51
|
Boucher E, Goldin-Blais L, Basiren Q, Mandato CA. Actin dynamics and myosin contractility during plasma membrane repair and restoration: Does one ring really heal them all? CURRENT TOPICS IN MEMBRANES 2019; 84:17-41. [PMID: 31610862 DOI: 10.1016/bs.ctm.2019.07.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
In order to survive daily insults, cells have evolved various mechanisms that detect, stabilize and repair damages done to their plasma membrane and cytoskeletal structures. Damage to the PM endangers wounded cells by exposing them to uncontrolled exchanges with the extracellular milieu. The processes and molecular machinery enabling PM repair are therefore at the center of the bulk of the investigations into single-cell repair program. Wounds are repaired by dynamically remodeling the composition and shape of the injured area through exocytosis-mediated release of intracellular membrane components to the wounded area, endocytosis-mediated removal of the injured area, or the shedding of the injury. The wound healing program of Xenopus oocytes and early Drosophila embryos is by contrast, mostly characterized by the rapid formation of a large membrane patch over the wound that eventually fuse with the plasma membrane which restores plasma membrane continuity and lead to the shedding of patch material into the extracellular space. Formation and contraction of actomyosin ring restores normal plasma membrane composition and organizes cytoskeletal repairs. The extend of the contributions of the cytoskeleton to the wound healing program of somatic cells have comparatively received little attention. This review offers a survey of the current knowledge on how actin dynamics, myosin-based contraction and other cytoskeletal structures affects PM and cortical cytoskeleton repair of somatic cells.
Collapse
Affiliation(s)
- Eric Boucher
- Department of Anatomy and Cell Biology, Faculty of Medicine, McGill University, Montreal, QC, Canada
| | - Laurence Goldin-Blais
- Department of Anatomy and Cell Biology, Faculty of Medicine, McGill University, Montreal, QC, Canada
| | - Quentin Basiren
- Department of Anatomy and Cell Biology, Faculty of Medicine, McGill University, Montreal, QC, Canada
| | - Craig A Mandato
- Department of Anatomy and Cell Biology, Faculty of Medicine, McGill University, Montreal, QC, Canada.
| |
Collapse
|
52
|
Bittel DC, Jaiswal JK. Contribution of Extracellular Vesicles in Rebuilding Injured Muscles. Front Physiol 2019; 10:828. [PMID: 31379590 PMCID: PMC6658195 DOI: 10.3389/fphys.2019.00828] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 06/13/2019] [Indexed: 12/22/2022] Open
Abstract
Skeletal myofibers are injured due to mechanical stresses experienced during physical activity, or due to myofiber fragility caused by genetic diseases. The injured myofiber needs to be repaired or regenerated to restore the loss in muscle tissue function. Myofiber repair and regeneration requires coordinated action of various intercellular signaling factors-including proteins, inflammatory cytokines, miRNAs, and membrane lipids. It is increasingly being recognized release and transmission of these signaling factors involves extracellular vesicle (EV) released by myofibers and other cells in the injured muscle. Intercellular signaling by these EVs alters the phenotype of their target cells either by directly delivering the functional proteins and lipids or by modifying longer-term gene expression. These changes in the target cells activate downstream pathways involved in tissue homeostasis and repair. The EVs are heterogeneous with regards to their size, composition, cargo, location, as well as time-course of genesis and release. These differences impact on the subsequent repair and regeneration of injured skeletal muscles. This review focuses on how intracellular vesicle production, cargo packaging, and secretion by injured muscle, modulates specific reparative, and regenerative processes. Insights into the formation of these vesicles and their signaling properties offer new understandings of the orchestrated response necessary for optimal muscle repair and regeneration.
Collapse
Affiliation(s)
- Daniel C Bittel
- Children's National Health System, Center for Genetic Medicine Research, Washington, DC, United States
| | - Jyoti K Jaiswal
- Children's National Health System, Center for Genetic Medicine Research, Washington, DC, United States.,Department of Genomics and Precision Medicine, George Washington University School of Medicine and Health Sciences, Washington, DC, United States
| |
Collapse
|
53
|
Spaulding HR, Quindry T, Hammer K, Quindry JC, Selsby JT. Nutraceutical and pharmaceutical cocktails did not improve muscle function or reduce histological damage in D2-mdx mice. J Appl Physiol (1985) 2019; 127:1058-1066. [PMID: 31295065 DOI: 10.1152/japplphysiol.00162.2019] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Progressive muscle injury and weakness are hallmarks of Duchenne muscular dystrophy. We showed previously that quercetin (Q) partially protected dystrophic limb muscles from disease-related injury. As quercetin activates PGC-1α through Sirtuin-1, an NAD+-dependent deacetylase, the depleted NAD+ in dystrophic skeletal muscle may limit quercetin efficacy; hence, supplementation with the NAD+ donor, nicotinamide riboside (NR), may facilitate quercetin efficacy. Lisinopril (Lis) protects skeletal muscle and improves cardiac function in dystrophin-deficient mice; therefore, it was included in this study to evaluate the effects of lisinopril used with quercetin and NR. Our purpose was to determine the extent to which Q, NR, and Lis decreased dystrophic injury. We hypothesized that Q, NR, or Lis alone would improve muscle function and decrease histological injury and when used in combination would have additive effects. Muscle function of 11-mo-old DBA (healthy), D2-mdx (dystrophin-deficient), and D2-mdx mice was assessed after treatment with Q, NR, and/or Lis for 7 mo. To mimic typical pharmacology of patients with Duchenne muscular dystrophy, a group was treated with prednisolone (Pred) in combination with Q, NR, and Lis. At 11 mo of age, dystrophin deficiency decreased specific tension and tetanic force in the soleus and extensor digitorum longus muscles and was not corrected by any treatment. Dystrophic muscle was more sensitive to contraction-induced injury, which was partially offset in the QNRLisPred group, whereas fatigue was similar between all groups. Treatments did not decrease histological damage. These data suggest that treatment with Q, NR, Lis, and Pred failed to adequately maintain dystrophic limb muscle function or decrease histological damage.NEW & NOTEWORTHY Despite a compelling rationale and previous evidence to the contrary in short-term investigations, quercetin, nicotinamide riboside, or Lisinopril, alone or in combination, failed to restore muscle function or decrease histological injury in dystrophic limb muscle from D2-mdx mice after long-term administration. Importantly, we also found that in the D2-mdx model, an emerging and relatively understudied model of Duchenne muscular dystrophy dystrophin deficiency caused profound muscle dysfunction and histopathology in skeletal muscle.
Collapse
Affiliation(s)
| | - Tiffany Quindry
- Department of Health and Human Performance, University of Montana, Missoula, Montana
| | - Kayleen Hammer
- Department of Animal Science, Iowa State University, Ames, Iowa
| | - John C Quindry
- Department of Health and Human Performance, University of Montana, Missoula, Montana
| | - Joshua T Selsby
- Department of Animal Science, Iowa State University, Ames, Iowa
| |
Collapse
|
54
|
Young CS, Mokhonova E, Quinonez M, Pyle AD, Spencer MJ. Creation of a Novel Humanized Dystrophic Mouse Model of Duchenne Muscular Dystrophy and Application of a CRISPR/Cas9 Gene Editing Therapy. J Neuromuscul Dis 2019; 4:139-145. [PMID: 28505980 PMCID: PMC5565771 DOI: 10.3233/jnd-170218] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Duchenne muscular dystrophy is caused by mutations in DMD which disrupt the reading frame. Therapeutic strategies that restore DMD's reading frame, such as exon skipping and CRISPR/Cas9, need to be tested in the context of the human DMD sequence in vivo. We have developed a novel dystrophic mouse model by using CRISPR/Cas9 to delete exon 45 in the human DMD gene in hDMD mice, which places DMD out-of-frame. We have utilized this model to demonstrate that our clinically-relevant CRISPR/Cas9 platform, which targets deletion of human DMD exons 45-55, can be directly applied in vivo to restore dystrophin.
Collapse
Affiliation(s)
- Courtney S. Young
- Molecular Biology Interdepartmental Program, University of California, Los Angeles, CA, USA
- Center for Duchenne Muscular Dystrophy at UCLA, University of California, Los Angeles, CA, USA
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, CA, USA
| | - Ekaterina Mokhonova
- Center for Duchenne Muscular Dystrophy at UCLA, University of California, Los Angeles, CA, USA
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Marbella Quinonez
- Center for Duchenne Muscular Dystrophy at UCLA, University of California, Los Angeles, CA, USA
- Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - April D. Pyle
- Center for Duchenne Muscular Dystrophy at UCLA, University of California, Los Angeles, CA, USA
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, CA, USA
- Department of Microbiology, Immunology and Molecular Genetics, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Melissa J. Spencer
- Molecular Biology Interdepartmental Program, University of California, Los Angeles, CA, USA
- Center for Duchenne Muscular Dystrophy at UCLA, University of California, Los Angeles, CA, USA
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, CA, USA
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| |
Collapse
|
55
|
Vo AH, Swaggart KA, Woo A, Gao QQ, Demonbreun AR, Fallon KS, Quattrocelli M, Hadhazy M, Page PGT, Chen Z, Eskin A, Squire K, Nelson SF, McNally EM. Dusp6 is a genetic modifier of growth through enhanced ERK activity. Hum Mol Genet 2019; 28:279-289. [PMID: 30289454 DOI: 10.1093/hmg/ddy349] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Accepted: 09/26/2018] [Indexed: 12/21/2022] Open
Abstract
Like other single-gene disorders, muscular dystrophy displays a range of phenotypic heterogeneity even with the same primary mutation. Identifying genetic modifiers capable of altering the course of muscular dystrophy is one approach to deciphering gene-gene interactions that can be exploited for therapy development. To this end, we used an intercross strategy in mice to map modifiers of muscular dystrophy. We interrogated genes of interest in an interval on mouse chromosome 10 associated with body mass in muscular dystrophy as skeletal muscle contributes significantly to total body mass. Using whole-genome sequencing of the two parental mouse strains combined with deep RNA sequencing, we identified the Met62Ile substitution in the dual-specificity phosphatase 6 (Dusp6) gene from the DBA/2 J (D2) mouse strain. DUSP6 is a broadly expressed dual-specificity phosphatase protein, which binds and dephosphorylates extracellular-signal-regulated kinase (ERK), leading to decreased ERK activity. We found that the Met62Ile substitution reduced the interaction between DUSP6 and ERK resulting in increased ERK phosphorylation and ERK activity. In dystrophic muscle, DUSP6 Met62Ile is strongly upregulated to counteract its reduced activity. We found that myoblasts from the D2 background were insensitive to a specific small molecule inhibitor of DUSP6, while myoblasts expressing the canonical DUSP6 displayed enhanced proliferation after exposure to DUSP6 inhibition. These data identify DUSP6 as an important regulator of ERK activity in the setting of muscle growth and muscular dystrophy.
Collapse
Affiliation(s)
- Andy H Vo
- Committee on Development, Regeneration and Stem Cell Biology, The University of Chicago, Chicago, IL
| | | | - Anna Woo
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago IL
| | - Quan Q Gao
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago IL
| | - Alexis R Demonbreun
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago IL
| | - Katherine S Fallon
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago IL
| | - Mattia Quattrocelli
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago IL
| | - Michele Hadhazy
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago IL
| | - Patrick G T Page
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago IL
| | - Zugen Chen
- Departments of Human Genetics and Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Ascia Eskin
- Departments of Human Genetics and Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Kevin Squire
- Departments of Human Genetics and Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Stanley F Nelson
- Departments of Human Genetics and Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Elizabeth M McNally
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago IL
| |
Collapse
|
56
|
van Putten M, Putker K, Overzier M, Adamzek WA, Pasteuning-Vuhman S, Plomp JJ, Aartsma-Rus A. Natural disease history of the D2 -mdx mouse model for Duchenne muscular dystrophy. FASEB J 2019; 33:8110-8124. [PMID: 30933664 PMCID: PMC6593893 DOI: 10.1096/fj.201802488r] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The C57BL/10ScSn-Dmdmdx/J (BL10-mdx) mouse has been the most commonly used model for Duchenne muscular dystrophy (DMD) for decades. Their muscle dysfunction and pathology is, however, less severe than in patients with DMD, which complicates preclinical studies. Recent discoveries indicate that disease severity is exacerbated when muscular dystrophy mouse models are generated on a DBA2/J genetic background. Knowledge on the natural history of animal models is pivotal for high-quality preclinical testing. However, for BL10-mdx mice on a DBA2/J background (D2-mdx), limited data are available. We addressed this gap in the natural history knowledge. First, we compared histopathological aspects in skeletal muscles of young D2-mdx, BL10-mdx, and wild-type mice. Pathology was more pronounced in D2-mdx mice and differed in severity between muscles within individuals. Secondly, we subjected D2-mdx mice to a functional test regime for 34 weeks and identified that female D2-mdx mice outperform severely impaired males, making females less useful for functional preclinical studies. Direct comparisons between 10- and 34-wk-old D2-mdx mice revealed that disease pathology ameliorates with age. Heart pathology was progressive, with some features already evident at a young age. This natural history study of the D2-mdx mouse will be instrumental for experimental design of future preclinical studies.-Van Putten, M., Putker, K., Overzier, M., Adamzek, W. A., Pasteuning-Vuhman, S., Plomp, J. J., Aartsma-Rus, A. Natural disease history of the D2-mdx mouse model for Duchenne muscular dystrophy.
Collapse
Affiliation(s)
- Maaike van Putten
- Department of Human Genetics, Leiden University Medical Centre, Leiden, The Netherlands
| | - Kayleigh Putker
- Department of Human Genetics, Leiden University Medical Centre, Leiden, The Netherlands
| | - Maurice Overzier
- Department of Human Genetics, Leiden University Medical Centre, Leiden, The Netherlands
| | - W A Adamzek
- Department of Human Genetics, Leiden University Medical Centre, Leiden, The Netherlands
| | | | - Jaap J Plomp
- Department of Neurology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Annemieke Aartsma-Rus
- Department of Human Genetics, Leiden University Medical Centre, Leiden, The Netherlands
| |
Collapse
|
57
|
Gordish-Dressman H, Willmann R, Dalle Pazze L, Kreibich A, van Putten M, Heydemann A, Bogdanik L, Lutz C, Davies K, Demonbreun AR, Duan D, Elsey D, Fukada SI, Girgenrath M, Patrick Gonzalez J, Grounds MD, Nichols A, Partridge T, Passini M, Sanarica F, Schnell FJ, Wells DJ, Yokota T, Young CS, Zhong Z, Spurney C, Spencer M, De Luca A, Nagaraju K, Aartsma-Rus A. "Of Mice and Measures": A Project to Improve How We Advance Duchenne Muscular Dystrophy Therapies to the Clinic. J Neuromuscul Dis 2019; 5:407-417. [PMID: 30198876 PMCID: PMC6218134 DOI: 10.3233/jnd-180324] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
A new line of dystrophic mdx mice on the DBA/2J (D2) background has emerged as a candidate to study the efficacy of therapeutic approaches for Duchenne muscular dystrophy (DMD). These mice harbor genetic polymorphisms that appear to increase the severity of the dystropathology, with disease modifiers that also occur in DMD patients, making them attractive for efficacy studies and drug development. This workshop aimed at collecting and consolidating available data on the pathological features and the natural history of these new D2/mdx mice, for comparison with classic mdx mice and controls, and to identify gaps in information and their potential value. The overall aim is to establish guidance on how to best use the D2/mdx mouse model in preclinical studies.
Collapse
Affiliation(s)
| | | | | | | | - Maaike van Putten
- Department of Human Genetics, Leiden University Medical Center, the Netherlands
| | - Ahlke Heydemann
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, IL, USA
| | | | | | - Kay Davies
- Department of Physiology, University of Oxford, Anatomy and Genetics, Oxford, UK
| | - Alexis R Demonbreun
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Dongsheng Duan
- Department of Molecular Microbiology and Immunology, Department of Neurology, Department of Biomedical Sciences and Department of Bioengineering, University of Missouri, Columbia, MO, USA
| | - David Elsey
- Summit Therapeutics, Abingdon, Oxfordshire, UK
| | - So-Ichiro Fukada
- Laboratory of Molecular and Cellular Physiology, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
| | | | | | - Miranda D Grounds
- School of Human Science, the University of Western Australia, Perth, Australia
| | | | | | | | - Francesca Sanarica
- Department of Pharmacy and Drug Sciences, Unit of Pharmacology, University of Bari "Aldo Moro", Italy
| | | | - Dominic J Wells
- Department of Comparative Biomedical Sciences, Royal Veterinary College, London, UK
| | | | - Courtney S Young
- Department of Neurology, Molecular Biology Institute, Center for Duchenne Muscular Dystrophy at UCLA, University of California, Los Angeles, CA, USA
| | | | | | - Melissa Spencer
- Department of Neurology, Molecular Biology Institute, Center for Duchenne Muscular Dystrophy at UCLA, University of California, Los Angeles, CA, USA
| | - Annamaria De Luca
- Department of Pharmacy and Drug Sciences, Unit of Pharmacology, University of Bari "Aldo Moro", Italy
| | - Kanneboyina Nagaraju
- School of Pharmacy and Pharmaceutical Sciences, Binghamton University, New York, USA
| | | |
Collapse
|
58
|
Whalen DS, Widatalla SE, Korolkova OY, Nangami GS, Beasley HK, Williams SD, Virgous C, Lehmann BD, Ochieng J, Sakwe AM. Implication of calcium activated RasGRF2 in Annexin A6-mediated breast tumor cell growth and motility. Oncotarget 2019; 10:133-151. [PMID: 30719209 PMCID: PMC6349432 DOI: 10.18632/oncotarget.26512] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2015] [Accepted: 12/16/2018] [Indexed: 01/10/2023] Open
Abstract
The role of AnxA6 in breast cancer and in particular, the mechanisms underlying its contribution to tumor cell growth and/or motility remain poorly understood. In this study, we established the tumor suppressor function of AnxA6 in triple negative breast cancer (TNBC) cells by showing that loss of AnxA6 is associated with early onset and rapid growth of xenograft TNBC tumors in mice. We also identified the Ca2+ activated RasGRF2 as an effector of AnxA6 mediated TNBC cell growth and motility. Activation of Ca2+ mobilizing oncogenic receptors such as epidermal growth factor receptor (EGFR) in TNBC cells or pharmacological stimulation of Ca2+ influx led to activation, subsequent degradation and altered effector functions of RasGRF2. Inhibition of Ca2+ influx or overexpression of AnxA6 blocked the activation/degradation of RasGRF2. We also show that AnxA6 acts as a scaffold for RasGRF2 and Ras proteins and that its interaction with RasGRF2 is modulated by GTP and/or activation of Ras proteins. Meanwhile, down-regulation of RasGRF2 and treatment of cells with the EGFR-targeted tyrosine kinase inhibitor (TKI) lapatinib strongly attenuated the growth of otherwise EGFR-TKI resistant AnxA6 high TNBC cells. These data not only suggest that AnxA6 modulated Ca2+ influx and effector functions of RasGRF2 underlie at least in part, the AnxA6 mediated TNBC cell growth and/or motility, but also provide a rationale to target Ras-driven TNBC with EGFR targeted therapies in combination with inhibition of RasGRF2.
Collapse
Affiliation(s)
- Diva S Whalen
- Department of Biochemistry and Cancer Biology, School of Graduate Studies and Research, Meharry Medical College, Nashville, TN, USA
| | - Sarrah E Widatalla
- Department of Biochemistry and Cancer Biology, School of Graduate Studies and Research, Meharry Medical College, Nashville, TN, USA
| | - Olga Y Korolkova
- Department of Biochemistry and Cancer Biology, School of Graduate Studies and Research, Meharry Medical College, Nashville, TN, USA
| | - Gladys S Nangami
- Department of Biochemistry and Cancer Biology, School of Graduate Studies and Research, Meharry Medical College, Nashville, TN, USA
| | - Heather K Beasley
- Department of Biochemistry and Cancer Biology, School of Graduate Studies and Research, Meharry Medical College, Nashville, TN, USA
| | - Stephen D Williams
- Department of Biochemistry and Cancer Biology, School of Graduate Studies and Research, Meharry Medical College, Nashville, TN, USA
| | - Carlos Virgous
- Animal Care Facility, Meharry Medical College, Nashville, TN, USA
| | - Brian D Lehmann
- Department of Biochemistry, Vanderbilt University, Nashville, TN, USA
| | - Josiah Ochieng
- Department of Biochemistry and Cancer Biology, School of Graduate Studies and Research, Meharry Medical College, Nashville, TN, USA
| | - Amos M Sakwe
- Department of Biochemistry and Cancer Biology, School of Graduate Studies and Research, Meharry Medical College, Nashville, TN, USA
| |
Collapse
|
59
|
Houang EM, Sham YY, Bates FS, Metzger JM. Muscle membrane integrity in Duchenne muscular dystrophy: recent advances in copolymer-based muscle membrane stabilizers. Skelet Muscle 2018; 8:31. [PMID: 30305165 PMCID: PMC6180502 DOI: 10.1186/s13395-018-0177-7] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Accepted: 09/13/2018] [Indexed: 02/07/2023] Open
Abstract
The scientific premise, design, and structure-function analysis of chemical-based muscle membrane stabilizing block copolymers are reviewed here for applications in striated muscle membrane injury. Synthetic block copolymers have a rich history and wide array of applications from industry to biology. Potential for discovery is enabled by a large chemical space for block copolymers, including modifications in block copolymer mass, composition, and molecular architecture. Collectively, this presents an impressive chemical landscape to leverage distinct structure-function outcomes. Of particular relevance to biology and medicine, stabilization of damaged phospholipid membranes using amphiphilic block copolymers, classified as poloxamers or pluronics, has been the subject of increasing scientific inquiry. This review focuses on implementing block copolymers to protect fragile muscle membranes against mechanical stress. The review highlights interventions in Duchenne muscular dystrophy, a fatal disease of progressive muscle deterioration owing to marked instability of the striated muscle membrane. Biophysical and chemical engineering advances are presented that delineate and expand upon current understanding of copolymer-lipid membrane interactions and the mechanism of stabilization. The studies presented here serve to underscore the utility of copolymer discovery leading toward the therapeutic application of block copolymers in Duchenne muscular dystrophy and potentially other biomedical applications in which membrane integrity is compromised.
Collapse
Affiliation(s)
- Evelyne M. Houang
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, 6-125 Jackson Hall, 321 Church Street SE, Minneapolis, MN 55455 USA
| | - Yuk Y. Sham
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, 6-125 Jackson Hall, 321 Church Street SE, Minneapolis, MN 55455 USA
- University of Minnesota Informatics Institute, MN, USA
- Bioinformatics and Computational Biology Program, University of Minnesota, MN, USA
| | - Frank S. Bates
- Department of Chemical Engineering and Materials Science, University of Minnesota, MN, USA
| | - Joseph M. Metzger
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, 6-125 Jackson Hall, 321 Church Street SE, Minneapolis, MN 55455 USA
| |
Collapse
|
60
|
Leigh F, Ferlini A, Biggar D, Bushby K, Finkel R, Morgenroth LP, Wagner KR. Neurology Care, Diagnostics, and Emerging Therapies of the Patient With Duchenne Muscular Dystrophy. Pediatrics 2018; 142:S5-S16. [PMID: 30275245 DOI: 10.1542/peds.2018-0333c] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/26/2018] [Indexed: 11/24/2022] Open
Abstract
Duchenne muscular dystrophy is the most common form of childhood muscular dystrophy. A mutation in the DMD gene disrupts dystrophin (protein) production, causing damage to muscle integrity, weakness, loss of ambulation, and cardiopulmonary compromise by the second decade of life. Life expectancy has improved from mid-teenage years to mid-20s with the use of glucocorticoids and beyond the third decade with ventilator support and multidisciplinary care. However, Duchenne muscular dystrophy is associated with comorbidities and is a fatal disease. Glucocorticoids prolong ambulation, but their side effects are significant. Emerging investigational therapies have surfaced over the past decade and have rapidly been tested in clinical trials. Gene-specific strategies include nonsense readthrough, exon skipping, gene editing, utrophin modulation, and gene replacement. Other mechanisms include muscle regeneration, antioxidants, and antifibrosis and anti-inflammatory pathways. With potential therapies emerging, early diagnosis is needed to initiate treatment early enough to minimize morbidity and mortality. Newborn screening can be used to significantly improve early diagnosis, especially for gene-specific therapeutics.
Collapse
Affiliation(s)
- Fawn Leigh
- Massachusetts General Hospital and Harvard Medical School, Harvard University, Cambridge, Massachusetts; .,Seattle Children's Hospital, University of Washington, Seattle, Washington
| | | | - Doug Biggar
- Holland Bloorview Kids Rehabilitation Hospital, Toronto, Ontario, Canada
| | - Katharine Bushby
- John Walton Centre for Muscular Dystrophy Research, Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom
| | | | | | - Kathryn R Wagner
- Kennedy Krieger Institute, Baltimore, Maryland; and.,School of Medicine, Johns Hopkins University, Baltimore, Maryland
| |
Collapse
|
61
|
Harsini FM, Chebrolu S, Fuson KL, White MA, Rice AM, Sutton RB. FerA is a Membrane-Associating Four-Helix Bundle Domain in the Ferlin Family of Membrane-Fusion Proteins. Sci Rep 2018; 8:10949. [PMID: 30026467 PMCID: PMC6053371 DOI: 10.1038/s41598-018-29184-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2018] [Accepted: 07/04/2018] [Indexed: 12/15/2022] Open
Abstract
Ferlin proteins participate in such diverse biological events as vesicle fusion in C. elegans, fusion of myoblast membranes to form myotubes, Ca2+-sensing during exocytosis in the hair cells of the inner ear, and Ca2+-dependent membrane repair in skeletal muscle cells. Ferlins are Ca2+-dependent, phospholipid-binding, multi-C2 domain-containing proteins with a single transmembrane helix that spans a vesicle membrane. The overall domain composition of the ferlins resembles the proteins involved in exocytosis; therefore, it is thought that they participate in membrane fusion at some level. But if ferlins do fuse membranes, then they are distinct from other known fusion proteins. Here we show that the central FerA domain from dysferlin, myoferlin, and otoferlin is a novel four-helix bundle fold with its own Ca2+-dependent phospholipid-binding activity. Small-angle X-ray scattering (SAXS), spectroscopic, and thermodynamic analysis of the dysferlin, myoferlin, and otoferlin FerA domains, in addition to clinically-defined dysferlin FerA mutations, suggests that the FerA domain interacts with the membrane and that this interaction is enhanced by the presence of Ca2+.
Collapse
Affiliation(s)
- Faraz M Harsini
- Department of Cell Physiology and Molecular Biophysics, Texas Tech University Health Sciences Center, Lubbock, TX, 79430-6551, USA
| | - Sukanya Chebrolu
- Department of Cell Physiology and Molecular Biophysics, Texas Tech University Health Sciences Center, Lubbock, TX, 79430-6551, USA
| | - Kerry L Fuson
- Department of Cell Physiology and Molecular Biophysics, Texas Tech University Health Sciences Center, Lubbock, TX, 79430-6551, USA
| | - Mark A White
- Department of Biochemistry and Molecular Biology, The University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Anne M Rice
- Department of Biophysics, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - R Bryan Sutton
- Department of Cell Physiology and Molecular Biophysics, Texas Tech University Health Sciences Center, Lubbock, TX, 79430-6551, USA. .,Center for Membrane Protein Research, Texas Tech University Health Sciences Center, Lubbock, TX, 79430-6551, USA.
| |
Collapse
|
62
|
Annexins in Translational Research: Hidden Treasures to Be Found. Int J Mol Sci 2018; 19:ijms19061781. [PMID: 29914106 PMCID: PMC6032224 DOI: 10.3390/ijms19061781] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Revised: 06/06/2018] [Accepted: 06/12/2018] [Indexed: 12/12/2022] Open
Abstract
The vertebrate annexin superfamily (AnxA) consists of 12 members of a calcium (Ca2+) and phospholipid binding protein family which share a high structural homology. In keeping with this hallmark feature, annexins have been implicated in the Ca2+-controlled regulation of a broad range of membrane events. In this review, we identify and discuss several themes of annexin actions that hold a potential therapeutic value, namely, the regulation of the immune response and the control of tissue homeostasis, and that repeatedly surface in the annexin activity profile. Our aim is to identify and discuss those annexin properties which might be exploited from a translational science and specifically, a clinical point of view.
Collapse
|
63
|
Hildyard JC, Taylor-Brown F, Massey C, Wells DJ, Piercy RJ. Determination of qPCR Reference Genes Suitable for Normalizing Gene Expression in a Canine Model of Duchenne Muscular Dystrophy. J Neuromuscul Dis 2018; 5:177-191. [DOI: 10.3233/jnd-170267] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Affiliation(s)
- John C.W. Hildyard
- Department of Clinical Science and Services, Comparative Neuromuscular Diseases Laboratory, Royal Veterinary College, London, UK
| | - Frances Taylor-Brown
- Department of Clinical Science and Services, Comparative Neuromuscular Diseases Laboratory, Royal Veterinary College, London, UK
| | - Claire Massey
- Department of Clinical Science and Services, Comparative Neuromuscular Diseases Laboratory, Royal Veterinary College, London, UK
| | - Dominic J. Wells
- Department of Comparative Biomedical Sciences, Royal Veterinary College, London, UK
| | - Richard J. Piercy
- Department of Clinical Science and Services, Comparative Neuromuscular Diseases Laboratory, Royal Veterinary College, London, UK
| |
Collapse
|
64
|
Pervin MS, Itoh G, Talukder MSU, Fujimoto K, Morimoto YV, Tanaka M, Ueda M, Yumura S. A study of wound repair in Dictyostelium cells by using novel laserporation. Sci Rep 2018; 8:7969. [PMID: 29789591 PMCID: PMC5964096 DOI: 10.1038/s41598-018-26337-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Accepted: 05/10/2018] [Indexed: 11/09/2022] Open
Abstract
We examined the mechanism of cell membrane repair in Dictyostelium cells by using a novel laser-based cell poration method. The dynamics of wound pores opening and closing were characterized by live imaging of fluorescent cell membrane proteins, influx of fluorescent dye, and Ca2+ imaging. The wound closed within 2-4 sec, depending on the wound size. Cells could tolerate a wound size of less than 2.0 µm. In the absence of Ca2+ in the external medium, the wound pore did not close and cells ruptured. The release of Ca2+ from intracellular stores also contributed to the elevation of cytoplasmic Ca2+ but not to wound repair. Annexin C1 immediately accumulated at the wound site depending on the external Ca2+ concentration, and annexin C1 knockout cells had a defect in wound repair, but it was not essential. Dictyostelium cells were able to respond to multiple repeated wounds with the same time courses, in contrast to previous reports showing that the first wound accelerates the second wound repair in fibroblasts.
Collapse
Affiliation(s)
- Mst Shaela Pervin
- Department of Functional Molecular Biology, Graduate School of Medicine, Yamaguchi University, Yamaguchi, 753-8512, Japan
| | - Go Itoh
- Department of Molecular Medicine and Biochemistry, Akita University Graduate School of Medicine, Akita, 010-8543, Japan
| | - Md Shahabe Uddin Talukder
- Department of Functional Molecular Biology, Graduate School of Medicine, Yamaguchi University, Yamaguchi, 753-8512, Japan.,Institute of Food and Radiation Biology, Atomic Energy Research Establishment, Savar, GPO Box 3787, Dhaka, 1000, Bangladesh
| | - Koushiro Fujimoto
- Department of Functional Molecular Biology, Graduate School of Medicine, Yamaguchi University, Yamaguchi, 753-8512, Japan
| | - Yusuke V Morimoto
- Department of Bioscience and Bioinformatics, Faculty of Computer Science and Systems Engineering, Kyushu Institute of Technology, 680-4 Kawazu, Iizuka, Fukuoka, 820-8502, Japan.,Quantitative Biology Center (QBiC), RIKEN, 6-2-3 Furuedai, Suita, Osaka, 565-0871, Japan
| | - Masamitsu Tanaka
- Department of Molecular Medicine and Biochemistry, Akita University Graduate School of Medicine, Akita, 010-8543, Japan
| | - Masahiro Ueda
- Quantitative Biology Center (QBiC), RIKEN, 6-2-3 Furuedai, Suita, Osaka, 565-0871, Japan.,Graduate School of Frontier Biosciences, Osaka University, Osaka, 565-0871, Japan
| | - Shigehiko Yumura
- Department of Functional Molecular Biology, Graduate School of Medicine, Yamaguchi University, Yamaguchi, 753-8512, Japan.
| |
Collapse
|
65
|
|
66
|
Lee JJA, Maruyama R, Sakurai H, Yokota T. Cell Membrane Repair Assay Using a Two-photon Laser Microscope. J Vis Exp 2018. [PMID: 29364240 DOI: 10.3791/56999] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Numerous pathophysiological insults can cause damage to cell membranes and, when coupled with innate defects in cell membrane repair or integrity, can result in disease. Understanding the underlying molecular mechanisms surrounding cell membrane repair is, therefore, an important objective to the development of novel therapeutic strategies for diseases associated with dysfunctional cell membrane dynamics. Many in vitro and in vivo studies aimed at understanding cell membrane resealing in various disease contexts utilize two-photon laser ablation as a standard for determining functional outcomes following experimental treatments. In this assay, cell membranes are subjected to wounding with a two-photon laser, which causes the cell membrane to rupture and fluorescent dye to infiltrate the cell. The intensity of fluorescence within the cell can then be monitored to quantify the cell's ability to reseal itself. There are several alternative methods for assessing cell membrane response to injury, as well as great variation in the two-photon laser wounding approach itself, therefore, a single, unified model of cell wounding would beneficially serve to decrease the variation between these methodologies. In this article, we outline a simple two-photon laser wounding protocol for assessing cell membrane repair in vitro in both healthy and dysferlinopathy patient fibroblast cells transfected with or without a full-length dysferlin plasmid.
Collapse
Affiliation(s)
- Joshua J A Lee
- Department of Medical Genetics, University of Alberta Faculty of Medicine and Dentistry
| | - Rika Maruyama
- Department of Medical Genetics, University of Alberta Faculty of Medicine and Dentistry
| | | | - Toshifumi Yokota
- Department of Medical Genetics, University of Alberta Faculty of Medicine and Dentistry;
| |
Collapse
|
67
|
Wang J, Liu J, Cao Y, Hu M, Hua Z. Domain IV of Annexin A5 Is Critical for Binding Calcium and Guarantees Its Maximum Binding to the Phosphatidylserine Membrane. Molecules 2017; 22:molecules22122256. [PMID: 29257055 PMCID: PMC6149819 DOI: 10.3390/molecules22122256] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Revised: 12/08/2017] [Accepted: 12/14/2017] [Indexed: 11/16/2022] Open
Abstract
Background: Although domain IV of annexin A5 (anxA5) may be less effective in binding phosphatidylserine (PS), the four domains together may guarantee the maximum binding of anxA5 to the PS membrane. Additionally, previous research has shown that annexin mutants lacking one or more domain(s) have different biological activities compared to the wild-type. The present research mainly aims to study the role of domain IV in the crucial PS-binding function of anxA5. Methods: The domain IV-truncated anxA5 protein was constructed and purified. Isothermal titration calorimetry, flow cytometry and activated partial thromboplastin time were adopted to examine the function of domain IV in anxA5-PS binding directly or indirectly. Results: The domain IV-truncated form of anxA5 is impaired in binding PS liposome and apoptotic cells, and anticoagulation activity. The mutant cannot bind calcium, but binds PS only in the presence of calcium. Conclusions: Truncation of domain IV of anxA5 destroys its calcium-binding ability and impairs its PS-binding activity. Truncation of domain IV may induce conformation change of anxA5 or reduce the hydrophobic interactions between protein and membrane, which may explain the decrease of PS-binding affinity of the mutant.
Collapse
Affiliation(s)
- Jie Wang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, China.
| | - Jing Liu
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, China.
| | - Yulu Cao
- Jiangsu TargetPharma Laboratories Inc., Changzhou High-Tech Research Institute of Nanjing University, Changzhou 213164, China.
| | - Minjin Hu
- Jiangsu TargetPharma Laboratories Inc., Changzhou High-Tech Research Institute of Nanjing University, Changzhou 213164, China.
| | - Zichun Hua
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, China.
- Shenzhen Research Institute of Nanjing University, Shenzhen 518057, China.
| |
Collapse
|
68
|
McElhanon KE, Bhattacharya S. Altered membrane integrity in the progression of muscle diseases. Life Sci 2017; 192:166-172. [PMID: 29183798 DOI: 10.1016/j.lfs.2017.11.035] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Revised: 11/12/2017] [Accepted: 11/22/2017] [Indexed: 12/27/2022]
Abstract
Sarcolemmal integrity is orchestrated through the interplay of preserving membrane strength and fast tracking the membrane repair process during an event of compromised membrane fragility. Several molecular players have been identified that act in a concerted fashion to maintain the barrier function of the muscle membrane. Substantial research findings in the field of muscle biology point out the importance of maintaining membrane integrity as a key contributory factor to cellular homeostasis. Innumerable data on the progression of membrane pathology associated with compromised muscle membrane integrity support targeting sarcolemmal integrity in skeletal and cardiac muscle as a model therapeutic strategy to alleviate some of the pathologic conditions. This review will discuss strategies that researchers have undertaken to compensate for an imbalance in sarcolemma membrane fragility and membrane repair to maintain muscle membrane integrity.
Collapse
Affiliation(s)
- Kevin E McElhanon
- Department of Physiology and Cell Biology, Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, 473 W. 12th Ave, Columbus, OH 43210-1252, United States
| | - Sayak Bhattacharya
- Department of Physiology and Cell Biology, Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, 473 W. 12th Ave, Columbus, OH 43210-1252, United States.
| |
Collapse
|
69
|
Annexin A4 and A6 induce membrane curvature and constriction during cell membrane repair. Nat Commun 2017; 8:1623. [PMID: 29158488 PMCID: PMC5696365 DOI: 10.1038/s41467-017-01743-6] [Citation(s) in RCA: 119] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Accepted: 10/12/2017] [Indexed: 11/12/2022] Open
Abstract
Efficient cell membrane repair mechanisms are essential for maintaining membrane integrity and thus for cell life. Here we show that the Ca2+- and phospholipid-binding proteins annexin A4 and A6 are involved in plasma membrane repair and needed for rapid closure of micron-size holes. We demonstrate that annexin A4 binds to artificial membranes and generates curvature force initiated from free edges, whereas annexin A6 induces constriction force. In cells, plasma membrane injury and Ca2+ influx recruit annexin A4 to the vicinity of membrane wound edges where its homo-trimerization leads to membrane curvature near the edges. We propose that curvature force is utilized together with annexin A6-mediated constriction force to pull the wound edges together for eventual fusion. We show that annexin A4 can counteract various plasma membrane disruptions including holes of several micrometers indicating that induction of curvature force around wound edges is an early key event in cell membrane repair. The role of annexins in cell membrane repair is largely undefined. Here the authors use a model lipid bilayer to show that annexin A4 induces curvature at the membrane free edge and annexin A6 induces constriction force, and find that both annexins are recruited to wound edges in cells and are required for repair.
Collapse
|
70
|
Giacomazzi G, Holvoet B, Trenson S, Caluwé E, Kravic B, Grosemans H, Cortés-Calabuig Á, Deroose CM, Huylebroeck D, Hashemolhosseini S, Janssens S, McNally E, Quattrocelli M, Sampaolesi M. MicroRNAs promote skeletal muscle differentiation of mesodermal iPSC-derived progenitors. Nat Commun 2017; 8:1249. [PMID: 29093487 PMCID: PMC5665910 DOI: 10.1038/s41467-017-01359-w] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2016] [Accepted: 09/12/2017] [Indexed: 02/06/2023] Open
Abstract
Muscular dystrophies (MDs) are often characterized by impairment of both skeletal and cardiac muscle. Regenerative strategies for both compartments therefore constitute a therapeutic avenue. Mesodermal iPSC-derived progenitors (MiPs) can regenerate both striated muscle types simultaneously in mice. Importantly, MiP myogenic propensity is influenced by somatic lineage retention. However, it is still unknown whether human MiPs have in vivo potential. Furthermore, methods to enhance the intrinsic myogenic properties of MiPs are likely needed, given the scope and need to correct large amounts of muscle in the MDs. Here, we document that human MiPs can successfully engraft into the skeletal muscle and hearts of dystrophic mice. Utilizing non-invasive live imaging and selectively induced apoptosis, we report evidence of striated muscle regeneration in vivo in mice by human MiPs. Finally, combining RNA-seq and miRNA-seq data, we define miRNA cocktails that promote the myogenic potential of human MiPs.
Collapse
Affiliation(s)
- Giorgia Giacomazzi
- Translational Cardiomyology, Department of Development and Regeneration, KU Leuven, 3000, Leuven, Belgium
| | - Bryan Holvoet
- Department of Imaging and Pathology, Nuclear Medicine and Molecular Imaging, KU Leuven, 3000, Leuven, Belgium
| | - Sander Trenson
- Department of Cardiovascular Sciences, KU Leuven, 3000, Leuven, Belgium
| | - Ellen Caluwé
- Department of Cardiovascular Sciences, KU Leuven, 3000, Leuven, Belgium
| | - Bojana Kravic
- Institute of Biochemistry, Friedrich-Alexander University of Erlangen-Nürnberg, 91054, Erlangen, Germany
| | - Hanne Grosemans
- Translational Cardiomyology, Department of Development and Regeneration, KU Leuven, 3000, Leuven, Belgium
| | | | - Christophe M Deroose
- Department of Imaging and Pathology, Nuclear Medicine and Molecular Imaging, KU Leuven, 3000, Leuven, Belgium
| | - Danny Huylebroeck
- Department of Cell Biology, Erasmus MC, 3015 CN, Rotterdam, The Netherlands.,Laboratory of Molecular Biology (Celgen), Department of Development and Regeneration, KU Leuven, 3000, Leuven, Belgium
| | - Said Hashemolhosseini
- Institute of Biochemistry, Friedrich-Alexander University of Erlangen-Nürnberg, 91054, Erlangen, Germany
| | - Stefan Janssens
- Department of Cardiovascular Sciences, KU Leuven, 3000, Leuven, Belgium
| | - Elizabeth McNally
- Center for Genetic Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Mattia Quattrocelli
- Translational Cardiomyology, Department of Development and Regeneration, KU Leuven, 3000, Leuven, Belgium.,Center for Genetic Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Maurilio Sampaolesi
- Translational Cardiomyology, Department of Development and Regeneration, KU Leuven, 3000, Leuven, Belgium. .,Human Anatomy Unit, Department of Public Health, Experimental and Forensic Medicine, University of Pavia, Pavia, 27100, Italy.
| |
Collapse
|
71
|
Defour A, Medikayala S, Van der Meulen JH, Hogarth MW, Holdreith N, Malatras A, Duddy W, Boehler J, Nagaraju K, Jaiswal JK. Annexin A2 links poor myofiber repair with inflammation and adipogenic replacement of the injured muscle. Hum Mol Genet 2017; 26:1979-1991. [PMID: 28334824 DOI: 10.1093/hmg/ddx065] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Accepted: 02/17/2017] [Indexed: 01/12/2023] Open
Abstract
Repair of skeletal muscle after sarcolemmal damage involves dysferlin and dysferlin-interacting proteins such as annexins. Mice and patient lacking dysferlin exhibit chronic muscle inflammation and adipogenic replacement of the myofibers. Here, we show that similar to dysferlin, lack of annexin A2 (AnxA2) also results in poor myofiber repair and progressive muscle weakening with age. By longitudinal analysis of AnxA2-deficient muscle we find that poor myofiber repair due to the lack of AnxA2 does not result in chronic inflammation or adipogenic replacement of the myofibers. Further, deletion of AnxA2 in dysferlin deficient mice reduced muscle inflammation, adipogenic replacement of myofibers, and improved muscle function. These results identify multiple roles of AnxA2 in muscle repair, which includes facilitating myofiber repair, chronic muscle inflammation and adipogenic replacement of dysferlinopathic muscle. It also identifies inhibition of AnxA2-mediated inflammation as a novel therapeutic avenue for treating muscle loss in dysferlinopathy.
Collapse
Affiliation(s)
- Aurelia Defour
- Center for Genetic Medicine Research, Children's National Health System, Washington, DC 20010, USA
| | - Sushma Medikayala
- Center for Genetic Medicine Research, Children's National Health System, Washington, DC 20010, USA
| | - Jack H Van der Meulen
- Center for Genetic Medicine Research, Children's National Health System, Washington, DC 20010, USA
| | - Marshall W Hogarth
- Center for Genetic Medicine Research, Children's National Health System, Washington, DC 20010, USA
| | - Nicholas Holdreith
- Center for Genetic Medicine Research, Children's National Health System, Washington, DC 20010, USA
| | - Apostolos Malatras
- Center for Research in Myology 75013, Sorbonne Universités, UPMC University Paris 06, INSERM UMRS975, CNRS FRE3617, GH Pitié Salpêtrière, Paris 13, Paris, France
| | - William Duddy
- Center for Research in Myology 75013, Sorbonne Universités, UPMC University Paris 06, INSERM UMRS975, CNRS FRE3617, GH Pitié Salpêtrière, Paris 13, Paris, France
- Northern Ireland Centre for Stratified Medicine, Altnagelvin Hospital Campus, Ulster University, Londonderry, Northern Ireland, BT52 1SJ UK
| | - Jessica Boehler
- Center for Genetic Medicine Research, Children's National Health System, Washington, DC 20010, USA
| | - Kanneboyina Nagaraju
- Center for Genetic Medicine Research, Children's National Health System, Washington, DC 20010, USA
- Department of Integrative Systems Biology, George Washington University School of Medicine and Health Sciences, Washington, DC, 20052 USA
| | - Jyoti K Jaiswal
- Center for Genetic Medicine Research, Children's National Health System, Washington, DC 20010, USA
- Department of Integrative Systems Biology, George Washington University School of Medicine and Health Sciences, Washington, DC, 20052 USA
| |
Collapse
|
72
|
Quattrocelli M, Capote J, Ohiri JC, Warner JL, Vo AH, Earley JU, Hadhazy M, Demonbreun AR, Spencer MJ, McNally EM. Genetic modifiers of muscular dystrophy act on sarcolemmal resealing and recovery from injury. PLoS Genet 2017; 13:e1007070. [PMID: 29065150 PMCID: PMC5669489 DOI: 10.1371/journal.pgen.1007070] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 11/03/2017] [Accepted: 10/11/2017] [Indexed: 12/17/2022] Open
Abstract
Genetic disruption of the dystrophin complex produces muscular dystrophy characterized by a fragile muscle plasma membrane leading to excessive muscle degeneration. Two genetic modifiers of Duchenne Muscular Dystrophy implicate the transforming growth factor β (TGFβ) pathway, osteopontin encoded by the SPP1 gene and latent TGFβ binding protein 4 (LTBP4). We now evaluated the functional effect of these modifiers in the context of muscle injury and repair to elucidate their mechanisms of action. We found that excess osteopontin exacerbated sarcolemmal injury, and correspondingly, that loss of osteopontin reduced injury extent both in isolated myofibers and in muscle in vivo. We found that ablation of osteopontin was associated with reduced expression of TGFβ and TGFβ-associated pathways. We identified that increased TGFβ resulted in reduced expression of Anxa1 and Anxa6, genes encoding key components of the muscle sarcolemma resealing process. Genetic manipulation of Ltbp4 in dystrophic muscle also directly modulated sarcolemmal resealing, and Ltbp4 alleles acted in concert with Anxa6, a distinct modifier of muscular dystrophy. These data provide a model in which a feed forward loop of TGFβ and osteopontin directly impacts the capacity of muscle to recover from injury, and identifies an intersection of genetic modifiers on muscular dystrophy.
Collapse
MESH Headings
- Animals
- Annexin A1/genetics
- Annexin A1/metabolism
- Annexin A6/genetics
- Annexin A6/metabolism
- Female
- Gene Expression Regulation
- Genes, Modifier
- Latent TGF-beta Binding Proteins/physiology
- Male
- Mice
- Mice, Inbred DBA
- Mice, Knockout
- Muscle, Skeletal/injuries
- Muscle, Skeletal/physiology
- Muscular Dystrophy, Animal/genetics
- Muscular Dystrophy, Animal/metabolism
- Muscular Dystrophy, Animal/pathology
- Osteopontin/genetics
- Osteopontin/metabolism
- Receptors, Transforming Growth Factor beta/genetics
- Receptors, Transforming Growth Factor beta/metabolism
- Recovery of Function
- Sarcolemma/physiology
Collapse
Affiliation(s)
- Mattia Quattrocelli
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States of America
| | - Joanna Capote
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, California, United States of America
| | - Joyce C. Ohiri
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States of America
| | - James L. Warner
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States of America
| | - Andy H. Vo
- Committee on Development, Regeneration, and Stem Cell Biology, The University of Chicago, Chicago, Illinois, United States of America
| | - Judy U. Earley
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States of America
| | - Michele Hadhazy
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States of America
| | - Alexis R. Demonbreun
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States of America
| | - Melissa J. Spencer
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, California, United States of America
| | - Elizabeth M. McNally
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States of America
| |
Collapse
|
73
|
Tsuda T, Fitzgerald KK. Dystrophic Cardiomyopathy: Complex Pathobiological Processes to Generate Clinical Phenotype. J Cardiovasc Dev Dis 2017; 4:jcdd4030014. [PMID: 29367543 PMCID: PMC5715712 DOI: 10.3390/jcdd4030014] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Revised: 08/27/2017] [Accepted: 08/30/2017] [Indexed: 02/06/2023] Open
Abstract
Duchenne muscular dystrophy (DMD), Becker muscular dystrophy (BMD), and X-linked dilated cardiomyopathy (XL-DCM) consist of a unique clinical entity, the dystrophinopathies, which are due to variable mutations in the dystrophin gene. Dilated cardiomyopathy (DCM) is a common complication of dystrophinopathies, but the onset, progression, and severity of heart disease differ among these subgroups. Extensive molecular genetic studies have been conducted to assess genotype-phenotype correlation in DMD, BMD, and XL-DCM to understand the underlying mechanisms of these diseases, but the results are not always conclusive, suggesting the involvement of complex multi-layers of pathological processes that generate the final clinical phenotype. Dystrophin protein is a part of dystrophin-glycoprotein complex (DGC) that is localized in skeletal muscles, myocardium, smooth muscles, and neuronal tissues. Diversity of cardiac phenotype in dystrophinopathies suggests multiple layers of pathogenetic mechanisms in forming dystrophic cardiomyopathy. In this review article, we review the complex molecular interactions involving the pathogenesis of dystrophic cardiomyopathy, including primary gene mutations and loss of structural integrity, secondary cellular responses, and certain epigenetic and other factors that modulate gene expressions. Involvement of epigenetic gene regulation appears to lead to specific cardiac phenotypes in dystrophic hearts.
Collapse
Affiliation(s)
- Takeshi Tsuda
- Nemours Cardiac Center, Nemours/Alfred I. duPont Hospital for Children, Wilmington, 1600 Rockland Rd, DE 19803, USA.
| | - Kristi K Fitzgerald
- Nemours Cardiac Center, Nemours/Alfred I. duPont Hospital for Children, Wilmington, 1600 Rockland Rd, DE 19803, USA.
| |
Collapse
|
74
|
Quattrocelli M, Salamone IM, Page PG, Warner JL, Demonbreun AR, McNally EM. Intermittent Glucocorticoid Dosing Improves Muscle Repair and Function in Mice with Limb-Girdle Muscular Dystrophy. THE AMERICAN JOURNAL OF PATHOLOGY 2017; 187:2520-2535. [PMID: 28823869 DOI: 10.1016/j.ajpath.2017.07.017] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Revised: 07/03/2017] [Accepted: 07/13/2017] [Indexed: 12/17/2022]
Abstract
The muscular dystrophies are genetically diverse. Shared pathological features among muscular dystrophies include breakdown, or loss of muscle, and accompanying fibrotic replacement. Novel strategies are needed to enhance muscle repair and function and to slow this pathological remodeling. Glucocorticoid steroids, like prednisone, are known to delay loss of ambulation in patients with Duchenne muscular dystrophy but are accompanied by prominent adverse effects. However, less is known about the effects of steroid administration in other types of muscular dystrophies, including limb-girdle muscular dystrophies (LGMDs). LGMD 2B is caused by loss of dysferlin, a membrane repair protein, and LGMD 2C is caused by loss of the dystrophin-associated protein, γ-sarcoglycan. Herein, we assessed the efficacy of steroid dosing on sarcolemmal repair, muscle function, histopathology, and the regenerative capacity of primary muscle cells. We found that in murine models of LGMD 2B and 2C, daily prednisone dosing reduced muscle damage and fibroinflammatory infiltration. However, daily prednisone dosing also correlated with increased muscle adipogenesis and atrophic remodeling. Conversely, intermittent dosing of prednisone, provided once weekly, enhanced muscle repair and did not induce atrophy or adipogenesis, and was associated with improved muscle function. These data indicate that dosing frequency of glucocorticoid steroids affects muscle remodeling in non-Duchenne muscular dystrophies, suggesting a positive outcome associated with intermittent steroid dosing in LGMD 2B and 2C muscle.
Collapse
Affiliation(s)
- Mattia Quattrocelli
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Isabella M Salamone
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Patrick G Page
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - James L Warner
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Alexis R Demonbreun
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Elizabeth M McNally
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois.
| |
Collapse
|
75
|
Plasma membrane repair: the adaptable cell life-insurance. Curr Opin Cell Biol 2017; 47:99-107. [DOI: 10.1016/j.ceb.2017.03.011] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Accepted: 03/16/2017] [Indexed: 12/17/2022]
|
76
|
Grewal T, Wason SJ, Enrich C, Rentero C. Annexins - insights from knockout mice. Biol Chem 2017; 397:1031-53. [PMID: 27318360 DOI: 10.1515/hsz-2016-0168] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Accepted: 06/14/2016] [Indexed: 12/23/2022]
Abstract
Annexins are a highly conserved protein family that bind to phospholipids in a calcium (Ca2+) - dependent manner. Studies with purified annexins, as well as overexpression and knockdown approaches identified multiple functions predominantly linked to their dynamic and reversible membrane binding behavior. However, most annexins are found at multiple locations and interact with numerous proteins. Furthermore, similar membrane binding characteristics, overlapping localizations and shared interaction partners have complicated identification of their precise functions. To gain insight into annexin function in vivo, mouse models deficient of annexin A1 (AnxA1), A2, A4, A5, A6 and A7 have been generated. Interestingly, with the exception of one study, all mice strains lacking one or even two annexins are viable and develop normally. This suggested redundancy within annexins, but examining these knockout (KO) strains under stress conditions revealed striking phenotypes, identifying underlying mechanisms specific for individual annexins, often supporting Ca2+ homeostasis and membrane transport as central for annexin biology. Conversely, mice lacking AnxA1 or A2 show extracellular functions relevant in health and disease that appear independent of membrane trafficking or Ca2+ signaling. This review will summarize the mechanistic insights gained from studies utilizing mouse models lacking members of the annexin family.
Collapse
|
77
|
Quattrocelli M, Barefield DY, Warner JL, Vo AH, Hadhazy M, Earley JU, Demonbreun AR, McNally EM. Intermittent glucocorticoid steroid dosing enhances muscle repair without eliciting muscle atrophy. J Clin Invest 2017; 127:2418-2432. [PMID: 28481224 DOI: 10.1172/jci91445] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Accepted: 03/09/2017] [Indexed: 12/20/2022] Open
Abstract
Glucocorticoid steroids such as prednisone are prescribed for chronic muscle conditions such as Duchenne muscular dystrophy, where their use is associated with prolonged ambulation. The positive effects of chronic steroid treatment in muscular dystrophy are paradoxical because these steroids are also known to trigger muscle atrophy. Chronic steroid use usually involves once-daily dosing, although weekly dosing in children has been suggested for its reduced side effects on behavior. In this work, we tested steroid dosing in mice and found that a single pulse of glucocorticoid steroids improved sarcolemmal repair through increased expression of annexins A1 and A6, which mediate myofiber repair. This increased expression was dependent on glucocorticoid response elements upstream of annexins and was reinforced by the expression of forkhead box O1 (FOXO1). We compared weekly versus daily steroid treatment in mouse models of acute muscle injury and in muscular dystrophy and determined that both regimens provided comparable benefits in terms of annexin gene expression and muscle repair. However, daily dosing activated atrophic pathways, including F-box protein 32 (Fbxo32), which encodes atrogin-1. Conversely, weekly steroid treatment in mdx mice improved muscle function and histopathology and concomitantly induced the ergogenic transcription factor Krüppel-like factor 15 (Klf15) while decreasing Fbxo32. These findings suggest that intermittent, rather than daily, glucocorticoid steroid regimen promotes sarcolemmal repair and muscle recovery from injury while limiting atrophic remodeling.
Collapse
|
78
|
Konopka-Postupolska D, Clark G. Annexins as Overlooked Regulators of Membrane Trafficking in Plant Cells. Int J Mol Sci 2017; 18:E863. [PMID: 28422051 PMCID: PMC5412444 DOI: 10.3390/ijms18040863] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2016] [Revised: 04/03/2017] [Accepted: 04/06/2017] [Indexed: 12/11/2022] Open
Abstract
Annexins are an evolutionary conserved superfamily of proteins able to bind membrane phospholipids in a calcium-dependent manner. Their physiological roles are still being intensively examined and it seems that, despite their general structural similarity, individual proteins are specialized toward specific functions. However, due to their general ability to coordinate membranes in a calcium-sensitive fashion they are thought to participate in membrane flow. In this review, we present a summary of the current understanding of cellular transport in plant cells and consider the possible roles of annexins in different stages of vesicular transport.
Collapse
Affiliation(s)
- Dorota Konopka-Postupolska
- Plant Biochemistry Department, Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw 02-106, Poland.
| | - Greg Clark
- Molecular, Cell, and Developmental Biology, University of Texas, Austin, TX 78712, USA.
| |
Collapse
|
79
|
Shah A, Schiffmacher AT, Taneyhill LA. Annexin A6 controls neuronal membrane dynamics throughout chick cranial sensory gangliogenesis. Dev Biol 2017; 425:85-99. [PMID: 28315296 DOI: 10.1016/j.ydbio.2017.03.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Revised: 03/10/2017] [Accepted: 03/11/2017] [Indexed: 12/19/2022]
Abstract
Cranial sensory ganglia are components of the peripheral nervous system that possess a significant somatosensory role and include neurons within the trigeminal and epibranchial nerve bundles. Although it is well established that these ganglia arise from interactions between neural crest and neurogenic placode cells, the molecular basis of ganglia assembly is still poorly understood. Members of the Annexin protein superfamily play key roles in sensory nervous system development throughout metazoans. Annexin A6 is expressed in chick trigeminal and epibranchial placode cell-derived neuroblasts and neurons, but its function in cranial ganglia formation has not been elucidated. To this end, we interrogated the role of Annexin A6 using gene perturbation studies in the chick embryo. Our data reveal that placode cell-derived neuroblasts with reduced Annexin A6 levels ingress and migrate normally to the ganglionic anlage, where neural crest cell corridors correctly form around them. Strikingly, while Annexin A6-depleted placode cell-derived neurons still express mature neuronal markers, they fail to form two long processes, which are considered morphological features of mature neurons, and no longer innervate their designated targets due to the absence of this bipolar morphology. Moreover, overexpression of Annexin A6 causes some placode cell-derived neurons to form extra protrusions alongside these bipolar processes. These data demonstrate that the molecular program associated with neuronal maturation is distinct from that orchestrating changes in neuronal morphology, and, importantly, reveal Annexin A6 to be a key membrane scaffolding protein during sensory neuron membrane biogenesis. Collectively, our results provide novel insight into mechanisms underscoring morphological changes within placode cell-derived neurons that are essential for cranial gangliogenesis.
Collapse
Affiliation(s)
- Ankita Shah
- Department of Animal and Avian Sciences, University of Maryland, College Park, MD 20742, USA
| | - Andrew T Schiffmacher
- Department of Animal and Avian Sciences, University of Maryland, College Park, MD 20742, USA
| | - Lisa A Taneyhill
- Department of Animal and Avian Sciences, University of Maryland, College Park, MD 20742, USA.
| |
Collapse
|
80
|
Quattrocelli M, Spencer MJ, McNally EM. Outside in: The matrix as a modifier of muscular dystrophy. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2017; 1864:572-579. [PMID: 28011285 PMCID: PMC5262521 DOI: 10.1016/j.bbamcr.2016.12.020] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Revised: 12/14/2016] [Accepted: 12/19/2016] [Indexed: 02/07/2023]
Abstract
Muscular dystrophies are genetic conditions leading to muscle degeneration and often, impaired regeneration. Duchenne Muscular Dystrophy is a prototypical form of muscular dystrophy, and like other forms of genetically inherited muscle diseases, pathological progression is variable. Variability in muscular dystrophy can arise from differences in the manner in which the primary mutation impacts the affected protein's function; however, clinical heterogeneity also derives from secondary mutations in other genes that can enhance or reduce pathogenic features of disease. These genes, called genetic modifiers, regulate the pathophysiological context of dystrophic degeneration and regeneration. Understanding the mechanistic links between genetic modifiers and dystrophic progression sheds light on pathologic remodeling, and provides novel avenues to therapeutically intervene to reduce muscle degeneration. Based on targeted genetic approaches and unbiased genomewide screens, several modifiers have been identified for muscular dystrophy, including extracellular agonists of signaling cascades. This review will focus on identification and possible mechanisms of recently identified modifiers for muscular dystrophy, including osteopontin, latent TGFβ binding protein 4 (LTBP4) and Jagged1. Moreover, we will review the investigational approaches that aim to target modifier pathways and thereby counteract dystrophic muscle wasting.
Collapse
Affiliation(s)
| | - Melissa J Spencer
- Dept of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | | |
Collapse
|
81
|
Grewal T, Hoque M, Conway JRW, Reverter M, Wahba M, Beevi SS, Timpson P, Enrich C, Rentero C. Annexin A6-A multifunctional scaffold in cell motility. Cell Adh Migr 2017; 11:288-304. [PMID: 28060548 DOI: 10.1080/19336918.2016.1268318] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Annexin A6 (AnxA6) belongs to a highly conserved protein family characterized by their calcium (Ca2+)-dependent binding to phospholipids. Over the years, immunohistochemistry, subcellular fractionations, and live cell microscopy established that AnxA6 is predominantly found at the plasma membrane and endosomal compartments. In these locations, AnxA6 acts as a multifunctional scaffold protein, recruiting signaling proteins, modulating cholesterol and membrane transport and influencing actin dynamics. These activities enable AnxA6 to contribute to the formation of multifactorial protein complexes and membrane domains relevant in signal transduction, cholesterol homeostasis and endo-/exocytic membrane transport. Hence, AnxA6 has been implicated in many biological processes, including cell proliferation, survival, differentiation, inflammation, but also membrane repair and viral infection. More recently, we and others identified roles for AnxA6 in cancer cell migration and invasion. This review will discuss how the multiple scaffold functions may enable AnxA6 to modulate migratory cell behavior in health and disease.
Collapse
Affiliation(s)
- Thomas Grewal
- a Faculty of Pharmacy , University of Sydney , Sydney , NSW , Australia
| | - Monira Hoque
- a Faculty of Pharmacy , University of Sydney , Sydney , NSW , Australia
| | - James R W Conway
- b The Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Cancer Division, St Vincent's Clinical School, Faculty of Medicine , University of New South Wales , Sydney , NSW , Australia
| | - Meritxell Reverter
- c Departament de Biomedicina, Unitat de Biologia Cel·lular, Centre de Recerca Biomèdica CELLEX, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Facultat de Medicina , Universitat de Barcelona , Barcelona , Spain
| | - Mohamed Wahba
- a Faculty of Pharmacy , University of Sydney , Sydney , NSW , Australia
| | - Syed S Beevi
- a Faculty of Pharmacy , University of Sydney , Sydney , NSW , Australia
| | - Paul Timpson
- b The Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Cancer Division, St Vincent's Clinical School, Faculty of Medicine , University of New South Wales , Sydney , NSW , Australia
| | - Carlos Enrich
- c Departament de Biomedicina, Unitat de Biologia Cel·lular, Centre de Recerca Biomèdica CELLEX, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Facultat de Medicina , Universitat de Barcelona , Barcelona , Spain
| | - Carles Rentero
- c Departament de Biomedicina, Unitat de Biologia Cel·lular, Centre de Recerca Biomèdica CELLEX, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Facultat de Medicina , Universitat de Barcelona , Barcelona , Spain
| |
Collapse
|
82
|
Enrich C, Rentero C, Meneses-Salas E, Tebar F, Grewal T. Annexins: Ca 2+ Effectors Determining Membrane Trafficking in the Late Endocytic Compartment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 981:351-385. [PMID: 29594868 DOI: 10.1007/978-3-319-55858-5_14] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Despite the discovery of annexins 40 years ago, we are just beginning to understand some of the functions of these still enigmatic proteins. Defined and characterized by their ability to bind anionic membrane lipids in a Ca2+-dependent manner, each annexin has to be considered a multifunctional protein, with a multitude of cellular locations and diverse activities. Underlying causes for this considerable functional diversity include their capability to associate with multiple cytosolic and membrane proteins. In recent years, the increasingly recognized establishment of membrane contact sites between subcellular compartments opens a new scenario for annexins as instrumental players to link Ca2+ signalling with the integration of membrane trafficking in many facets of cell physiology. In this chapter, we review and discuss current knowledge on the contribution of annexins in the biogenesis and functioning of the late endocytic compartment, affecting endo- and exocytic pathways in a variety of physiological consequences ranging from membrane repair, lysosomal exocytosis, to cell migration.
Collapse
Affiliation(s)
- Carlos Enrich
- Departament de Biomedicina, Unitat de Biologia Cel·lular, Centre de Recerca Biomèdica (CELLEX), Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain. .,Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Barcelona, Spain.
| | - Carles Rentero
- Departament de Biomedicina, Unitat de Biologia Cel·lular, Centre de Recerca Biomèdica (CELLEX), Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Barcelona, Spain
| | - Elsa Meneses-Salas
- Departament de Biomedicina, Unitat de Biologia Cel·lular, Centre de Recerca Biomèdica (CELLEX), Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Barcelona, Spain
| | - Francesc Tebar
- Departament de Biomedicina, Unitat de Biologia Cel·lular, Centre de Recerca Biomèdica (CELLEX), Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Barcelona, Spain
| | - Thomas Grewal
- Faculty of Pharmacy, University of Sydney, Sydney, Australia
| |
Collapse
|
83
|
Milani-Nejad N, Schultz EJ, Slabaugh JL, Janssen PML, Rafael-Fortney JA. Myocardial Contractile Dysfunction Is Present without Histopathology in a Mouse Model of Limb-Girdle Muscular Dystrophy-2F and Is Prevented after Claudin-5 Virotherapy. Front Physiol 2016; 7:539. [PMID: 27999547 PMCID: PMC5138189 DOI: 10.3389/fphys.2016.00539] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Accepted: 10/27/2016] [Indexed: 01/18/2023] Open
Abstract
Mutations in several members of the dystrophin glycoprotein complex lead to skeletal and cardiomyopathies. Cardiac care for these muscular dystrophies consists of management of symptoms with standard heart medications after detection of reduced whole heart function. Recent evidence from both Duchenne muscular dystrophy patients and animal models suggests that myocardial dysfunction is present before myocardial damage or deficiencies in whole heart function, and that treatment prior to heart failure symptoms may be beneficial. To determine whether this same early myocardial dysfunction is present in other muscular dystrophy cardiomyopathies, we conducted a physiological assessment of cardiac function at the tissue level in the δ-sarcoglycan null mouse model (Sgcd−/−) of Limb-girdle muscular dystrophy type 2F. Baseline cardiac contractile force measurements using ex vivo intact linear muscle preparations, were severely depressed in these mice without the presence of histopathology. Virotherapy withclaudin-5 prevents the onset of cardiomyopathy in another muscular dystrophy model. After virotherapy with claudin-5, the cardiac contractile force deficits in Sgcd−/− mice are no longer significant. These studies suggest that screening Limb-girdle muscular dystrophy patients using methods that detect earlier functional changes may provide a longer therapeutic window for cardiac care.
Collapse
Affiliation(s)
- Nima Milani-Nejad
- Department of Physiology and Cell Biology, The Ohio State University Wexner Medical CenterColumbus, OH, USA; Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical CenterColumbus, OH, USA; Medical Scientist Training Program and Biomedical Sciences Graduate Program, The Ohio State University Wexner Medical CenterColumbus, OH, USA
| | - Eric J Schultz
- Department of Physiology and Cell Biology, The Ohio State University Wexner Medical CenterColumbus, OH, USA; Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical CenterColumbus, OH, USA; Department of Molecular and Cellular Biochemistry, The Ohio State University Wexner Medical CenterColumbus, OH, USA
| | - Jessica L Slabaugh
- Department of Physiology and Cell Biology, The Ohio State University Wexner Medical CenterColumbus, OH, USA; Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical CenterColumbus, OH, USA
| | - Paul M L Janssen
- Department of Physiology and Cell Biology, The Ohio State University Wexner Medical CenterColumbus, OH, USA; Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical CenterColumbus, OH, USA
| | - Jill A Rafael-Fortney
- Department of Physiology and Cell Biology, The Ohio State University Wexner Medical CenterColumbus, OH, USA; Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical CenterColumbus, OH, USA; Department of Molecular and Cellular Biochemistry, The Ohio State University Wexner Medical CenterColumbus, OH, USA
| |
Collapse
|
84
|
Whitlock JM, Hartzell HC. Anoctamins/TMEM16 Proteins: Chloride Channels Flirting with Lipids and Extracellular Vesicles. Annu Rev Physiol 2016; 79:119-143. [PMID: 27860832 DOI: 10.1146/annurev-physiol-022516-034031] [Citation(s) in RCA: 122] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Anoctamin (ANO)/TMEM16 proteins exhibit diverse functions in cells throughout the body and are implicated in several human diseases. Although the founding members ANO1 (TMEM16A) and ANO2 (TMEM16B) are Ca2+-activated Cl- channels, most ANO paralogs are Ca2+-dependent phospholipid scramblases that serve as channels facilitating the movement (scrambling) of phospholipids between leaflets of the membrane bilayer. Phospholipid scrambling significantly alters the physical properties of the membrane and its landscape and has vast downstream signaling consequences. In particular, phosphatidylserine exposed on the external leaflet of the plasma membrane functions as a ligand for receptors vital for cell-cell communication. A major consequence of Ca2+-dependent scrambling is the release of extracellular vesicles that function as intercellular messengers by delivering signaling proteins and noncoding RNAs to alter target cell function. We discuss the physiological implications of Ca2+-dependent phospholipid scrambling, the extracellular vesicles associated with this activity, and the roles of ANOs in these processes.
Collapse
Affiliation(s)
- Jarred M Whitlock
- Department of Cell Biology, Emory University School of Medicine, Atlanta, Georgia 30322;
| | - H Criss Hartzell
- Department of Cell Biology, Emory University School of Medicine, Atlanta, Georgia 30322;
| |
Collapse
|
85
|
Blazek AD, Paleo BJ, Weisleder N. Plasma Membrane Repair: A Central Process for Maintaining Cellular Homeostasis. Physiology (Bethesda) 2016; 30:438-48. [PMID: 26525343 DOI: 10.1152/physiol.00019.2015] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Plasma membrane repair is a conserved cellular response mediating active resealing of membrane disruptions to maintain homeostasis and prevent cell death and progression of multiple diseases. Cell membrane repair repurposes mechanisms from various cellular functions, including vesicle trafficking, exocytosis, and endocytosis, to mend the broken membrane. Recent studies increased our understanding of membrane repair by establishing the molecular machinery contributing to membrane resealing. Here, we review some of the key proteins linked to cell membrane repair.
Collapse
Affiliation(s)
- Alisa D Blazek
- Department of Physiology and Cell Biology, Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Brian J Paleo
- Department of Physiology and Cell Biology, Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Noah Weisleder
- Department of Physiology and Cell Biology, Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, Ohio
| |
Collapse
|
86
|
Boye TL, Nylandsted J. Annexins in plasma membrane repair. Biol Chem 2016; 397:961-9. [DOI: 10.1515/hsz-2016-0171] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Accepted: 06/14/2016] [Indexed: 01/01/2023]
Abstract
Abstract
Disruption of the plasma membrane poses deadly threat to eukaryotic cells and survival requires a rapid membrane repair system. Recent evidence reveal various plasma membrane repair mechanisms, which are required for cells to cope with membrane lesions including membrane fusion and replacement strategies, remodeling of cortical actin cytoskeleton and vesicle wound patching. Members of the annexin protein family, which are Ca2+-triggered phospholipid-binding proteins emerge as important components of the plasma membrane repair system. Here, we discuss the mechanisms of plasma membrane repair involving annexins spanning from yeast to human cancer cells.
Collapse
|
87
|
Dongsheng H, Zhuo Z, Jiamin L, Hailan M, Lijuan H, Fan C, Dan Y, He Z, Yun X. Proteomic Analysis of the Peri-Infarct Area after Human Umbilical Cord Mesenchymal Stem Cell Transplantation in Experimental Stroke. Aging Dis 2016; 7:623-634. [PMID: 27699085 PMCID: PMC5036957 DOI: 10.14336/ad.2016.0121] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2015] [Accepted: 01/21/2016] [Indexed: 12/27/2022] Open
Abstract
Among various therapeutic approaches for stroke, treatment with human umbilical cord mesenchymal stem cells (hUC-MSCs) has acquired some promising results. However, the underlying mechanisms remain unclear. We analyzed the protein expression spectrum of the cortical peri-infarction region after ischemic stroke followed by treatment with hUC-MSCs, and found 16 proteins expressed differentially between groups treated with or without hUC-MSCs. These proteins were further determined by Gene Ontology term analysis and network with CD200-CD200R1, CCL21-CXCR3 and transcription factors. Three of them: Abca13, Grb2 and Ptgds were verified by qPCR and ELISA. We found the protein level of Abca13 and the mRNA level of Grb2 consistent with results from the proteomic analysis. Finally, the function of these proteins was described and the potential proteins that deserve to be further studied was also highlighted. Our data may provide possible underlying mechanisms for the treatment of stroke using hUC-MSCs.
Collapse
Affiliation(s)
- He Dongsheng
- 1Department of Neurology, Affiliated Drum Tower Hospital, and; 2Jiangsu Key Laboratory for Molecular Medicine, Nanjing University Medical School, Nanjing 210008, China.; 3The State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing 210008, China; 5Jiangsu Province Stroke Center for Diagnosis and Therapy, Nanjing 210008, China; 6Nanjing Neuropsychiatry Clinic Medical Center, Nanjing 210008, China
| | - Zhang Zhuo
- 4Department of Gastroenterology, Children's Hospital of Nanjing, Nanjing Medical University, Nanjing 210008, China
| | - Lao Jiamin
- 1Department of Neurology, Affiliated Drum Tower Hospital, and; 2Jiangsu Key Laboratory for Molecular Medicine, Nanjing University Medical School, Nanjing 210008, China.; 3The State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing 210008, China; 5Jiangsu Province Stroke Center for Diagnosis and Therapy, Nanjing 210008, China; 6Nanjing Neuropsychiatry Clinic Medical Center, Nanjing 210008, China
| | - Meng Hailan
- 1Department of Neurology, Affiliated Drum Tower Hospital, and; 2Jiangsu Key Laboratory for Molecular Medicine, Nanjing University Medical School, Nanjing 210008, China.; 3The State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing 210008, China; 5Jiangsu Province Stroke Center for Diagnosis and Therapy, Nanjing 210008, China; 6Nanjing Neuropsychiatry Clinic Medical Center, Nanjing 210008, China
| | - Han Lijuan
- 1Department of Neurology, Affiliated Drum Tower Hospital, and; 2Jiangsu Key Laboratory for Molecular Medicine, Nanjing University Medical School, Nanjing 210008, China.; 3The State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing 210008, China; 5Jiangsu Province Stroke Center for Diagnosis and Therapy, Nanjing 210008, China; 6Nanjing Neuropsychiatry Clinic Medical Center, Nanjing 210008, China
| | - Chen Fan
- 1Department of Neurology, Affiliated Drum Tower Hospital, and; 2Jiangsu Key Laboratory for Molecular Medicine, Nanjing University Medical School, Nanjing 210008, China.; 3The State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing 210008, China; 5Jiangsu Province Stroke Center for Diagnosis and Therapy, Nanjing 210008, China; 6Nanjing Neuropsychiatry Clinic Medical Center, Nanjing 210008, China
| | - Ye Dan
- 1Department of Neurology, Affiliated Drum Tower Hospital, and; 2Jiangsu Key Laboratory for Molecular Medicine, Nanjing University Medical School, Nanjing 210008, China.; 3The State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing 210008, China; 5Jiangsu Province Stroke Center for Diagnosis and Therapy, Nanjing 210008, China; 6Nanjing Neuropsychiatry Clinic Medical Center, Nanjing 210008, China
| | - Zhang He
- 1Department of Neurology, Affiliated Drum Tower Hospital, and; 2Jiangsu Key Laboratory for Molecular Medicine, Nanjing University Medical School, Nanjing 210008, China.; 3The State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing 210008, China; 5Jiangsu Province Stroke Center for Diagnosis and Therapy, Nanjing 210008, China; 6Nanjing Neuropsychiatry Clinic Medical Center, Nanjing 210008, China
| | - Xu Yun
- 1Department of Neurology, Affiliated Drum Tower Hospital, and; 2Jiangsu Key Laboratory for Molecular Medicine, Nanjing University Medical School, Nanjing 210008, China.; 3The State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing 210008, China; 5Jiangsu Province Stroke Center for Diagnosis and Therapy, Nanjing 210008, China; 6Nanjing Neuropsychiatry Clinic Medical Center, Nanjing 210008, China
| |
Collapse
|
88
|
Cárdenas AM, González-Jamett AM, Cea LA, Bevilacqua JA, Caviedes P. Dysferlin function in skeletal muscle: Possible pathological mechanisms and therapeutical targets in dysferlinopathies. Exp Neurol 2016; 283:246-54. [PMID: 27349407 DOI: 10.1016/j.expneurol.2016.06.026] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Revised: 06/22/2016] [Accepted: 06/23/2016] [Indexed: 12/18/2022]
Abstract
Mutations in the dysferlin gene are linked to a group of muscular dystrophies known as dysferlinopathies. These myopathies are characterized by progressive atrophy. Studies in muscle tissue from dysferlinopathy patients or dysferlin-deficient mice point out its importance in membrane repair. However, expression of dysferlin homologous proteins that restore sarcolemma repair function in dysferlinopathy animal models fail to arrest muscle wasting, therefore suggesting that dysferlin plays other critical roles in muscle function. In the present review, we discuss dysferlin functions in the skeletal muscle, as well as pathological mechanisms related to dysferlin mutations. Particular focus is presented related the effect of dysferlin on cell membrane related function, which affect its repair, vesicle trafficking, as well as Ca(2+) homeostasis. Such mechanisms could provide accessible targets for pharmacological therapies.
Collapse
Affiliation(s)
- Ana M Cárdenas
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile.
| | - Arlek M González-Jamett
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile; Programa de Anatomía y Biología del Desarrollo, ICBM, Facultad de Medicina, Departamento de Neurología y Neurocirugía, Hospital Clínico Universidad de Chile, Universidad de Chile, Santiago, Chile
| | - Luis A Cea
- Programa de Anatomía y Biología del Desarrollo, ICBM, Facultad de Medicina, Departamento de Neurología y Neurocirugía, Hospital Clínico Universidad de Chile, Universidad de Chile, Santiago, Chile
| | - Jorge A Bevilacqua
- Programa de Anatomía y Biología del Desarrollo, ICBM, Facultad de Medicina, Departamento de Neurología y Neurocirugía, Hospital Clínico Universidad de Chile, Universidad de Chile, Santiago, Chile
| | - Pablo Caviedes
- Programa de Farmacología Molecular y Clinica, ICBM, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| |
Collapse
|
89
|
Demonbreun AR, Quattrocelli M, Barefield DY, Allen MV, Swanson KE, McNally EM. An actin-dependent annexin complex mediates plasma membrane repair in muscle. J Cell Biol 2016; 213:705-18. [PMID: 27298325 PMCID: PMC4915191 DOI: 10.1083/jcb.201512022] [Citation(s) in RCA: 146] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2015] [Accepted: 05/19/2016] [Indexed: 01/03/2023] Open
Abstract
Disruption of the plasma membrane often accompanies cellular injury, and in muscle, plasma membrane resealing is essential for efficient recovery from injury. Muscle contraction, especially of lengthened muscle, disrupts the sarcolemma. To define the molecular machinery that directs repair, we applied laser wounding to live mammalian myofibers and assessed translocation of fluorescently tagged proteins using high-resolution microscopy. Within seconds of membrane disruption, annexins A1, A2, A5, and A6 formed a tight repair "cap." Actin was recruited to the site of damage, and annexin A6 cap formation was both actin dependent and Ca(2+) regulated. Repair proteins, including dysferlin, EHD1, EHD2, MG53, and BIN1, localized adjacent to the repair cap in a "shoulder" region enriched with phosphatidlyserine. Dye influx into muscle fibers lacking both dysferlin and the related protein myoferlin was substantially greater than control or individual null muscle fibers, underscoring the importance of shoulder-localized proteins. These data define the cap and shoulder as subdomains within the repair complex accumulating distinct and nonoverlapping components.
Collapse
Affiliation(s)
| | | | - David Y Barefield
- Center for Genetic Medicine, Northwestern University, Chicago, IL 60611
| | - Madison V Allen
- Center for Genetic Medicine, Northwestern University, Chicago, IL 60611
| | - Kaitlin E Swanson
- Center for Genetic Medicine, Northwestern University, Chicago, IL 60611 Department of Pathology, The University of Chicago, Chicago, IL 60637
| | | |
Collapse
|
90
|
Abstract
PURPOSE OF REVIEW Recently, genetic pathways that modify the clinical severity of Duchenne muscular dystrophy (DMD) have been identified. The pathways uncovered as modifiers are useful to predict prognosis and also elucidate molecular signatures that can be manipulated therapeutically. RECENT FINDINGS Modifiers have been identified using combinations of transcriptome and genome profiling. Osteopontin, encoded by the SPP1 gene, was found using gene expression profiling. Latent TGFβ binding protein 4, encoding latent TGFβ binding protein 4 was initially discovered using a genome-wide screen in mice and then validated in cohorts of DMD patients. These two pathways converge in that they both regulate TGFβ. A third modifier, Anxa6 that specifies annexin A6, is a calcium binding protein that has been identified using mouse models, and regulates the injury pathway and sarcolemmal resealing. SUMMARY Genetic modifiers can serve as biomarkers for outcomes in DMD. Modifiers can alter strength and ambulation in muscular dystrophy, and these same features can be used as endpoints used in clinical trials. Moreover, because genetic modifiers can influence outcomes, these genetic markers should be considered when stratifying results in muscular dystrophy.
Collapse
Affiliation(s)
- Andy H Vo
- Committee on Development, Regeneration and Stem Cell Biology, Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, The University of Chicago, Chicago, Illinois, USA
| | | |
Collapse
|
91
|
Membrane repair of human skeletal muscle cells requires Annexin-A5. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2016; 1863:2267-79. [PMID: 27286750 DOI: 10.1016/j.bbamcr.2016.06.003] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Revised: 05/31/2016] [Accepted: 06/03/2016] [Indexed: 11/20/2022]
Abstract
Defect in membrane repair contributes to the development of limb girdle muscular dystrophy type 2B (LGMD2B) and Miyoshi myopathy. In healthy skeletal muscle, unraveling membrane repair mechanisms requires to establish an exhaustive list of the components of the resealing machinery. Here we show that human myotubes rendered deficient for Annexin-A5 (AnxA5) suffer from a severe defect in membrane resealing. This defect is rescued by the addition of recombinant AnxA5 while an AnxA5 mutant, which is unable to form 2D protein arrays, has no effect. Using correlative light and electron microscopy, we show that AnxA5 binds to the edges of the torn membrane, as early as a few seconds after sarcolemma injury, where it probably self-assembles into 2D arrays. In addition, we observed that membrane resealing is associated with the presence of a cluster of lipid vesicles at the wounded site. AnxA5 is present at the surface of these vesicles and may thus participate in plugging the cell membrane disruption. Finally, we show that AnxA5 behaves similarly in myotubes from a muscle cell line established from a patient suffering from LGMD2B, a myopathy due to dysferlin mutations, which indicates that trafficking of AnxA5 during sarcolemma damage is independent of the presence of dysferlin.
Collapse
|
92
|
Davenport NR, Sonnemann KJ, Eliceiri KW, Bement WM. Membrane dynamics during cellular wound repair. Mol Biol Cell 2016; 27:2272-85. [PMID: 27226483 PMCID: PMC4945144 DOI: 10.1091/mbc.e16-04-0223] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Accepted: 05/18/2016] [Indexed: 12/31/2022] Open
Abstract
Fusion of intracellular compartments with each other and the plasma membrane has been hypothesized to occur at sites of cellular injury but has never been directly visualized. High-speed microscopy reveals this process and shows that resealing is accompanied by intracellular patterning of proteins, ions, and membrane lipids. Cells rapidly reseal after damage, but how they do so is unknown. It has been hypothesized that resealing occurs due to formation of a patch derived from rapid fusion of intracellular compartments at the wound site. However, patching has never been directly visualized. Here we study membrane dynamics in wounded Xenopus laevis oocytes at high spatiotemporal resolution. Consistent with the patch hypothesis, we find that damage triggers rampant fusion of intracellular compartments, generating a barrier that limits influx of extracellular dextrans. Patch formation is accompanied by compound exocytosis, local accumulation and aggregation of vesicles, and rupture of compartments facing the external environment. Subcellular patterning is evident as annexin A1, dysferlin, diacylglycerol, active Rho, and active Cdc42 are recruited to compartments confined to different regions around the wound. We also find that a ring of elevated intracellular calcium overlaps the region where membrane dynamics are most evident and persists for several minutes. The results provide the first direct visualization of membrane patching during membrane repair, reveal novel features of the repair process, and show that a remarkable degree of spatial patterning accompanies damage-induced membrane dynamics.
Collapse
Affiliation(s)
- Nicholas R Davenport
- Program in Cellular and Molecular Biology, University of Wisconsin-Madison, Madison, WI 53706
| | - Kevin J Sonnemann
- Laboratory of Cell and Molecular Biology, University of Wisconsin-Madison, Madison, WI 53706
| | - Kevin W Eliceiri
- Laboratory of Cell and Molecular Biology, University of Wisconsin-Madison, Madison, WI 53706 Laboratory for Optical and Computational Instrumentation, University of Wisconsin-Madison, Madison, WI 53706
| | - William M Bement
- Program in Cellular and Molecular Biology, University of Wisconsin-Madison, Madison, WI 53706 Laboratory of Cell and Molecular Biology, University of Wisconsin-Madison, Madison, WI 53706 Department of Zoology, University of Wisconsin-Madison, Madison, WI 53706
| |
Collapse
|
93
|
Demonbreun AR, Allen MV, Warner JL, Barefield DY, Krishnan S, Swanson KE, Earley JU, McNally EM. Enhanced Muscular Dystrophy from Loss of Dysferlin Is Accompanied by Impaired Annexin A6 Translocation after Sarcolemmal Disruption. THE AMERICAN JOURNAL OF PATHOLOGY 2016; 186:1610-22. [PMID: 27070822 DOI: 10.1016/j.ajpath.2016.02.005] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Revised: 01/20/2016] [Accepted: 02/11/2016] [Indexed: 02/03/2023]
Abstract
Dysferlin is a membrane-associated protein implicated in membrane resealing; loss of dysferlin leads to muscular dystrophy. We examined the same loss-of-function Dysf mutation in two different mouse strains, 129T2/SvEmsJ (Dysf(129)) and C57BL/6J (Dysf(B6)). Although there are many genetic differences between these two strains, we focused on polymorphisms in Anxa6 because these variants were previously associated with modifying a pathologically distinct form of muscular dystrophy and increased the production of a truncated annexin A6 protein. Dysferlin deficiency in the C57BL/6J background was associated with increased Evan's Blue dye uptake into muscle and increased serum creatine kinase compared to the 129T2/SvEmsJ background. In the C57BL/6J background, dysferlin loss was associated with enhanced pathologic severity, characterized by decreased mean fiber cross-sectional area, increased internalized nuclei, and increased fibrosis, compared to that in Dysf(129) mice. Macrophage infiltrate was also increased in Dysf(B6) muscle. High-resolution imaging of live myofibers demonstrated that fibers from Dysf(B6) mice displayed reduced translocation of full-length annexin A6 to the site of laser-induced sarcolemmal disruption compared to Dysf(129) myofibers, and impaired translocation of annexin A6 associated with impaired resealing of the sarcolemma. These results provide one mechanism by which the C57BL/6J background intensifies dysferlinopathy, giving rise to a more severe form of muscular dystrophy in the Dysf(B6) mouse model through increased membrane leak and inflammation.
Collapse
Affiliation(s)
| | - Madison V Allen
- Center for Genetic Medicine, Northwestern University, Chicago, Illinois
| | - James L Warner
- Center for Genetic Medicine, Northwestern University, Chicago, Illinois
| | - David Y Barefield
- Center for Genetic Medicine, Northwestern University, Chicago, Illinois
| | - Swathi Krishnan
- Department of Medicine, The University of Chicago, Chicago, Illinois
| | - Kaitlin E Swanson
- Department of Pathology, The University of Chicago, Chicago, Illinois
| | - Judy U Earley
- Center for Genetic Medicine, Northwestern University, Chicago, Illinois
| | | |
Collapse
|
94
|
Castro-Gomes T, Corrotte M, Tam C, Andrews NW. Plasma Membrane Repair Is Regulated Extracellularly by Proteases Released from Lysosomes. PLoS One 2016; 11:e0152583. [PMID: 27028538 PMCID: PMC4814109 DOI: 10.1371/journal.pone.0152583] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Accepted: 03/16/2016] [Indexed: 12/28/2022] Open
Abstract
Eukaryotic cells rapidly repair wounds on their plasma membrane. Resealing is Ca2+-dependent, and involves exocytosis of lysosomes followed by massive endocytosis. Extracellular activity of the lysosomal enzyme acid sphingomyelinase was previously shown to promote endocytosis and wound removal. However, whether lysosomal proteases released during cell injury participate in resealing is unknown. Here we show that lysosomal proteases regulate plasma membrane repair. Extracellular proteolysis is detected shortly after cell wounding, and inhibition of this process blocks repair. Conversely, surface protein degradation facilitates plasma membrane resealing. The abundant lysosomal cysteine proteases cathepsin B and L, known to proteolytically remodel the extracellular matrix, are rapidly released upon cell injury and are required for efficient plasma membrane repair. In contrast, inhibition of aspartyl proteases or RNAi-mediated silencing of the lysosomal aspartyl protease cathepsin D enhances resealing, an effect associated with the accumulation of active acid sphingomyelinase on the cell surface. Thus, secreted lysosomal cysteine proteases may promote repair by facilitating membrane access of lysosomal acid sphingomyelinase, which promotes wound removal and is subsequently downregulated extracellularly by a process involving cathepsin D.
Collapse
Affiliation(s)
- Thiago Castro-Gomes
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, Maryland, 20742, United States of America
| | - Matthias Corrotte
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, Maryland, 20742, United States of America
| | - Christina Tam
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, Maryland, 20742, United States of America
| | - Norma W. Andrews
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, Maryland, 20742, United States of America
- * E-mail:
| |
Collapse
|
95
|
Calyjur PC, Almeida CDF, Ayub-Guerrieri D, Ribeiro AF, Fernandes SDA, Ishiba R, dos Santos ALF, Onofre-Oliveira P, Vainzof M. The mdx Mutation in the 129/Sv Background Results in a Milder Phenotype: Transcriptome Comparative Analysis Searching for the Protective Factors. PLoS One 2016; 11:e0150748. [PMID: 26954670 PMCID: PMC4783004 DOI: 10.1371/journal.pone.0150748] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Accepted: 02/18/2016] [Indexed: 12/23/2022] Open
Abstract
The mdx mouse is a good genetic and molecular murine model for Duchenne Muscular Dystrophy (DMD), a progressive and devastating muscle disease. However, this model is inappropriate for testing new therapies due to its mild phenotype. Here, we transferred the mdx mutation to the 129/Sv strain with the aim to create a more severe model for DMD. Unexpectedly, functional analysis of the first three generations of mdx129 showed a progressive amelioration of the phenotype, associated to less connective tissue replacement, and more regeneration than the original mdxC57BL. Transcriptome comparative analysis was performed to identify what is protecting this new model from the dystrophic characteristics. The mdxC57BL presents three times more differentially expressed genes (DEGs) than the mdx129 (371 and 137 DEGs respectively). However, both models present more overexpressed genes than underexpressed, indicating that the dystrophic and regenerative alterations are associated with the activation rather than repression of genes. As to functional categories, the DEGs of both mdx models showed a predominance of immune system genes. Excluding this category, the mdx129 model showed a decreased participation of the endo/exocytic pathway and homeostasis categories, and an increased participation of the extracellular matrix and enzymatic activity categories. Spp1 gene overexpression was the most significant DEG exclusively expressed in the mdx129 strain. This was confirmed through relative mRNA analysis and osteopontin protein quantification. The amount of the 66 kDa band of the protein, representing the post-translational product of the gene, was about 4,8 times higher on western blotting. Spp1 is a known DMD prognostic biomarker, and our data indicate that its upregulation can benefit phenotype. Modeling the expression of the DEGs involved in the mdx mutation with a benign course should be tested as a possible therapeutic target for the dystrophic process.
Collapse
Affiliation(s)
- Priscila Clara Calyjur
- Laboratory of Muscle Proteins and Comparative Histopathology, Human Genome and Stem-cell Research Center, Biosciences Institute, University of São Paulo, São Paulo, Brazil
| | - Camila de Freitas Almeida
- Laboratory of Muscle Proteins and Comparative Histopathology, Human Genome and Stem-cell Research Center, Biosciences Institute, University of São Paulo, São Paulo, Brazil
| | - Danielle Ayub-Guerrieri
- Laboratory of Muscle Proteins and Comparative Histopathology, Human Genome and Stem-cell Research Center, Biosciences Institute, University of São Paulo, São Paulo, Brazil
| | - Antonio Fernando Ribeiro
- Laboratory of Muscle Proteins and Comparative Histopathology, Human Genome and Stem-cell Research Center, Biosciences Institute, University of São Paulo, São Paulo, Brazil
| | - Stephanie de Alcântara Fernandes
- Laboratory of Muscle Proteins and Comparative Histopathology, Human Genome and Stem-cell Research Center, Biosciences Institute, University of São Paulo, São Paulo, Brazil
| | - Renata Ishiba
- Laboratory of Muscle Proteins and Comparative Histopathology, Human Genome and Stem-cell Research Center, Biosciences Institute, University of São Paulo, São Paulo, Brazil
| | - Andre Luis Fernandes dos Santos
- Laboratory of Muscle Proteins and Comparative Histopathology, Human Genome and Stem-cell Research Center, Biosciences Institute, University of São Paulo, São Paulo, Brazil
| | - Paula Onofre-Oliveira
- Laboratory of Muscle Proteins and Comparative Histopathology, Human Genome and Stem-cell Research Center, Biosciences Institute, University of São Paulo, São Paulo, Brazil
| | - Mariz Vainzof
- Laboratory of Muscle Proteins and Comparative Histopathology, Human Genome and Stem-cell Research Center, Biosciences Institute, University of São Paulo, São Paulo, Brazil
- * E-mail:
| |
Collapse
|
96
|
A Common Variant of PROK1 (V67I) Acts as a Genetic Modifier in Early Human Pregnancy through Down-Regulation of Gene Expression. Int J Mol Sci 2016; 17:ijms17020162. [PMID: 26828479 PMCID: PMC4783896 DOI: 10.3390/ijms17020162] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Revised: 01/16/2016] [Accepted: 01/16/2016] [Indexed: 11/30/2022] Open
Abstract
PROK1-V67I has been shown to play a role as a modifier gene in the PROK1-PROKR system of human early pregnancy. To explore the related modifier mechanism of PROK1-V67I, we carried out a comparison study at the gene expression level and the cell function alternation of V67I, and its wild-type (WT), in transiently-transfected cells. We, respectively, performed quantitative RT-PCR and ELISA assays to evaluate the protein and/or transcript level of V67I and WT in HTR-8/SV neo, JAR, Ishikawa, and HEK293 cells. Transiently V67I- or WT-transfected HTR-8/SV neo and HEK293 cells were used to investigate cell function alternations. The transcript and protein expressions were down-regulated in all cell lines, ranging from 20% to 70%, compared with WT. There were no significant differences in the ligand activities of V67I and WT with regard to cell proliferation, cell invasion, calcium influx, and tubal formation. Both PROK1 alleles promoted cell invasion and intracellular calcium mobilization, whereas they had no significant effects on cell proliferation and tubal formation. In conclusion, the biological effects of PROK1-V67I on cell functions are similar to those of WT, and the common variant of V67I may act as a modifier in the PROK1-PROKR system through down-regulation of PROK1 expression. This study may provide a general mechanism that the common variant of V67I, modifying the disease severity of PROK1-related pathophysiologies.
Collapse
|
97
|
Abstract
Mature muscle has a unique structure that is amenable to live cell imaging. Herein, we describe the experimental protocol for expressing fluorescently labeled proteins in the flexor digitorum brevis (FDB) muscle. Conditions have been optimized to provide a large number of high quality myofibers expressing the electroporated plasmid while minimizing muscle damage. The method employs fluorescent tags on various proteins. Combining this expression method with high resolution confocal microscopy permits live cell imaging, including imaging after laser-induced damage. Fluorescent dyes combined with imaging of fluorescently-tagged proteins provides information regarding the basic structure of muscle and its response to stimuli.
Collapse
|
98
|
Leikina E, Defour A, Melikov K, Van der Meulen JH, Nagaraju K, Bhuvanendran S, Gebert C, Pfeifer K, Chernomordik LV, Jaiswal JK. Annexin A1 Deficiency does not Affect Myofiber Repair but Delays Regeneration of Injured Muscles. Sci Rep 2015; 5:18246. [PMID: 26667898 PMCID: PMC4678367 DOI: 10.1038/srep18246] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Accepted: 11/13/2015] [Indexed: 12/28/2022] Open
Abstract
Repair and regeneration of the injured skeletal myofiber involves fusion of intracellular vesicles with sarcolemma and fusion of the muscle progenitor cells respectively. In vitro experiments have identified involvement of Annexin A1 (Anx A1) in both these fusion processes. To determine if Anx A1 contributes to these processes during muscle repair in vivo, we have assessed muscle growth and repair in Anx A1-deficient mouse (AnxA1-/-). We found that the lack of Anx A1 does not affect the muscle size and repair of myofibers following focal sarcolemmal injury and lengthening contraction injury. However, the lack of Anx A1 delayed muscle regeneration after notexin-induced injury. This delay in muscle regeneration was not caused by a slowdown in proliferation and differentiation of satellite cells. Instead, lack of Anx A1 lowered the proportion of differentiating myoblasts that managed to fuse with the injured myofibers by days 5 and 7 after notexin injury as compared to the wild type (w.t.) mice. Despite this early slowdown in fusion of Anx A1-/- myoblasts, regeneration caught up at later times post injury. These results establish in vivo role of Anx A1 in cell fusion required for myofiber regeneration and not in intracellular vesicle fusion needed for repair of myofiber sarcolemma.
Collapse
Affiliation(s)
- Evgenia Leikina
- Section on Membrane Biology, Program of Physical Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bldg. 10/Rm. 10D05, 10 Center Dr. Bethesda, Maryland 20892-1855, USA
| | - Aurelia Defour
- Children's National Medical Center, Center for Genetic Medicine Research, 111 Michigan Avenue, NW, Washington DC 20010-2970, USA
| | - Kamran Melikov
- Section on Membrane Biology, Program of Physical Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bldg. 10/Rm. 10D05, 10 Center Dr. Bethesda, Maryland 20892-1855, USA
| | - Jack H Van der Meulen
- Children's National Medical Center, Center for Genetic Medicine Research, 111 Michigan Avenue, NW, Washington DC 20010-2970, USA
| | - Kanneboyina Nagaraju
- Children's National Medical Center, Center for Genetic Medicine Research, 111 Michigan Avenue, NW, Washington DC 20010-2970, USA.,Department of Integrative Systems Biology, George Washington University School of Medicine and Health Sciences, Washington DC, USA
| | - Shivaprasad Bhuvanendran
- Children's National Medical Center, Center for Genetic Medicine Research, 111 Michigan Avenue, NW, Washington DC 20010-2970, USA
| | - Claudia Gebert
- Section on Genome Imprinting, Program on Genomics of Differentiation, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, USA
| | - Karl Pfeifer
- Section on Genome Imprinting, Program on Genomics of Differentiation, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, USA
| | - Leonid V Chernomordik
- Section on Membrane Biology, Program of Physical Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bldg. 10/Rm. 10D05, 10 Center Dr. Bethesda, Maryland 20892-1855, USA
| | - Jyoti K Jaiswal
- Children's National Medical Center, Center for Genetic Medicine Research, 111 Michigan Avenue, NW, Washington DC 20010-2970, USA.,Department of Integrative Systems Biology, George Washington University School of Medicine and Health Sciences, Washington DC, USA
| |
Collapse
|
99
|
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
|
100
|
Abstract
Since an intact membrane is required for normal cellular homeostasis, membrane repair is essential for cell survival. Human genetic studies, combined with the development of novel animal models and refinement of techniques to study cellular injury, have now uncovered series of repair proteins highly relevant for human health. Many of the deficient repair pathways manifest in skeletal muscle, where defective repair processes result in myopathies or other forms of muscle disease. Dysferlin is a membrane-associated protein implicated in sarcolemmal repair and also linked to other membrane functions including the maintenance of transverse tubules in muscle. MG53, annexins, and Eps15 homology domain-containing proteins interact with dysferlin to form a membrane repair complex and similarly have roles in membrane trafficking in muscle. These molecular features of membrane repair are not unique to skeletal muscle, but rather skeletal muscle, due to its high demands, is more dependent on an efficient repair process. Phosphatidylserine and phosphatidylinositol 4,5-bisphosphate, as well as Ca(2+), are central regulators of membrane organization during repair. Given the importance of muscle health in disease and in aging, these pathways are targets to enhance muscle function and recovery from injury.
Collapse
|