulder tendon tissue from surgically-induced tenotomy-RCI rat model and cultured swine tenocytes. The tendon tissue was collected post-injury on 3-5 (Group-A), 10-12 (Group-B), and 22-24 (Group-C), days and the corresponding contralateral tendons were used as control for each group. There was higher protein expression of citrate synthase (P < 0.0001) [10.22 MFI (mean fluorescent intensity)] and complex-1 (P = 0.0008) (7.86 MFI) in Group-A and Group-B that decreased in Group-C [(P = 0.0201) (5.78 MFI and (P = 0.7915) (2.32 MFI), respectively] compared to control tendons. The ratio of BAX:Bcl2 (Bcl2 associated x protein:B cell lymphoma 2) in RCI tendons increased by 50.5% (Group-A) and 68.4% (Group-B) and decreased by 25.8% (Group-C) compared to normoxic controls. Hypoxia increased β-tubulin expression (P = 0067) and reduced PGC1-α (P = 0412) expression in the isolated swine tenocytes with no effect on the protein expression of Complex-1 (P = 7409) and citrate synthase (P = 0.3290). Also, the hypoxic tenocytes exhibited about 4-fold increase in mitochondrial superoxide (P < 0.0001), altered morphology and mitochondrial pore integrity, and increase in mitochondrial density compared to normoxic controls. These findings suggest the critical role of mitochondria in the RCI healing response.
Collapse
Affiliation(s)
- Finosh G Thankam
- Departments of Clinical & Translational Science and Orthopedic Surgery, Creighton University School of Medicine, Omaha, NE, 68178, USA
| | - Isaiah S Chandra
- Departments of Clinical & Translational Science and Orthopedic Surgery, Creighton University School of Medicine, Omaha, NE, 68178, USA
| | - Anuradha N Kovilam
- Departments of Clinical & Translational Science and Orthopedic Surgery, Creighton University School of Medicine, Omaha, NE, 68178, USA
| | - Connor G Diaz
- Departments of Clinical & Translational Science and Orthopedic Surgery, Creighton University School of Medicine, Omaha, NE, 68178, USA
| | - Benjamin T Volberding
- Departments of Clinical & Translational Science and Orthopedic Surgery, Creighton University School of Medicine, Omaha, NE, 68178, USA
| | - Matthew F Dilisio
- Departments of Clinical & Translational Science and Orthopedic Surgery, Creighton University School of Medicine, Omaha, NE, 68178, USA
| | - Mohamed M Radwan
- Departments of Clinical & Translational Science and Orthopedic Surgery, Creighton University School of Medicine, Omaha, NE, 68178, USA
| | - R Michael Gross
- Departments of Clinical & Translational Science and Orthopedic Surgery, Creighton University School of Medicine, Omaha, NE, 68178, USA
| | - Devendra K Agrawal
- Departments of Clinical & Translational Science and Orthopedic Surgery, Creighton University School of Medicine, Omaha, NE, 68178, USA.
| |
Collapse
|
24
|
Dekraker C, Boucher E, Mandato CA. Regulation and Assembly of Actomyosin Contractile Rings in Cytokinesis and Cell Repair. Anat Rec (Hoboken) 2018; 301:2051-2066. [PMID: 30312008 DOI: 10.1002/ar.23962] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Revised: 08/24/2018] [Accepted: 08/27/2018] [Indexed: 01/17/2023]
Abstract
Cytokinesis and single-cell wound repair both involve contractile assemblies of filamentous actin (F-actin) and myosin II organized into characteristic ring-like arrays. The assembly of these actomyosin contractile rings (CRs) is specified spatially and temporally by small Rho GTPases, which trigger local actin polymerization and myosin II contractility via a variety of downstream effectors. We now have a much clearer view of the Rho GTPase signaling cascade that leads to the formation of CRs, but some factors involved in CR positioning, assembly, and function remain poorly understood. Recent studies show that this regulation is multifactorial and goes beyond the long-established Ca2+ -dependent processes. There is substantial evidence that the Ca2+ -independent changes in cell shape, tension, and plasma membrane composition that characterize cytokinesis and single-cell wound repair also regulate CR formation. Elucidating the regulation and mechanistic properties of CRs is important to our understanding of basic cell biology and holds potential for therapeutic applications in human disease. In this review, we present a primer on the factors influencing and regulating CR positioning, assembly, and contraction as they occur in a variety of cytokinetic and single-cell wound repair models. Anat Rec, 301:2051-2066, 2018. © 2018 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Corina Dekraker
- Department of Anatomy and Cell Biology, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
| | - Eric Boucher
- Department of Anatomy and Cell Biology, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
| | - Craig A Mandato
- Department of Anatomy and Cell Biology, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
25
|
Ryan TE, Yamaguchi DJ, Schmidt CA, Zeczycki TN, Shaikh SR, Brophy P, Green TD, Tarpey MD, Karnekar R, Goldberg EJ, Sparagna GC, Torres MJ, Annex BH, Neufer PD, Spangenburg EE, McClung JM. Extensive skeletal muscle cell mitochondriopathy distinguishes critical limb ischemia patients from claudicants. JCI Insight 2018; 3:123235. [PMID: 30385731 DOI: 10.1172/jci.insight.123235] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Accepted: 10/02/2018] [Indexed: 12/31/2022] Open
Abstract
The most severe manifestation of peripheral arterial disease (PAD) is critical limb ischemia (CLI). CLI patients suffer high rates of amputation and mortality; accordingly, there remains a clear need both to better understand CLI and to develop more effective treatments. Gastrocnemius muscle was obtained from 32 older (51-84 years) non-PAD controls, 27 claudicating PAD patients (ankle-brachial index [ABI] 0.65 ± 0.21 SD), and 19 CLI patients (ABI 0.35 ± 0.30 SD) for whole transcriptome sequencing and comprehensive mitochondrial phenotyping. Comparable permeabilized myofiber mitochondrial function was paralleled by both similar mitochondrial content and related mRNA expression profiles in non-PAD control and claudicating patient tissues. Tissues from CLI patients, despite being histologically intact and harboring equivalent mitochondrial content, presented a unique bioenergetic signature. This signature was defined by deficits in permeabilized myofiber mitochondrial function and a unique pattern of both nuclear and mitochondrial encoded gene suppression. Moreover, isolated muscle progenitor cells retained both mitochondrial functional deficits and gene suppression observed in the tissue. These findings indicate that muscle tissues from claudicating patients and non-PAD controls were similar in both their bioenergetics profile and mitochondrial phenotypes. In contrast, CLI patient limb skeletal muscles harbor a unique skeletal muscle mitochondriopathy that represents a potentially novel therapeutic site for intervention.
Collapse
Affiliation(s)
- Terence E Ryan
- Department of Physiology.,East Carolina Diabetes and Obesity Institute
| | | | - Cameron A Schmidt
- Department of Physiology.,East Carolina Diabetes and Obesity Institute
| | - Tonya N Zeczycki
- East Carolina Diabetes and Obesity Institute.,Department of Biochemistry and Molecular Biology, Brody School of Medicine, East Carolina University, Greenville, North Carolina, USA
| | - Saame Raza Shaikh
- Department of Nutrition, Gillings School of Global Public Health, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | | | - Thomas D Green
- Department of Physiology.,East Carolina Diabetes and Obesity Institute
| | - Michael D Tarpey
- Department of Physiology.,East Carolina Diabetes and Obesity Institute
| | - Reema Karnekar
- Department of Physiology.,East Carolina Diabetes and Obesity Institute
| | - Emma J Goldberg
- Department of Physiology.,East Carolina Diabetes and Obesity Institute
| | | | | | - Brian H Annex
- Division of Cardiovascular Medicine, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - P Darrell Neufer
- Department of Physiology.,East Carolina Diabetes and Obesity Institute
| | | | - Joseph M McClung
- Department of Physiology.,East Carolina Diabetes and Obesity Institute.,Department of Cardiovascular Sciences
| |
Collapse
|
26
|
Horn A, Jaiswal JK. Cellular mechanisms and signals that coordinate plasma membrane repair. Cell Mol Life Sci 2018; 75:3751-3770. [PMID: 30051163 PMCID: PMC6541445 DOI: 10.1007/s00018-018-2888-7] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2018] [Revised: 07/13/2018] [Accepted: 07/23/2018] [Indexed: 02/08/2023]
Abstract
Plasma membrane forms the barrier between the cytoplasm and the environment. Cells constantly and selectively transport molecules across their plasma membrane without disrupting it. Any disruption in the plasma membrane compromises its selective permeability and is lethal, if not rapidly repaired. There is a growing understanding of the organelles, proteins, lipids, and small molecules that help cells signal and efficiently coordinate plasma membrane repair. This review aims to summarize how these subcellular responses are coordinated and how cellular signals generated due to plasma membrane injury interact with each other to spatially and temporally coordinate repair. With the involvement of calcium and redox signaling in single cell and tissue repair, we will discuss how these and other related signals extend from single cell repair to tissue level repair. These signals link repair processes that are activated immediately after plasma membrane injury with longer term processes regulating repair and regeneration of the damaged tissue. We propose that investigating cell and tissue repair as part of a continuum of wound repair mechanisms would be of value in treating degenerative diseases.
Collapse
Affiliation(s)
- Adam Horn
- Center for Genetic Medicine Research, Children's National Health System, 111 Michigan Avenue, NW, Washington, DC, 20010-2970, USA
- Department of Genomics and Precision Medicine, George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | - Jyoti K Jaiswal
- Center for Genetic Medicine Research, Children's National Health System, 111 Michigan Avenue, NW, Washington, DC, 20010-2970, USA.
- Department of Genomics and Precision Medicine, George Washington University School of Medicine and Health Sciences, Washington, DC, USA.
| |
Collapse
|
27
|
Horn A, Van der Meulen JH, Defour A, Hogarth M, Sreetama SC, Reed A, Scheffer L, Chandel NS, Jaiswal JK. Mitochondrial redox signaling enables repair of injured skeletal muscle cells. Sci Signal 2017; 10:eaaj1978. [PMID: 28874604 PMCID: PMC5949579 DOI: 10.1126/scisignal.aaj1978] [Citation(s) in RCA: 114] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Strain and physical trauma to mechanically active cells, such as skeletal muscle myofibers, injures their plasma membranes, and mitochondrial function is required for their repair. We found that mitochondrial function was also needed for plasma membrane repair in myoblasts as well as nonmuscle cells, which depended on mitochondrial uptake of calcium through the mitochondrial calcium uniporter (MCU). Calcium uptake transiently increased the mitochondrial production of reactive oxygen species (ROS), which locally activated the guanosine triphosphatase (GTPase) RhoA, triggering F-actin accumulation at the site of injury and facilitating membrane repair. Blocking mitochondrial calcium uptake or ROS production prevented injury-triggered RhoA activation, actin polymerization, and plasma membrane repair. This repair mechanism was shared between myoblasts, nonmuscle cells, and mature skeletal myofibers. Quenching mitochondrial ROS in myofibers during eccentric exercise ex vivo caused increased damage to myofibers, resulting in a greater loss of muscle force. These results suggest a physiological role for mitochondria in plasma membrane repair in injured cells, a role that highlights a beneficial effect of ROS.
Collapse
Affiliation(s)
- Adam Horn
- Children's National Health System, Center for Genetic Medicine Research, 111 Michigan Avenue Northwest, Washington, DC 20010-2970, USA
- Department of Integrative Systems Biology, George Washington University School of Medicine and Health Sciences, Washington, DC 20010-2970, USA
| | - Jack H Van der Meulen
- Children's National Health System, Center for Genetic Medicine Research, 111 Michigan Avenue Northwest, Washington, DC 20010-2970, USA
| | - Aurelia Defour
- Children's National Health System, Center for Genetic Medicine Research, 111 Michigan Avenue Northwest, Washington, DC 20010-2970, USA
| | - Marshall Hogarth
- Children's National Health System, Center for Genetic Medicine Research, 111 Michigan Avenue Northwest, Washington, DC 20010-2970, USA
| | - Sen Chandra Sreetama
- Children's National Health System, Center for Genetic Medicine Research, 111 Michigan Avenue Northwest, Washington, DC 20010-2970, USA
| | - Aaron Reed
- Children's National Health System, Center for Genetic Medicine Research, 111 Michigan Avenue Northwest, Washington, DC 20010-2970, USA
| | - Luana Scheffer
- Children's National Health System, Center for Genetic Medicine Research, 111 Michigan Avenue Northwest, Washington, DC 20010-2970, USA
| | - Navdeep S Chandel
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Jyoti K Jaiswal
- Children's National Health System, Center for Genetic Medicine Research, 111 Michigan Avenue Northwest, Washington, DC 20010-2970, USA.
- Department of Integrative Systems Biology, George Washington University School of Medicine and Health Sciences, Washington, DC 20010-2970, USA
| |
Collapse
|
28
|
Growing functions of the ESCRT machinery in cell biology and viral replication. Biochem Soc Trans 2017; 45:613-634. [PMID: 28620025 DOI: 10.1042/bst20160479] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Revised: 02/17/2017] [Accepted: 02/21/2017] [Indexed: 01/31/2023]
Abstract
The vast expansion in recent years of the cellular processes promoted by the endosomal sorting complex required for transport (ESCRT) machinery has reinforced its identity as a modular system that uses multiple adaptors to recruit the core membrane remodelling activity at different intracellular sites and facilitate membrane scission. Functional connections to processes such as the aurora B-dependent abscission checkpoint also highlight the importance of the spatiotemporal regulation of the ESCRT machinery. Here, we summarise the role of ESCRTs in viral budding, and what we have learned about the ESCRT pathway from studying this process. These advances are discussed in the context of areas of cell biology that have been transformed by research in the ESCRT field, including cytokinetic abscission, nuclear envelope resealing and plasma membrane repair.
Collapse
|
29
|
Davenport NR, Bement WM. Cell repair: Revisiting the patch hypothesis. Commun Integr Biol 2016; 9:e1253643. [PMID: 28042380 PMCID: PMC5193046 DOI: 10.1080/19420889.2016.1253643] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Accepted: 10/22/2016] [Indexed: 11/23/2022] Open
Abstract
Plasma membrane damage elicits a complex and dynamic cellular response. A vital component of this response, membrane resealing, is thought to arise from fusion of intracellular membranous compartments to form a temporary, impermeant patch at the site of damage; however, this hypothesis has been difficult to confirm visually. By utilizing advanced microscopy technologies with high spatiotemporal resolution in wounded Xenopus laevis oocytes, we provide the first direct visualization of the membrane fusion events predicted by the patch hypothesis; we show the barrier formed by patching is capable of abating exchange of material across the plasma membrane within seconds. Profound changes also occur to the plasma membrane surrounding wounds; lipid remodeling is accompanied by membrane fusion events, both conventional (e.g., exocytosis) and novel (e.g., “explodosis”). Further, we reveal additional complexity in wound-induced subcellular patterning, supporting existing evidence that extensive interactions between lipid, protein, and ionic signaling pathways shape the cellular wound response.
Collapse
Affiliation(s)
- Nicholas R Davenport
- Laboratory of Cell & Molecular Biology University of Wisconsin-Madison , Madison, WI, USA
| | - William M Bement
- Laboratory of Cell & Molecular Biology University of Wisconsin-Madison, Madison, WI, USA; Department of Zoology; University of Wisconsin-Madison, Madison, WI, USA
| |
Collapse
|
30
|
Mitochondria mediate cell membrane repair and contribute to Duchenne muscular dystrophy. Cell Death Differ 2016; 24:330-342. [PMID: 27834955 PMCID: PMC5299714 DOI: 10.1038/cdd.2016.127] [Citation(s) in RCA: 106] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Revised: 09/02/2016] [Accepted: 09/28/2016] [Indexed: 12/31/2022] Open
Abstract
Dystrophin deficiency is the genetic basis for Duchenne muscular dystrophy (DMD), but the cellular basis of progressive myofiber death in DMD is not fully understood. Using two dystrophin-deficient mdx mouse models, we find that the mitochondrial dysfunction is among the earliest cellular deficits of mdx muscles. Mitochondria in dystrophic myofibers also respond poorly to sarcolemmal injury. These mitochondrial deficits reduce the ability of dystrophic muscle cell membranes to repair and are associated with a compensatory increase in dysferlin-mediated membrane repair proteins. Dysferlin deficit in mdx mice further compromises myofiber cell membrane repair and enhances the muscle pathology at an asymptomatic age for dysferlin-deficient mice. Restoring partial dystrophin expression by exon skipping improves mitochondrial function and offers potential to improve myofiber repair. These findings identify that mitochondrial deficit in muscular dystrophy compromises the repair of injured myofibers and show that this repair mechanism is distinct from and complimentary to the dysferlin-mediated repair of injured myofibers.
Collapse
|
31
|
Vila MC, Klimek MB, Novak JS, Rayavarapu S, Uaesoontrachoon K, Boehler JF, Fiorillo AA, Hogarth MW, Zhang A, Shaughnessy C, Gordish-Dressman H, Burki U, Straub V, Lu QL, Partridge TA, Brown KJ, Hathout Y, van den Anker J, Hoffman EP, Nagaraju K. Elusive sources of variability of dystrophin rescue by exon skipping. Skelet Muscle 2015; 5:44. [PMID: 26634117 PMCID: PMC4667482 DOI: 10.1186/s13395-015-0070-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Accepted: 11/24/2015] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Systemic delivery of anti-sense oligonucleotides to Duchenne muscular dystrophy (DMD) patients to induce de novo dystrophin protein expression in muscle (exon skipping) is a promising therapy. Treatment with Phosphorodiamidate morpholino oligomers (PMO) lead to shorter de novo dystrophin protein in both animal models and DMD boys who otherwise lack dystrophin; however, restoration of dystrophin has been observed to be highly variable. Understanding the factors causing highly variable induction of dystrophin expression in pre-clinical models would likely lead to more effective means of exon skipping in both pre-clinical studies and human clinical trials. METHODS In the present study, we investigated possible factors that might lead to the variable success of exon skipping using morpholino drugs in the mdx mouse model. We tested whether specific muscle groups or fiber types showed better success than others and also correlated residual PMO concentration in muscle with the amount of de novo dystrophin protein 1 month after a single high-dose morpholino injection (800 mg/kg). We compared the results from six muscle groups using three different methods of dystrophin quantification: immunostaining, immunoblotting, and mass spectrometry assays. RESULTS The triceps muscle showed the greatest degree of rescue (average 38±28 % by immunostaining). All three dystrophin detection methods were generally concordant for all muscles. We show that dystrophin rescue occurs in a sporadic patchy pattern with high geographic variability across muscle sections. We did not find a correlation between residual morpholino drug in muscle tissue and the degree of dystrophin expression. CONCLUSIONS While we found some evidence of muscle group enhancement and successful rescue, our data also suggest that other yet-undefined factors may underlie the observed variability in the success of exon skipping. Our study highlights the challenges associated with quantifying dystrophin in clinical trials where a single small muscle biopsy is taken from a DMD patient.
Collapse
Affiliation(s)
- Maria Candida Vila
- Research Center for Genetic Medicine, Children's National Health System, 111 Michigan Avenue N.W., Washington, DC, 20010 USA.,Institute of Biomedical Sciences, The George Washington University, Washington, DC, USA
| | - Margaret Benny Klimek
- Research Center for Genetic Medicine, Children's National Health System, 111 Michigan Avenue N.W., Washington, DC, 20010 USA
| | - James S Novak
- Research Center for Genetic Medicine, Children's National Health System, 111 Michigan Avenue N.W., Washington, DC, 20010 USA
| | - Sree Rayavarapu
- Research Center for Genetic Medicine, Children's National Health System, 111 Michigan Avenue N.W., Washington, DC, 20010 USA
| | - Kitipong Uaesoontrachoon
- Research Center for Genetic Medicine, Children's National Health System, 111 Michigan Avenue N.W., Washington, DC, 20010 USA
| | - Jessica F Boehler
- Research Center for Genetic Medicine, Children's National Health System, 111 Michigan Avenue N.W., Washington, DC, 20010 USA.,Institute of Biomedical Sciences, The George Washington University, Washington, DC, USA
| | - Alyson A Fiorillo
- Research Center for Genetic Medicine, Children's National Health System, 111 Michigan Avenue N.W., Washington, DC, 20010 USA
| | - Marshall W Hogarth
- Research Center for Genetic Medicine, Children's National Health System, 111 Michigan Avenue N.W., Washington, DC, 20010 USA
| | - Aiping Zhang
- Research Center for Genetic Medicine, Children's National Health System, 111 Michigan Avenue N.W., Washington, DC, 20010 USA
| | - Conner Shaughnessy
- Research Center for Genetic Medicine, Children's National Health System, 111 Michigan Avenue N.W., Washington, DC, 20010 USA
| | - Heather Gordish-Dressman
- Research Center for Genetic Medicine, Children's National Health System, 111 Michigan Avenue N.W., Washington, DC, 20010 USA.,Institute of Biomedical Sciences, The George Washington University, Washington, DC, USA
| | - Umar Burki
- The John Walton Muscular Dystrophy Research Centre, MRC Centre for Neuromuscular Diseases at Newcastle, Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne, UK
| | - Volker Straub
- The John Walton Muscular Dystrophy Research Centre, MRC Centre for Neuromuscular Diseases at Newcastle, Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne, UK
| | - Qi Long Lu
- McColl-Lockwood Laboratory for Muscular Dystrophy Research, Neuromuscular/ALS Center, Department of Neurology, Carolinas Medical Center, Charlotte, NC USA
| | - Terence A Partridge
- Research Center for Genetic Medicine, Children's National Health System, 111 Michigan Avenue N.W., Washington, DC, 20010 USA.,Institute of Biomedical Sciences, The George Washington University, Washington, DC, USA
| | - Kristy J Brown
- Research Center for Genetic Medicine, Children's National Health System, 111 Michigan Avenue N.W., Washington, DC, 20010 USA.,Institute of Biomedical Sciences, The George Washington University, Washington, DC, USA
| | - Yetrib Hathout
- Research Center for Genetic Medicine, Children's National Health System, 111 Michigan Avenue N.W., Washington, DC, 20010 USA.,Institute of Biomedical Sciences, The George Washington University, Washington, DC, USA
| | - John van den Anker
- Research Center for Genetic Medicine, Children's National Health System, 111 Michigan Avenue N.W., Washington, DC, 20010 USA.,Center for Translational Science, Children's National Health System, Washington, DC, USA
| | - Eric P Hoffman
- Research Center for Genetic Medicine, Children's National Health System, 111 Michigan Avenue N.W., Washington, DC, 20010 USA.,Institute of Biomedical Sciences, The George Washington University, Washington, DC, USA
| | - Kanneboyina Nagaraju
- Research Center for Genetic Medicine, Children's National Health System, 111 Michigan Avenue N.W., Washington, DC, 20010 USA.,Institute of Biomedical Sciences, The George Washington University, Washington, DC, USA
| |
Collapse
|
32
|
Jaiswal JK, Nylandsted J. S100 and annexin proteins identify cell membrane damage as the Achilles heel of metastatic cancer cells. Cell Cycle 2015; 14:502-9. [PMID: 25565331 DOI: 10.1080/15384101.2014.995495] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Mechanical activity of cells and the stress imposed on them by extracellular environment is a constant source of injury to the plasma membrane (PM). In invasive tumor cells, increased motility together with the harsh environment of the tumor stroma further increases the risk of PM injury. The impact of these stresses on tumor cell plasma membrane and mechanism by which tumor cells repair the PM damage are poorly understood. Ca(2+) entry through the injured PM initiates repair of the PM. Depending on the cell type, different organelles and proteins respond to this Ca(2+) entry and facilitate repair of the damaged plasma membrane. We recently identified that proteins expressed in various metastatic cancers including Ca(2+)-binding EF hand protein S100A11 and its binding partner annexin A2 are used by tumor cells for plasma membrane repair (PMR). Here we will discuss the involvement of S100, annexin proteins and their regulation of actin cytoskeleton, leading to PMR. Additionally, we will show that another S100 member--S100A4 accumulates at the injured PM. These findings reveal a new role for the S100 and annexin protein up regulation in metastatic cancers and identify these proteins and PMR as targets for treating metastatic cancers.
Collapse
Affiliation(s)
- Jyoti K Jaiswal
- a Center for Genetic Medicine Research ; Children's National Medical Center ; Washington , DC USA
| | | |
Collapse
|
33
|
Lauritzen SP, Boye TL, Nylandsted J. Annexins are instrumental for efficient plasma membrane repair in cancer cells. Semin Cell Dev Biol 2015; 45:32-8. [DOI: 10.1016/j.semcdb.2015.10.028] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Accepted: 10/15/2015] [Indexed: 01/15/2023]
|
34
|
Scheffer LL, Sreetama SC, Sharma N, Medikayala S, Brown KJ, Defour A, Jaiswal JK. Mechanism of Ca²⁺-triggered ESCRT assembly and regulation of cell membrane repair. Nat Commun 2014; 5:5646. [PMID: 25534348 PMCID: PMC4333728 DOI: 10.1038/ncomms6646] [Citation(s) in RCA: 251] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2014] [Accepted: 10/22/2014] [Indexed: 12/11/2022] Open
Abstract
In muscle and other mechanically active tissue, cell membranes are constantly injured, and their repair depends on the injury-induced increase in cytosolic calcium. Here, we show that injury-triggered Ca(2+) increase results in assembly of ESCRT III and accessory proteins at the site of repair. This process is initiated by the calcium-binding protein-apoptosis-linked gene (ALG)-2. ALG-2 facilitates accumulation of ALG-2-interacting protein X (ALIX), ESCRT III and Vps4 complex at the injured cell membrane, which in turn results in cleavage and shedding of the damaged part of the cell membrane. Lack of ALG-2, ALIX or Vps4B each prevents shedding, and repair of the injured cell membrane. These results demonstrate Ca(2+)-dependent accumulation of ESCRT III-Vps4 complex following large focal injury to the cell membrane and identify the role of ALG-2 as the initiator of sequential ESCRT III-Vps4 complex assembly that facilitates scission and repair of the injured cell membrane.
Collapse
Affiliation(s)
- Luana L Scheffer
- Children's National Medical Center, Center for Genetic Medicine Research, 111 Michigan Avenue, NW, Washington DC 20010-2970, USA
| | - Sen Chandra Sreetama
- Children's National Medical Center, Center for Genetic Medicine Research, 111 Michigan Avenue, NW, Washington DC 20010-2970, USA
| | - Nimisha Sharma
- Children's National Medical Center, Center for Genetic Medicine Research, 111 Michigan Avenue, NW, Washington DC 20010-2970, USA
| | - Sushma Medikayala
- Children's National Medical Center, Center for Genetic Medicine Research, 111 Michigan Avenue, NW, Washington DC 20010-2970, USA
| | - Kristy J Brown
- 1] Children's National Medical Center, Center for Genetic Medicine Research, 111 Michigan Avenue, NW, Washington DC 20010-2970, USA [2] Department of Integrative Systems Biology, George Washington University School of Medicine and Health Sciences, Washington DC, USA
| | - Aurelia Defour
- Children's National Medical Center, Center for Genetic Medicine Research, 111 Michigan Avenue, NW, Washington DC 20010-2970, USA
| | - Jyoti K Jaiswal
- 1] Children's National Medical Center, Center for Genetic Medicine Research, 111 Michigan Avenue, NW, Washington DC 20010-2970, USA [2] Department of Integrative Systems Biology, George Washington University School of Medicine and Health Sciences, Washington DC, USA
| |
Collapse
|
35
|
Artemenko K, Horáková J, Steinberger B, Besenfelder U, Brem G, Bergquist J, Mayrhofer C. A proteomic approach to monitor the dynamic response of the female oviductal epithelial cell surface to male gametes. J Proteomics 2014; 113:1-14. [PMID: 25281772 DOI: 10.1016/j.jprot.2014.09.016] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2014] [Revised: 09/18/2014] [Accepted: 09/22/2014] [Indexed: 12/11/2022]
Abstract
UNLABELLED Sophisticated strategies to analyze cell surface proteins are indispensable to study fundamental biological processes, such as the response of cells to environmental changes or cell-cell communication. Herein, we describe a refined mass spectrometry-based approach for the specific characterization and quantitation of cell surface proteins expressed in the female reproductive tract. The strategy is based on in situ biotinylation of rabbit oviducts, affinity enrichment of surface exposed biotin tagged proteins and dimethyl labeling of the obtained tryptic peptides followed by LC-MS/MS analysis. This approach proved to be sensitive enough to analyze small sample amounts (<1μg) and allowed further to trace the dynamic composition of the surface proteome of the oviductal epithelium in response to male gametes. The relative protein expression ratios of 175 proteins were quantified. Thirty-one of them were found to be altered over time, namely immediately, 1h and 2h after insemination compared to the time-matched control groups. Functional analysis demonstrated that structural reorganization of the oviductal epithelial cell surface was involved in the early response of the female organ to semen. In summary, this study outlines a workflow that is capable to monitor alterations in the female oviduct that are related to key reproductive processes in vivo. BIOLOGICAL SIGNIFICANCE The proper interaction between the female reproductive tract, in particular, the oviduct and the male gametes, is fundamental to fertilization and embryonic development under physiological conditions. Thereby the oviductal epithelial cell surface proteins play an important role. Besides their direct interaction with male gametes, these molecules participate in signal transduction and, thus, are involved in the mandatory cellular response of the oviductal epithelium. In this study we present a refined LC-MS/MS based workflow that is capable to quantitatively analyze the expression of oviductal epithelial cell surface proteins in response to insemination in vivo. A special focus was on the very early interaction between the female organ and the male gametes. At first, this study clearly revealed an immediate response of the surface proteome to semen, which was modulated over time. The described methodology can be applied for studies of further distinct biological events in the oviduct and therefore contribute to a deeper insight into the formation of new life.
Collapse
Affiliation(s)
- Konstantin Artemenko
- Institute of Analytical Chemistry, Department of Chemistry - Biomedical Center and Science for Life Laboratory, Uppsala University, Uppsala, Sweden.
| | - Jana Horáková
- Institute of Analytical Chemistry, Department of Chemistry - Biomedical Center and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Birgit Steinberger
- Institute of Animal Breeding and Genetics, Department for Biomedical Sciences, University of Veterinary Medicine, Vienna, Austria; Institute of Biotechnology in Animal Production, Department for Agrobiotechnology (IFA Tulln), University of Natural Resources and Applied Life Sciences, Vienna, Tulln, Austria
| | - Urban Besenfelder
- Institute of Biotechnology in Animal Production, Department for Agrobiotechnology (IFA Tulln), University of Natural Resources and Applied Life Sciences, Vienna, Tulln, Austria
| | - Gottfried Brem
- Institute of Animal Breeding and Genetics, Department for Biomedical Sciences, University of Veterinary Medicine, Vienna, Austria
| | - Jonas Bergquist
- Institute of Analytical Chemistry, Department of Chemistry - Biomedical Center and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Corina Mayrhofer
- Institute of Animal Breeding and Genetics, Department for Biomedical Sciences, University of Veterinary Medicine, Vienna, Austria; Institute of Biotechnology in Animal Production, Department for Agrobiotechnology (IFA Tulln), University of Natural Resources and Applied Life Sciences, Vienna, Tulln, Austria
| |
Collapse
|
36
|
Andrews NW, Almeida PE, Corrotte M. Damage control: cellular mechanisms of plasma membrane repair. Trends Cell Biol 2014; 24:734-42. [PMID: 25150593 DOI: 10.1016/j.tcb.2014.07.008] [Citation(s) in RCA: 226] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2014] [Revised: 07/29/2014] [Accepted: 07/31/2014] [Indexed: 02/06/2023]
Abstract
When wounded, eukaryotic cells reseal in a few seconds. Ca(2+) influx induces exocytosis of lysosomes, a process previously thought to promote repair by 'patching' wounds. New evidence suggests that resealing involves direct wound removal. Exocytosis of lysosomal acid sphingomyelinase (ASM) triggers endocytosis of lesions followed by intracellular degradation. Characterization of injury-induced endosomes revealed a role for caveolae, sphingolipid-enriched plasma membrane invaginations that internalize toxin pores and are abundant in mechanically stressed cells. These findings provide a novel mechanistic explanation for the muscle pathology associated with mutations in caveolar proteins. Membrane remodeling by the ESCRT complex was also recently shown to participate in small-wound repair, emphasizing that cell resealing involves previously unrecognized mechanisms for lesion removal that are distinct from the patch model.
Collapse
Affiliation(s)
- Norma W Andrews
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD 20742-5815, USA.
| | - Patricia E Almeida
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD 20742-5815, USA; Department of Biology, Federal University of Juiz de Fora, Juiz de Fora, Minas Gerais, Brazil
| | - Matthias Corrotte
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD 20742-5815, USA
| |
Collapse
|
37
|
Defour A, Van der Meulen JH, Bhat R, Bigot A, Bashir R, Nagaraju K, Jaiswal JK. Dysferlin regulates cell membrane repair by facilitating injury-triggered acid sphingomyelinase secretion. Cell Death Dis 2014; 5:e1306. [PMID: 24967968 PMCID: PMC4079937 DOI: 10.1038/cddis.2014.272] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2014] [Revised: 05/08/2014] [Accepted: 05/20/2014] [Indexed: 01/17/2023]
Abstract
Dysferlin deficiency compromises the repair of injured muscle, but the underlying cellular mechanism remains elusive. To study this phenomenon, we have developed mouse and human myoblast models for dysferlinopathy. These dysferlinopathic myoblasts undergo normal differentiation but have a deficit in their ability to repair focal injury to their cell membrane. Imaging cells undergoing repair showed that dysferlin-deficit decreased the number of lysosomes present at the cell membrane, resulting in a delay and reduction in injury-triggered lysosomal exocytosis. We find repair of injured cells does not involve formation of intracellular membrane patch through lysosome-lysosome fusion; instead, individual lysosomes fuse with the injured cell membrane, releasing acid sphingomyelinase (ASM). ASM secretion was reduced in injured dysferlinopathic cells, and acute treatment with sphingomyelinase restored the repair ability of dysferlinopathic myoblasts and myofibers. Our results provide the mechanism for dysferlin-mediated repair of skeletal muscle sarcolemma and identify ASM as a potential therapy for dysferlinopathy.
Collapse
Affiliation(s)
- A Defour
- Center for Genetic Medicine Research, Children's National Medical Center, 111 Michigan Avenue NW, Washington, DC, USA
| | - J H Van der Meulen
- Center for Genetic Medicine Research, Children's National Medical Center, 111 Michigan Avenue NW, Washington, DC, USA
| | - R Bhat
- Center for Genetic Medicine Research, Children's National Medical Center, 111 Michigan Avenue NW, Washington, DC, USA
| | - A Bigot
- Institut de Myologie, UM76 Université Pierre et Marie Curie, U974 INSERM, UMR7215 CNRS, GH Pitié-Salpétrière, 47 bd de l'Hôpital, Paris, France
| | - R Bashir
- School of Biological and Biochemical Sciences, University of Durham, Durham, UK
| | - K Nagaraju
- Center for Genetic Medicine Research, Children's National Medical Center, 111 Michigan Avenue NW, Washington, DC, USA
- Department of Integrative Systems Biology, George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | - J K Jaiswal
- Center for Genetic Medicine Research, Children's National Medical Center, 111 Michigan Avenue NW, Washington, DC, USA
- Department of Integrative Systems Biology, George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| |
Collapse
|
38
|
Pratt SJP, Shah SB, Ward CW, Kerr JP, Stains JP, Lovering RM. Recovery of altered neuromuscular junction morphology and muscle function in mdx mice after injury. Cell Mol Life Sci 2014; 72:153-64. [PMID: 24947322 PMCID: PMC4282693 DOI: 10.1007/s00018-014-1663-7] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2014] [Revised: 05/14/2014] [Accepted: 06/02/2014] [Indexed: 12/02/2022]
Abstract
Duchenne muscular dystrophy (DMD) is a devastating neuromuscular disease in which weakness, increased susceptibility to muscle injury, and inadequate repair underlie the pathology. While most attention has focused within the muscle fiber, we recently demonstrated significant alterations in the neuromuscular junction (NMJ) morphology and resulting neuromuscular transmission failure (NTF) 24 h after injury in mdx mice (murine model for DMD). Here we determine the contribution of NMJ morphology and NTF to the recovery of muscle contractile function post-injury. NMJ morphology and NTF rates were assessed day 0 (immediately after injury) and days 1, 7, 14 and 21 after quadriceps injury. Eccentric injury of the quadriceps resulted in a significant loss of maximal torque in both WT (39 ± 6 %) and mdx (76 ± 8 %) with a full recovery in WT by day 7 and in mdx by day 21. Post-injury alterations in NMJ morphology and NTF were found only in mdx, were limited to days 0 and 1, and were independent of changes in MuSK or AChR expression. Such early changes at the NMJ after injury are consistent with mechanical disruption rather than newly forming NMJs. Furthermore, we show that the dense microtubule network that underlies the NMJ is significantly reduced and disorganized in mdx compared to WT. These structural changes at the NMJ may play a role in the increased NMJ disruption and the exaggerated loss of nerve-evoked muscle force seen after injury to dystrophic muscles.
Collapse
Affiliation(s)
- Stephen J. P. Pratt
- Department of Orthopaedics, University of Maryland School of Medicine, 100 Penn St. AHB, Room 540, Baltimore, MD 21201 USA
| | - Sameer B. Shah
- Department of Orthopaedic Surgery and Bioengineering, University of California, San Diego, USA
| | | | - Jaclyn P. Kerr
- Department of Physiology, University of Maryland School of Medicine, Baltimore, USA
| | - Joseph P. Stains
- Department of Orthopaedics, University of Maryland School of Medicine, 100 Penn St. AHB, Room 540, Baltimore, MD 21201 USA
| | - Richard M. Lovering
- Department of Orthopaedics, University of Maryland School of Medicine, 100 Penn St. AHB, Room 540, Baltimore, MD 21201 USA
- Department of Physiology, University of Maryland School of Medicine, Baltimore, USA
| |
Collapse
|
39
|
Abstract
Dystrophin and utrophin are highly similar proteins that both link cortical actin filaments with a complex of sarcolemmal glycoproteins, yet localize to different subcellular domains within normal muscle cells. In mdx mice and Duchenne muscular dystrophy patients, dystrophin is lacking and utrophin is consequently up-regulated and redistributed to locations normally occupied by dystrophin. Transgenic overexpression of utrophin has been shown to significantly improve aspects of the disease phenotype in the mdx mouse; therefore, utrophin up-regulation is under intense investigation as a potential therapy for Duchenne muscular dystrophy. Here we biochemically compared the previously documented microtubule binding activity of dystrophin with utrophin and analyzed several transgenic mouse models to identify phenotypes of the mdx mouse that remain despite transgenic utrophin overexpression. Our in vitro analyses revealed that dystrophin binds microtubules with high affinity and pauses microtubule polymerization, whereas utrophin has no activity in either assay. We also found that transgenic utrophin overexpression does not correct subsarcolemmal microtubule lattice disorganization, loss of torque production after in vivo eccentric contractions, or physical inactivity after mild exercise. Finally, our data suggest that exercise-induced inactivity correlates with loss of sarcolemmal neuronal NOS localization in mdx muscle, whereas loss of in vivo torque production after eccentric contraction-induced injury is associated with microtubule lattice disorganization.
Collapse
|
40
|
Albrecht DE, Rufibach LE, Williams BA, Lee ER, Windish HP, Hwang EY, Shira SR, Mittal P. 6th Dysferlin Conference, 3-6 April 2013, Arlington, Virginia, USA. Neuromuscul Disord 2014; 24:277-87. [PMID: 24480524 DOI: 10.1016/j.nmd.2013.11.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2013] [Accepted: 11/14/2013] [Indexed: 11/29/2022]
Abstract
The 2013 Dysferlin Conference, sponsored and organized by the Jain Foundation, was held from April 3-6, 2013 in Arlington, VA. Participants included 34 researcher speakers, 5 dysferlinopathy patients and all 8 members of the Jain Foundation team. Dysferlinopathy is a rare disease that typically robs patients of mobility during their second or third decade of life. The goals of these Dysferlin Conferences are to bring experts in the field together so that they will collaborate with one another, to quicken the pace of understanding the biology of the disease and to build effective platforms to ameliorate disease. This is important because the function of dysferlin and how to compensate for its absence is still not well understood, in spite of the fact that the dysferlin gene was identified more than a decade ago. The objective of this conference, therefore, was to share and discuss the newest unpublished research defining the role of dysferlin in skeletal muscle, why its absence causes muscular dystrophy and possible therapies for dysferlin-deficient muscular dystrophy patients.
Collapse
Affiliation(s)
| | - Laura E Rufibach
- Jain Foundation Inc., 9725 Third Ave NE, Suite 204, Seattle, WA, USA
| | | | - Elaine R Lee
- Jain Foundation Inc., 9725 Third Ave NE, Suite 204, Seattle, WA, USA
| | | | - Esther Y Hwang
- Jain Foundation Inc., 9725 Third Ave NE, Suite 204, Seattle, WA, USA
| | - Sarah R Shira
- Jain Foundation Inc., 9725 Third Ave NE, Suite 204, Seattle, WA, USA
| | - Plavi Mittal
- Jain Foundation Inc., 9725 Third Ave NE, Suite 204, Seattle, WA, USA.
| |
Collapse
|
41
|
Rayavarapu S, Coley W, Van der Meulen JH, Cakir E, Tappeta K, Kinder TB, Dillingham BC, Brown KJ, Hathout Y, Nagaraju K. Activation of the ubiquitin proteasome pathway in a mouse model of inflammatory myopathy: a potential therapeutic target. ACTA ACUST UNITED AC 2014; 65:3248-58. [PMID: 24022788 DOI: 10.1002/art.38180] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2013] [Accepted: 08/27/2013] [Indexed: 01/01/2023]
Abstract
OBJECTIVE Myositis is characterized by severe muscle weakness. We and others have previously shown that endoplasmic reticulum (ER) stress plays a role in the pathogenesis of myositis. The present study was undertaken to identify perturbed pathways and assess their contribution to muscle disease in a mouse myositis model. METHODS Stable isotope labeling with amino acids in cell culture (SILAC) was used to identify alterations in the skeletal muscle proteome of myositic mice in vivo. Differentially altered protein levels identified in the initial comparisons were validated using a liquid chromatography tandem mass spectrometry spike-in strategy and further confirmed by immunoblotting. In addition, we evaluated the effect of a proteasome inhibitor, bortezomib, on the disease phenotype, using well-standardized functional, histologic, and biochemical assessments. RESULTS With the SILAC technique we identified significant alterations in levels of proteins belonging to the ER stress response, ubiquitin proteasome pathway (UPP), oxidative phosphorylation, glycolysis, cytoskeleton, and muscle contractile apparatus categories. We validated the myositis-related changes in the UPP and demonstrated a significant increase in the ubiquitination of muscle proteins as well as a specific increase in ubiquitin carboxyl-terminal hydrolase isozyme L1 (UCHL-1) in myositis, but not in muscle affected by other dystrophies or normal muscle. Inhibition of the UPP with bortezomib significantly improved muscle function and also significantly reduced tumor necrosis factor α expression in the skeletal muscle of mice with myositis. CONCLUSION Our findings indicate that ER stress activates downstream UPPs and contributes to muscle degeneration and that UCHL-1 is a potential biomarker for disease progression. UPP inhibition offers a potential therapeutic strategy for myositis.
Collapse
Affiliation(s)
- Sree Rayavarapu
- Children's National Medical Center and George Washington University, Washington DC
| | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Heier CR, Damsker JM, Yu Q, Dillingham BC, Huynh T, Van der Meulen JH, Sali A, Miller BK, Phadke A, Scheffer L, Quinn J, Tatem K, Jordan S, Dadgar S, Rodriguez OC, Albanese C, Calhoun M, Gordish-Dressman H, Jaiswal JK, Connor EM, McCall JM, Hoffman EP, Reeves EKM, Nagaraju K. VBP15, a novel anti-inflammatory and membrane-stabilizer, improves muscular dystrophy without side effects. EMBO Mol Med 2013; 5:1569-85. [PMID: 24014378 PMCID: PMC3799580 DOI: 10.1002/emmm.201302621] [Citation(s) in RCA: 142] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2013] [Revised: 07/30/2013] [Accepted: 08/02/2013] [Indexed: 01/01/2023] Open
Abstract
Absence of dystrophin makes skeletal muscle more susceptible to injury, resulting in breaches of the plasma membrane and chronic inflammation in Duchenne muscular dystrophy (DMD). Current management by glucocorticoids has unclear molecular benefits and harsh side effects. It is uncertain whether therapies that avoid hormonal stunting of growth and development, and/or immunosuppression, would be more or less beneficial. Here, we discover an oral drug with mechanisms that provide efficacy through anti-inflammatory signaling and membrane-stabilizing pathways, independent of hormonal or immunosuppressive effects. We find VBP15 protects and promotes efficient repair of skeletal muscle cells upon laser injury, in opposition to prednisolone. Potent inhibition of NF-κB is mediated through protein interactions of the glucocorticoid receptor, however VBP15 shows significantly reduced hormonal receptor transcriptional activity. The translation of these drug mechanisms into DMD model mice improves muscle strength, live-imaging and pathology through both preventive and post-onset intervention regimens. These data demonstrate successful improvement of dystrophy independent of hormonal, growth, or immunosuppressive effects, indicating VBP15 merits clinical investigation for DMD and would benefit other chronic inflammatory diseases.
Collapse
Affiliation(s)
- Christopher R Heier
- Center for Genetic Medicine Research, Children's National Medical Center, Washington, DC, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Cohen TV, Gnocchi VF, Cohen JE, Phadke A, Liu H, Ellis JA, Foisner R, Stewart CL, Zammit PS, Partridge TA. Defective skeletal muscle growth in lamin A/C-deficient mice is rescued by loss of Lap2α. Hum Mol Genet 2013; 22:2852-69. [PMID: 23535822 DOI: 10.1093/hmg/ddt135] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Mutations in lamin A/C result in a range of tissue-specific disorders collectively called laminopathies. Of these, Emery-Dreifuss and Limb-Girdle muscular dystrophy 1B mainly affect striated muscle. A useful model for understanding both laminopathies and lamin A/C function is the Lmna(-/-) mouse. We found that skeletal muscle growth and muscle satellite (stem) cell proliferation were both reduced in Lmna(-/-) mice. Lamins A and C associate with lamina-associated polypeptide 2 alpha (Lap2α) and the retinoblastoma gene product, pRb, to regulate cell cycle exit. We found Lap2α to be upregulated in Lmna(-/-) myoblasts (MBs). To specifically test the contribution of elevated Lap2α to the phenotype of Lmna(-/-) mice, we generated Lmna(-/-)Lap2α(-/-) mice. Lifespan and body mass were increased in Lmna(-/-)Lap2α(-/-) mice compared with Lmna(-/-). Importantly, the satellite cell proliferation defect was rescued, resulting in improved myogenesis. Lmna(-/-) MBs also exhibited increased levels of Smad2/3, which were abnormally distributed in the cell and failed to respond to TGFβ1 stimulation as in control cells. However, using SIS3 to inhibit signaling via Smad3 reduced cell death and augmented MB fusion. Together, our results show that perturbed Lap2α/pRb and Smad2/3 signaling are important regulatory pathways mediating defective muscle growth in Lmna(-/-) mice, and that inhibition of either pathway alone or in combination can ameliorate this deleterious phenotype.
Collapse
Affiliation(s)
- Tatiana V Cohen
- Research Center for Genetic Medicine, Children’s National Medical Center, 111 Michigan Avenue NW, Washington, DC 20010, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Brown KJ, Marathi R, Fiorillo AA, Ciccimaro EF, Sharma S, Rowlands DS, Rayavarapu S, Nagaraju K, Hoffman EP, Hathout Y. Accurate Quantitation of Dystrophin Protein in Human Skeletal Muscle Using Mass Spectrometry. ACTA ACUST UNITED AC 2013; Suppl 7. [PMID: 23646235 DOI: 10.4172/1948-593x.s7-001] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Quantitation of human dystrophin protein in muscle biopsies is a clinically relevant endpoint for both diagnosis and response to dystrophin-replacement therapies for dystrophinopathies. A robust and accurate assay would enable the use of dystrophin as a surrogate biomarker, particularly in exploratory Phase 2 trials. Currently available methods to quantitate dystrophin rely on immunoblot or immunohistochemistry methods that are not considered robust. Here we present a mass spectrometry based approach to accurately quantitate dystrophin protein in a total protein extract from human muscle biopsies. Our approach uses a combination of stable isotope labeled dystrophin as a spike-in standard, gel electrophoresis and high precision mass spectrometry to detect and quantitate multiple peptides of dystrophin within a complex protein mixture. The method was found highly reproducible and linear over a wide dynamic range, detecting as low as 5% of dystrophin relative to the normal amount in healthy individuals.
Collapse
Affiliation(s)
- Kristy J Brown
- Children's National Medical Center, Center for Genetic Medicine Research, USA ; Department of Integrative Systems Biology, The George Washington University, 2300 Eye Street, N.W., Ross 605, Washington, D.C. USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|