1
|
Goret M, Thomas M, Edelweiss E, Messaddeq N, Laporte J. BIN1 reduction ameliorates DNM2-related Charcot-Marie-Tooth neuropathy. Proc Natl Acad Sci U S A 2025; 122:e2419244122. [PMID: 40042903 PMCID: PMC11912451 DOI: 10.1073/pnas.2419244122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 01/27/2025] [Indexed: 03/19/2025] Open
Abstract
Charcot-Marie-Tooth (CMT) disease, the most common inherited neuromuscular disorder, manifests as progressive muscle weakness and peripheral nerve defects. Dominant mutations in DNM2, encoding the large GTPase dynamin 2, result in CMT without any suggested therapeutic strategy. Different dominant mutations in DNM2 also cause centronuclear myopathy (CNM), and increasing BIN1 (amphiphysin 2), an endogenous modulator of DNM2, rescued CNM in mice. Here, we found that increasing BIN1 level exacerbated the phenotypes of the Dnm2K562E/+ mouse carrying the most common DNM2-CMT mutation. Conversely, whole-body reduction of Bin1 expression level, through the generation of Dnm2K562E/+ mice with heterozygous loss of BIN1, restored motor performance and ameliorated muscle organization and structural defects of peripheral nerves. The rescue of motor defects was maintained at least up to 1 y of age. BIN1 inhibited the GTPase activity of DNM2, and the rescue was driven by an increased activity of the K562E DNM2-CMT mutant, and a normalization of integrin localization in muscle. Overall, this study highlights BIN1 as a modifier of DNM2-CMT, and its reduction as a potential therapeutic strategy. It also revealed an opposite pathological mechanism and inverse therapeutic concepts for DNM2-CMT peripheral neuropathy versus DNM2-CNM myopathy.
Collapse
Affiliation(s)
- Marie Goret
- Institut de génétique, biologie moléculaire et cellulaire, département de Médecine translationnelle et neurogénétique, INSERM U1258, CNRS UMR7104, Université de Strasbourg, Illkirch 67404, France
| | - Morgane Thomas
- Institut de génétique, biologie moléculaire et cellulaire, département de Médecine translationnelle et neurogénétique, INSERM U1258, CNRS UMR7104, Université de Strasbourg, Illkirch 67404, France
| | - Evelina Edelweiss
- Institut de génétique, biologie moléculaire et cellulaire, département de Médecine translationnelle et neurogénétique, INSERM U1258, CNRS UMR7104, Université de Strasbourg, Illkirch 67404, France
| | - Nadia Messaddeq
- Institut de génétique, biologie moléculaire et cellulaire, département de Médecine translationnelle et neurogénétique, INSERM U1258, CNRS UMR7104, Université de Strasbourg, Illkirch 67404, France
| | - Jocelyn Laporte
- Institut de génétique, biologie moléculaire et cellulaire, département de Médecine translationnelle et neurogénétique, INSERM U1258, CNRS UMR7104, Université de Strasbourg, Illkirch 67404, France
| |
Collapse
|
2
|
Ritter AJ, Wallace A, Ronaghi N, Sanford J. junctionCounts: comprehensive alternative splicing analysis and prediction of isoform-level impacts to the coding sequence. NAR Genom Bioinform 2024; 6:lqae093. [PMID: 39131822 PMCID: PMC11310779 DOI: 10.1093/nargab/lqae093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 07/15/2024] [Accepted: 07/22/2024] [Indexed: 08/13/2024] Open
Abstract
Alternative splicing (AS) is emerging as an important regulatory process for complex biological processes. Transcriptomic studies therefore commonly involve the identification and quantification of alternative processing events, but the need for predicting the functional consequences of changes to the relative inclusion of alternative events remains largely unaddressed. Many tools exist for the former task, albeit each constrained to its own event type definitions. Few tools exist for the latter task; each with significant limitations. To address these issues we developed junctionCounts, which captures both simple and complex pairwise AS events and quantifies them with straightforward exon-exon and exon-intron junction reads in RNA-seq data, performing competitively among similar tools in terms of sensitivity, false discovery rate and quantification accuracy. Its partner utility, cdsInsertion, identifies transcript coding sequence (CDS) information via in silico translation from annotated start codons, including the presence of premature termination codons. Finally, findSwitchEvents connects AS events with CDS information to predict the impact of individual events to the isoform-level CDS. We used junctionCounts to characterize splicing dynamics and NMD regulation during neuronal differentiation across four primates, demonstrating junctionCounts' capacity to robustly characterize AS in a variety of organisms and to predict its effect on mRNA isoform fate.
Collapse
Affiliation(s)
- Alexander J Ritter
- Department of Biomolecular Engineering, University of California Santa Cruz, Santa Cruz, CA 95064, USA
| | - Andrew Wallace
- Department of Molecular, Cell and Developmental Biology, University of California Santa Cruz, Santa Cruz, CA 95064, USA
| | - Neda Ronaghi
- Department of Molecular, Cell and Developmental Biology, University of California Santa Cruz, Santa Cruz, CA 95064, USA
| | - Jeremy R Sanford
- Department of Molecular, Cell and Developmental Biology, University of California Santa Cruz, Santa Cruz, CA 95064, USA
| |
Collapse
|
3
|
Chechenova M, McLendon L, Dallas B, Stratton H, Kiani K, Gerberich E, Alekseyenko A, Tamba N, An S, Castillo L, Czajkowski E, Talley C, Brown A, Bryantsev AL. Muscle degeneration in aging Drosophila flies: the role of mechanical stress. Skelet Muscle 2024; 14:20. [PMID: 39164781 PMCID: PMC11334408 DOI: 10.1186/s13395-024-00352-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 08/09/2024] [Indexed: 08/22/2024] Open
Abstract
Muscle wasting is a universal hallmark of aging which is displayed by a wide range of organisms, although the causes and mechanisms of this phenomenon are not fully understood. We used Drosophila to characterize the phenomenon of spontaneous muscle fiber degeneration (SMFD) during aging. We found that SMFD occurs across diverse types of somatic muscles, progresses with chronological age, and positively correlates with functional muscle decline. Data from vital dyes and morphological markers imply that degenerative fibers most likely die by necrosis. Mechanistically, SMFD is driven by the damage resulting from muscle contractions, and the nervous system may play a significant role in this process. Our quantitative model of SMFD assessment can be useful in identifying and validating novel genetic factors that influence aging-related muscle wasting.
Collapse
Affiliation(s)
- Maria Chechenova
- Department of Molecular and Cellular Biology, Kennesaw State University, 105 Marietta Dr., NW, Room 4004, MD 1201, Kennesaw, GA, 30144, USA
- Present Affiliation: MNG Laboratories, A LabCorp Company, Atlanta, GA, USA
| | - Lilla McLendon
- Department of Molecular and Cellular Biology, Kennesaw State University, 105 Marietta Dr., NW, Room 4004, MD 1201, Kennesaw, GA, 30144, USA
| | - Bracey Dallas
- Department of Molecular and Cellular Biology, Kennesaw State University, 105 Marietta Dr., NW, Room 4004, MD 1201, Kennesaw, GA, 30144, USA
| | - Hannah Stratton
- Department of Molecular and Cellular Biology, Kennesaw State University, 105 Marietta Dr., NW, Room 4004, MD 1201, Kennesaw, GA, 30144, USA
| | - Kaveh Kiani
- Department of Molecular and Cellular Biology, Kennesaw State University, 105 Marietta Dr., NW, Room 4004, MD 1201, Kennesaw, GA, 30144, USA
| | - Erik Gerberich
- Department of Molecular and Cellular Biology, Kennesaw State University, 105 Marietta Dr., NW, Room 4004, MD 1201, Kennesaw, GA, 30144, USA
| | - Alesia Alekseyenko
- Department of Molecular and Cellular Biology, Kennesaw State University, 105 Marietta Dr., NW, Room 4004, MD 1201, Kennesaw, GA, 30144, USA
| | - Natasya Tamba
- Department of Molecular and Cellular Biology, Kennesaw State University, 105 Marietta Dr., NW, Room 4004, MD 1201, Kennesaw, GA, 30144, USA
| | - SooBin An
- Department of Molecular and Cellular Biology, Kennesaw State University, 105 Marietta Dr., NW, Room 4004, MD 1201, Kennesaw, GA, 30144, USA
| | - Lizzet Castillo
- Department of Biology, University of New Mexico, Albuquerque, NM, USA
| | - Emily Czajkowski
- Department of Biology, University of New Mexico, Albuquerque, NM, USA
- Present Affiliation: Department of Molecular Biosciences, Northwestern University, Evanston, IL, USA
| | - Christina Talley
- Department of Molecular and Cellular Biology, Kennesaw State University, 105 Marietta Dr., NW, Room 4004, MD 1201, Kennesaw, GA, 30144, USA
| | - Austin Brown
- Department of Mathematics, Kennesaw State University, Kennesaw, GA, USA
| | - Anton L Bryantsev
- Department of Molecular and Cellular Biology, Kennesaw State University, 105 Marietta Dr., NW, Room 4004, MD 1201, Kennesaw, GA, 30144, USA.
| |
Collapse
|
4
|
Zambo B, Edelweiss E, Morlet B, Negroni L, Pajkos M, Dosztanyi Z, Ostergaard S, Trave G, Laporte J, Gogl G. Uncovering the BIN1-SH3 interactome underpinning centronuclear myopathy. eLife 2024; 13:RP95397. [PMID: 38995680 PMCID: PMC11245310 DOI: 10.7554/elife.95397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/13/2024] Open
Abstract
Truncation of the protein-protein interaction SH3 domain of the membrane remodeling Bridging Integrator 1 (BIN1, Amphiphysin 2) protein leads to centronuclear myopathy. Here, we assessed the impact of a set of naturally observed, previously uncharacterized BIN1 SH3 domain variants using conventional in vitro and cell-based assays monitoring the BIN1 interaction with dynamin 2 (DNM2) and identified potentially harmful ones that can be also tentatively connected to neuromuscular disorders. However, SH3 domains are typically promiscuous and it is expected that other, so far unknown partners of BIN1 exist besides DNM2, that also participate in the development of centronuclear myopathy. In order to shed light on these other relevant interaction partners and to get a holistic picture of the pathomechanism behind BIN1 SH3 domain variants, we used affinity interactomics. We identified hundreds of new BIN1 interaction partners proteome-wide, among which many appear to participate in cell division, suggesting a critical role of BIN1 in the regulation of mitosis. Finally, we show that the identified BIN1 mutations indeed cause proteome-wide affinity perturbation, signifying the importance of employing unbiased affinity interactomic approaches.
Collapse
Affiliation(s)
- Boglarka Zambo
- Equipe Labellisee Ligue 2015, Departement de Biologie Structurale Integrative, Institut de Genetique et de Biologie Moleculaire et Cellulaire (IGBMC), INSERM U1258/CNRS UMR 7104/Universite de Strasbourg, Illkirch, France
| | - Evelina Edelweiss
- Institut de Genetique et de Biologie Moléculaire et Cellulaire (IGBMC), INSERM U1258/CNRS UMR 7104/Université de Strasbourg, Illkirch, France
| | - Bastien Morlet
- Institut de Genetique et de Biologie Moléculaire et Cellulaire (IGBMC), INSERM U1258/CNRS UMR 7104/Université de Strasbourg, Illkirch, France
| | - Luc Negroni
- Institut de Genetique et de Biologie Moléculaire et Cellulaire (IGBMC), INSERM U1258/CNRS UMR 7104/Université de Strasbourg, Illkirch, France
| | - Matyas Pajkos
- Department of Biochemistry, ELTE Eötvös Loránd University, Budapest, Hungary
| | - Zsuzsanna Dosztanyi
- Department of Biochemistry, ELTE Eötvös Loránd University, Budapest, Hungary
| | - Soren Ostergaard
- Novo Nordisk A/S, Global Research Technologies, Novo Nordisk Research Park, Maaloev, Denmark
| | - Gilles Trave
- Equipe Labellisee Ligue 2015, Departement de Biologie Structurale Integrative, Institut de Genetique et de Biologie Moleculaire et Cellulaire (IGBMC), INSERM U1258/CNRS UMR 7104/Universite de Strasbourg, Illkirch, France
| | - Jocelyn Laporte
- Institut de Genetique et de Biologie Moléculaire et Cellulaire (IGBMC), INSERM U1258/CNRS UMR 7104/Université de Strasbourg, Illkirch, France
| | - Gergo Gogl
- Equipe Labellisee Ligue 2015, Departement de Biologie Structurale Integrative, Institut de Genetique et de Biologie Moleculaire et Cellulaire (IGBMC), INSERM U1258/CNRS UMR 7104/Universite de Strasbourg, Illkirch, France
| |
Collapse
|
5
|
Castillo-Mancho V, Atienza-Manuel A, Sarmiento-Jiménez J, Ruiz-Gómez M, Culi J. Phospholipid scramblase 1: an essential component of the nephrocyte slit diaphragm. Cell Mol Life Sci 2024; 81:261. [PMID: 38878170 PMCID: PMC11335299 DOI: 10.1007/s00018-024-05287-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 05/03/2024] [Accepted: 05/20/2024] [Indexed: 06/29/2024]
Abstract
Blood ultrafiltration in nephrons critically depends on specialized intercellular junctions between podocytes, named slit diaphragms (SDs). Here, by studying a homologous structure found in Drosophila nephrocytes, we identify the phospholipid scramblase Scramb1 as an essential component of the SD, uncovering a novel link between membrane dynamics and SD formation. In scramb1 mutants, SDs fail to form. Instead, the SD components Sticks and stones/nephrin, Polychaetoid/ZO-1, and the Src-kinase Src64B/Fyn associate in cortical foci lacking the key SD protein Dumbfounded/NEPH1. Scramb1 interaction with Polychaetoid/ZO-1 and Flotillin2, the presence of essential putative palmitoylation sites and its capacity to oligomerize, suggest a function in promoting SD assembly within lipid raft microdomains. Furthermore, Scramb1 interactors as well as its functional sensitivity to temperature, suggest an active involvement in membrane remodeling processes during SD assembly. Remarkably, putative Ca2+-binding sites in Scramb1 are essential for its activity raising the possibility that Ca2+ signaling may control the assembly of SDs by impacting on Scramb1 activity.
Collapse
Affiliation(s)
- Vicente Castillo-Mancho
- Centro de Biología Molecular Severo Ochoa, CSIC and UAM, Nicolás Cabrera 1, Cantoblanco, Madrid, 28049, Spain
| | - Alexandra Atienza-Manuel
- Centro de Biología Molecular Severo Ochoa, CSIC and UAM, Nicolás Cabrera 1, Cantoblanco, Madrid, 28049, Spain
| | - Jorge Sarmiento-Jiménez
- Centro de Biología Molecular Severo Ochoa, CSIC and UAM, Nicolás Cabrera 1, Cantoblanco, Madrid, 28049, Spain
| | - Mar Ruiz-Gómez
- Centro de Biología Molecular Severo Ochoa, CSIC and UAM, Nicolás Cabrera 1, Cantoblanco, Madrid, 28049, Spain.
| | - Joaquim Culi
- Centro de Biología Molecular Severo Ochoa, CSIC and UAM, Nicolás Cabrera 1, Cantoblanco, Madrid, 28049, Spain.
| |
Collapse
|
6
|
Giraud Q, Laporte J. Amphiphysin-2 (BIN1) functions and defects in cardiac and skeletal muscle. Trends Mol Med 2024; 30:579-591. [PMID: 38514365 DOI: 10.1016/j.molmed.2024.02.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 02/11/2024] [Accepted: 02/14/2024] [Indexed: 03/23/2024]
Abstract
Amphiphysin-2 is a ubiquitously expressed protein also known as bridging integrator 1 (BIN1), playing a critical role in membrane remodeling, trafficking, and cytoskeleton dynamics in a wide range of tissues. Mutations in the gene encoding BIN1 cause centronuclear myopathies (CNM), and recent evidence has implicated BIN1 in heart failure, underlining its crucial role in both skeletal and cardiac muscle. Furthermore, altered expression of BIN1 is linked to an increased risk of late-onset Alzheimer's disease and several types of cancer, including breast, colon, prostate, and lung cancers. Recently, the first proof-of-concept for potential therapeutic strategies modulating BIN1 were obtained for muscle diseases. In this review article, we discuss the similarities and differences in BIN1's functions in cardiac and skeletal muscle, along with its associated diseases and potential therapies.
Collapse
Affiliation(s)
- Quentin Giraud
- Department of Translational Medicine and Neurogenetics, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC, INSERM U1258, CNRS UMR7104, Université de Strasbourg, Illkirch-Graffenstaden, 67400, France
| | - Jocelyn Laporte
- Department of Translational Medicine and Neurogenetics, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC, INSERM U1258, CNRS UMR7104, Université de Strasbourg, Illkirch-Graffenstaden, 67400, France.
| |
Collapse
|
7
|
Kawaguchi K, Fujita N. Shaping transverse-tubules: central mechanisms that play a role in the cytosol zoning for muscle contraction. J Biochem 2024; 175:125-131. [PMID: 37848047 PMCID: PMC10873525 DOI: 10.1093/jb/mvad083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 10/05/2023] [Accepted: 10/11/2023] [Indexed: 10/19/2023] Open
Abstract
A transverse-tubule (T-tubule) is an invagination of the plasma membrane penetrating deep into muscle cells. An extensive membrane network of T-tubules is crucial for rapid and synchronized signal transmission from the cell surface to the entire sarcoplasmic reticulum for Ca2+ release, leading to muscle contraction. T-tubules are also indispensable for the formation and positioning of other muscle organelles. Their structure and physiological roles are relatively well established; however, the mechanisms shaping T-tubules require further elucidation. Centronuclear myopathy (CNM), an inherited muscular disorder, accompanies structural defects in T-tubules. Membrane traffic-related genes, including MTM1 (Myotubularin 1), DNM2 (Dynamin 2), and BIN1 (Bridging Integrator-1), were identified as causative genes of CNM. In addition, causative genes for other muscle diseases are also reported to be involved in the formation and maintenance of T-tubules. This review summarizes current knowledge on the mechanisms of how T-tubule formation and maintenance is regulated.
Collapse
Affiliation(s)
- Kohei Kawaguchi
- Cell Biology Center, Institute of Innovative Research, Tokyo Institute of Technology, 4259 S2-11 Nagatsuta-cho, Midori-ku, Yokohama 226-8503, Japan
| | - Naonobu Fujita
- Cell Biology Center, Institute of Innovative Research, Tokyo Institute of Technology, 4259 S2-11 Nagatsuta-cho, Midori-ku, Yokohama 226-8503, Japan
- Graduate School of Life Science and Technology, Tokyo Institute of Technology, 4259 S2-11 Nagatsuta-cho, Midori-ku, Yokohama 226-8503, Japan
| |
Collapse
|
8
|
Fujita N, Girada S, Vogler G, Bodmer R, Kiger AA. PI(4,5)P 2 role in Transverse-tubule membrane formation and muscle function. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.31.578124. [PMID: 38352484 PMCID: PMC10862868 DOI: 10.1101/2024.01.31.578124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/20/2024]
Abstract
Transverse (T)-tubules - vast, tubulated domains of the muscle plasma membrane - are critical to maintain healthy skeletal and heart contractions. How the intricate T-tubule membranes are formed is not well understood, with challenges to systematically interrogate in muscle. We established the use of intact Drosophila larval body wall muscles as an ideal system to discover mechanisms that sculpt and maintain the T-tubule membrane network. A muscle-targeted genetic screen identified specific phosphoinositide lipid regulators necessary for T-tubule organization and muscle function. We show that a PI4KIIIα - Skittles/PIP5K pathway is needed for T-tubule localized PI(4)P to PI(4,5)P 2 synthesis, T-tubule organization, calcium regulation, and muscle and heart rate functions. Muscles deficient for PI4KIIIα or Amphiphysin , the homolog of human BIN1 , similarly exhibited specific loss of transversal T-tubule membranes and dyad junctions, yet retained longitudinal membranes and the associated dyads. Our results highlight the power of live muscle studies, uncovering distinct mechanisms and functions for sub-compartments of the T-tubule network relevant to human myopathy. Summary T-tubules - vast, tubulated domains of the muscle plasma membrane - are critical to maintain skeletal and heart contractions. Fujita et al . establish genetic screens and assays in intact Drosophila muscles that uncover PI(4,5)P 2 regulation critical for T-tubule maintenance and function. Key Findings PI4KIIIα is required for muscle T-tubule formation and larval mobility. A PI4KIIIα-Sktl pathway promotes PI(4)P and PI(4,5)P 2 function at T-tubules. PI4KIIIα is necessary for calcium dynamics and transversal but not longitudinal dyads. Disruption of PI(4,5)P 2 function in fly heart leads to fragmented T-tubules and abnormal heart rate.
Collapse
|
9
|
Giraud Q, Spiegelhalter C, Messaddeq N, Laporte J. MTM1 overexpression prevents and reverts BIN1-related centronuclear myopathy. Brain 2023; 146:4158-4173. [PMID: 37490306 PMCID: PMC10545525 DOI: 10.1093/brain/awad251] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 06/30/2023] [Accepted: 07/06/2023] [Indexed: 07/26/2023] Open
Abstract
Centronuclear and myotubular myopathies (CNM) are rare and severe genetic diseases associated with muscle weakness and atrophy as well as intracellular disorganization of myofibres. The main mutated proteins control lipid and membrane dynamics and are the lipid phosphatase myotubularin (MTM1), and the membrane remodelling proteins amphiphysin 2 (BIN1) and dynamin 2 (DNM2). There is no available therapy. Here, to validate a novel therapeutic strategy for BIN1- and DNM2-CNM, we evaluated adeno-associated virus-mediated MTM1 (AAV-MTM1 ) overexpression in relevant mouse models. Early systemic MTM1 overexpression prevented the development of the CNM pathology in Bin1mck-/- mice, while late intramuscular MTM1 expression partially reverted the established phenotypes after only 4 weeks of treatment. However, AAV-MTM1 injection did not change the DNM2-CNM mouse phenotypes. We investigated the mechanism of the rescue of the myopathy in BIN1-CNM and found that the lipid phosphatase activity of MTM1 was essential for the rescue of muscle atrophy and myofibre hypotrophy but dispensable for the rescue of myofibre disorganization including organelle mis-position and T-tubule defects. Furthermore, the improvement of T-tubule organization correlated with normalization of key regulators of T-tubule morphogenesis, dysferlin and caveolin. Overall, these data support the inclusion of BIN1-CNM patients in an AAV-MTM1 clinical trial.
Collapse
Affiliation(s)
- Quentin Giraud
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS UMR7104, INSERM U1258, Université de Strasbourg, 67404, Illkirch, France
| | - Coralie Spiegelhalter
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS UMR7104, INSERM U1258, Université de Strasbourg, 67404, Illkirch, France
| | - Nadia Messaddeq
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS UMR7104, INSERM U1258, Université de Strasbourg, 67404, Illkirch, France
| | - Jocelyn Laporte
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS UMR7104, INSERM U1258, Université de Strasbourg, 67404, Illkirch, France
| |
Collapse
|
10
|
de Carvalho Neves J, Moschovaki-Filippidou F, Böhm J, Laporte J. DNM2 levels normalization improves muscle phenotypes of a novel mouse model for moderate centronuclear myopathy. MOLECULAR THERAPY. NUCLEIC ACIDS 2023; 33:321-334. [PMID: 37547294 PMCID: PMC10400865 DOI: 10.1016/j.omtn.2023.07.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Accepted: 07/11/2023] [Indexed: 08/08/2023]
Abstract
Dynamin 2 (DNM2) is a ubiquitously expressed GTPase regulating membrane trafficking and cytoskeleton dynamics. Heterozygous dominant mutations in DNM2 cause centronuclear myopathy (CNM), associated with muscle weakness and atrophy and histopathological hallmarks as fiber hypotrophy and organelles mis-position. Different severities range from the severe neonatal onset form to the moderate form with childhood onset and to the mild adult onset form. No therapy is approved for CNM. Here we aimed to validate and rescue a mouse model for the moderate form of DNM2-CNM harboring the common DNM2 R369W missense mutation. Dnm2R369W/+ mice presented with increased DNM2 protein level in muscle and moderate CNM-like phenotypes with force deficit, muscle and fiber hypotrophy, impaired mTOR signaling, and progressive mitochondria and nuclei mis-position with age. Molecular analyses revealed a fiber type switch toward oxidative metabolism correlating with decreased force and alteration of mitophagy markers paralleling mitochondria structural defects. Normalization of DNM2 levels through intramuscular injection of AAV-shDnm2 targeting Dnm2 mRNA significantly improved histopathology and muscle and myofiber hypotrophy. These results showed that the Dnm2R369W/+ mouse is a faithful model for the moderate form of DNM2-CNM and revealed that DNM2 normalization after a short 4-week treatment is sufficient to improve the CNM phenotypes.
Collapse
Affiliation(s)
- Juliana de Carvalho Neves
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS UMR7104, INSERM U1258, 1 rue Laurent Fries, 67404 Illkirch Cedex, France
| | - Foteini Moschovaki-Filippidou
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS UMR7104, INSERM U1258, 1 rue Laurent Fries, 67404 Illkirch Cedex, France
| | - Johann Böhm
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS UMR7104, INSERM U1258, 1 rue Laurent Fries, 67404 Illkirch Cedex, France
| | - Jocelyn Laporte
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS UMR7104, INSERM U1258, 1 rue Laurent Fries, 67404 Illkirch Cedex, France
| |
Collapse
|
11
|
Saavedra P, Dumesic PA, Hu Y, Filine E, Jouandin P, Binari R, Wilensky SE, Rodiger J, Wang H, Chen W, Liu Y, Spiegelman BM, Perrimon N. REPTOR and CREBRF encode key regulators of muscle energy metabolism. Nat Commun 2023; 14:4943. [PMID: 37582831 PMCID: PMC10427696 DOI: 10.1038/s41467-023-40595-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 08/03/2023] [Indexed: 08/17/2023] Open
Abstract
Metabolic flexibility of muscle tissue describes the adaptive capacity to use different energy substrates according to their availability. The disruption of this ability associates with metabolic disease. Here, using a Drosophila model of systemic metabolic dysfunction triggered by yorkie-induced gut tumors, we show that the transcription factor REPTOR is an important regulator of energy metabolism in muscles. We present evidence that REPTOR is activated in muscles of adult flies with gut yorkie-tumors, where it modulates glucose metabolism. Further, in vivo studies indicate that sustained activity of REPTOR is sufficient in wildtype muscles to repress glycolysis and increase tricarboxylic acid (TCA) cycle metabolites. Consistent with the fly studies, higher levels of CREBRF, the mammalian ortholog of REPTOR, reduce glycolysis in mouse myotubes while promoting oxidative metabolism. Altogether, our results define a conserved function for REPTOR and CREBRF as key regulators of muscle energy metabolism.
Collapse
Affiliation(s)
- Pedro Saavedra
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, 02115, USA.
| | - Phillip A Dumesic
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, 02115, USA
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA, 02115, USA
| | - Yanhui Hu
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, 02115, USA
| | - Elizabeth Filine
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, 02115, USA
| | - Patrick Jouandin
- Institut de Recherche en Cancérologie de Montpellier, INSERM, Montpellier, France
| | - Richard Binari
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, 02115, USA
- Howard Hughes Medical Institute, Boston, MA, 02115, USA
| | - Sarah E Wilensky
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, 02115, USA
| | - Jonathan Rodiger
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, 02115, USA
| | - Haiyun Wang
- School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Weihang Chen
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, 02115, USA
| | - Ying Liu
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, 02115, USA
| | - Bruce M Spiegelman
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, 02115, USA
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA, 02115, USA
| | - Norbert Perrimon
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, 02115, USA.
- Howard Hughes Medical Institute, Boston, MA, 02115, USA.
| |
Collapse
|
12
|
Kermani F, Mosqueira M, Peters K, Lemma ED, Rapti K, Grimm D, Bastmeyer M, Laugsch M, Hecker M, Ullrich ND. Membrane remodelling triggers maturation of excitation-contraction coupling in 3D-shaped human-induced pluripotent stem cell-derived cardiomyocytes. Basic Res Cardiol 2023; 118:13. [PMID: 36988697 PMCID: PMC10060306 DOI: 10.1007/s00395-023-00984-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 03/14/2023] [Accepted: 03/14/2023] [Indexed: 03/30/2023]
Abstract
The prospective use of human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CM) for cardiac regenerative medicine strongly depends on the electro-mechanical properties of these cells, especially regarding the Ca2+-dependent excitation-contraction (EC) coupling mechanism. Currently, the immature structural and functional features of hiPSC-CM limit the progression towards clinical applications. Here, we show that a specific microarchitecture is essential for functional maturation of hiPSC-CM. Structural remodelling towards a cuboid cell shape and induction of BIN1, a facilitator of membrane invaginations, lead to transverse (t)-tubule-like structures. This transformation brings two Ca2+ channels critical for EC coupling in close proximity, the L-type Ca2+ channel at the sarcolemma and the ryanodine receptor at the sarcoplasmic reticulum. Consequently, the Ca2+-dependent functional interaction of these channels becomes more efficient, leading to improved spatio-temporal synchronisation of Ca2+ transients and higher EC coupling gain. Thus, functional maturation of hiPSC-cardiomyocytes by optimised cell microarchitecture needs to be considered for future cardiac regenerative approaches.
Collapse
Affiliation(s)
- Fatemeh Kermani
- Department of Cardiovascular Physiology, Heidelberg University, Heidelberg, Germany
| | - Matias Mosqueira
- Department of Cardiovascular Physiology, Heidelberg University, Heidelberg, Germany
| | - Kyra Peters
- Department of Cardiovascular Physiology, Heidelberg University, Heidelberg, Germany
| | - Enrico D Lemma
- Zoological Institute, Cell and Neurobiology, Karlsruhe Institute of Technology (KIT), Karlsruhe, Germany
- Department of Engineering, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Kleopatra Rapti
- Department of Infectious Diseases/Virology, Section Viral Vector Technologies, Heidelberg University, Heidelberg, Germany
| | - Dirk Grimm
- Department of Infectious Diseases/Virology, Section Viral Vector Technologies, Heidelberg University, Heidelberg, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Heidelberg/Mannheim, Heidelberg, Germany
- German Center for Infection Research (DZIF), Partner Site Heidelberg, Heidelberg, Germany
| | - Martin Bastmeyer
- Zoological Institute, Cell and Neurobiology, Karlsruhe Institute of Technology (KIT), Karlsruhe, Germany
- Institute of Biological and Chemical Systems-Biological information processing (IBCS-BIP), Karlsruhe Institute of Technology (KIT), Eggenstein-Leopoldshafen, Germany
- Research Bridge (Synthetic Biology), Heidelberg-Karlsruhe Research Partnership (HEiKA), Heidelberg University and Karlsruhe Institute of Technology, Heidelberg, Germany
| | - Magdalena Laugsch
- Institute of Human Genetics, Heidelberg University, Heidelberg, Germany
| | - Markus Hecker
- Department of Cardiovascular Physiology, Heidelberg University, Heidelberg, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Heidelberg/Mannheim, Heidelberg, Germany
| | - Nina D Ullrich
- Department of Cardiovascular Physiology, Heidelberg University, Heidelberg, Germany.
- German Center for Cardiovascular Research (DZHK), Partner Site Heidelberg/Mannheim, Heidelberg, Germany.
- Research Bridge (Synthetic Biology), Heidelberg-Karlsruhe Research Partnership (HEiKA), Heidelberg University and Karlsruhe Institute of Technology, Heidelberg, Germany.
| |
Collapse
|
13
|
Laiman J, Hsu YJ, Loh J, Tang WC, Chuang MC, Liu HK, Yang WS, Chen BC, Chuang LM, Chang YC, Liu YW. GSK3α phosphorylates dynamin-2 to promote GLUT4 endocytosis in muscle cells. J Cell Biol 2023; 222:e202102119. [PMID: 36445308 PMCID: PMC9712776 DOI: 10.1083/jcb.202102119] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 08/17/2022] [Accepted: 10/31/2022] [Indexed: 12/02/2022] Open
Abstract
Insulin-stimulated translocation of glucose transporter 4 (GLUT4) to plasma membrane of skeletal muscle is critical for postprandial glucose uptake; however, whether the internalization of GLUT4 is also regulated by insulin signaling remains unclear. Here, we discover that the activity of dynamin-2 (Dyn2) in catalyzing GLUT4 endocytosis is negatively regulated by insulin signaling in muscle cells. Mechanistically, the fission activity of Dyn2 is inhibited by binding with the SH3 domain of Bin1. In the absence of insulin, GSK3α phosphorylates Dyn2 to relieve the inhibition of Bin1 and promotes endocytosis. Conversely, insulin signaling inactivates GSK3α and leads to attenuated GLUT4 internalization. Furthermore, the isoform-specific pharmacological inhibition of GSK3α significantly improves insulin sensitivity and glucose tolerance in diet-induced insulin-resistant mice. Together, we identify a new role of GSK3α in insulin-stimulated glucose disposal by regulating Dyn2-mediated GLUT4 endocytosis in muscle cells. These results highlight the isoform-specific function of GSK3α on membrane trafficking and its potential as a therapeutic target for metabolic disorders.
Collapse
Affiliation(s)
- Jessica Laiman
- Institute of Molecular Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Yen-Jung Hsu
- Institute of Molecular Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Julie Loh
- Institute of Molecular Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Wei-Chun Tang
- ResearchCenter for Applied Sciences, Academia Sinica, Taipei, Taiwan
| | - Mei-Chun Chuang
- Institute of Molecular Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Hui-Kang Liu
- National Research Institute of Chinese Medicine, Ministry of Health and Welfare, Taipei, Taiwan
- Program in the Clinical Drug Development of Herbal Medicine, Taipei Medical University, Taipei, Taiwan
| | - Wei-Shun Yang
- Division of Nephrology, Department of Internal Medicine, National Taiwan University Hospital, Hsin-Chu Branch, Hsin-Chu, Taiwan
| | - Bi-Chang Chen
- ResearchCenter for Applied Sciences, Academia Sinica, Taipei, Taiwan
| | - Lee-Ming Chuang
- Institute of Molecular Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Yi-Cheng Chang
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
- Institute of Medical Genomics and Proteomics, College of Medicine, National Taiwan University, Taipei, Taiwan
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Ya-Wen Liu
- Institute of Molecular Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
- Center of Precision Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
14
|
Mackrill JJ. Evolution of the cardiac dyad. Philos Trans R Soc Lond B Biol Sci 2022; 377:20210329. [PMID: 36189805 PMCID: PMC9527923 DOI: 10.1098/rstb.2021.0329] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 12/22/2021] [Indexed: 12/30/2022] Open
Abstract
Cardiac dyads are the site of communication between the sarcoplasmic reticulum (SR) and infoldings of the sarcolemma called transverse-tubules (TT). During heart excitation-contraction coupling, Ca2+-influx through L-type Ca2+ channels in the TT is amplified by release of Ca2+-from the SR via type 2 ryanodine receptors, activating the contractile apparatus. Key proteins involved in cardiac dyad function are bridging integrator 1 (BIN1), junctophilin 2 and caveolin 3. The work presented here aims to reconstruct the evolutionary history of the cardiac dyad, by surveying the scientific literature for ultrastructural evidence of these junctions across all animal taxa; phylogenetically reconstructing the evolutionary history of BIN1; and by comparing peptide motifs involved in TT formation by this protein across metazoans. Key findings are that cardiac dyads have been identified in mammals, arthropods and molluscs, but not in other animals. Vertebrate BIN1 does not group with members of this protein family from other taxa, suggesting that invertebrate BINs are paralogues rather orthologues of this gene. Comparisons of BIN1 peptide sequences of mammals with those of other vertebrates reveals novel features that might contribute to TT and dyad formation. The analyses presented here suggest that the cardiac dyad evolved independently several times during metazoan evolution: an unexpected observation given the diversity of heart structure and function between different animal taxa. This article is part of the theme issue 'The cardiomyocyte: new revelations on the interplay between architecture and function in growth, health, and disease'.
Collapse
Affiliation(s)
- John James Mackrill
- Department of Physiology, School of Medicine, University College Cork, Western Gateway Building, Western Road, Cork T12 XF62, Republic of Ireland
| |
Collapse
|
15
|
Mallik B, Bhat S, Kumar V. Role of Bin‐Amphiphysin‐Rvs (BAR) domain proteins in mediating neuronal signaling and disease. Synapse 2022; 76:e22248. [DOI: 10.1002/syn.22248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 05/13/2022] [Accepted: 07/18/2022] [Indexed: 11/06/2022]
Affiliation(s)
- Bhagaban Mallik
- Department of Biological Sciences Indian Institute of Science Education and Research (IISER) Bhopal Indore Bypass Road Bhopal Madhya Pradesh 462 066 India
| | - Sajad Bhat
- Department of Biological Sciences Indian Institute of Science Education and Research (IISER) Bhopal Indore Bypass Road Bhopal Madhya Pradesh 462 066 India
| | - Vimlesh Kumar
- Department of Biological Sciences Indian Institute of Science Education and Research (IISER) Bhopal Indore Bypass Road Bhopal Madhya Pradesh 462 066 India
| |
Collapse
|
16
|
Fujise K, Noguchi S, Takeda T. Centronuclear Myopathy Caused by Defective Membrane Remodelling of Dynamin 2 and BIN1 Variants. Int J Mol Sci 2022; 23:ijms23116274. [PMID: 35682949 PMCID: PMC9181712 DOI: 10.3390/ijms23116274] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Revised: 05/31/2022] [Accepted: 06/01/2022] [Indexed: 02/01/2023] Open
Abstract
Centronuclear myopathy (CNM) is a congenital myopathy characterised by centralised nuclei in skeletal myofibers. T-tubules, sarcolemmal invaginations required for excitation-contraction coupling, are disorganised in the skeletal muscles of CNM patients. Previous studies showed that various endocytic proteins are involved in T-tubule biogenesis and their dysfunction is tightly associated with CNM pathogenesis. DNM2 and BIN1 are two causative genes for CNM that encode essential membrane remodelling proteins in endocytosis, dynamin 2 and BIN1, respectively. In this review, we overview the functions of dynamin 2 and BIN1 in T-tubule biogenesis and discuss how their dysfunction in membrane remodelling leads to CNM pathogenesis.
Collapse
Affiliation(s)
- Kenshiro Fujise
- Departments of Neuroscience and Cell Biology, Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT 06520-8001, USA;
| | - Satoru Noguchi
- National Institute of Neuroscience, National Center of Neurology and Psychiatry (NCNP), Tokyo 187-8502, Japan;
| | - Tetsuya Takeda
- Department of Biochemistry, Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Shikata-cho 2-5-1, Kita-ku, Okayama 700-8558, Japan
- Correspondence: ; Tel.: +81-86-235-7125; Fax: +81-86-235-7126
| |
Collapse
|
17
|
Silva-Rojas R, Nattarayan V, Jaque-Fernandez F, Gomez-Oca R, Menuet A, Reiss D, Goret M, Messaddeq N, Lionello VM, Kretz C, Cowling BS, Jacquemond V, Laporte J. Mice with muscle-specific deletion of Bin1 recapitulate centronuclear myopathy and acute downregulation of dynamin 2 improves their phenotypes. Mol Ther 2022; 30:868-880. [PMID: 34371181 PMCID: PMC8821932 DOI: 10.1016/j.ymthe.2021.08.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 07/07/2021] [Accepted: 07/29/2021] [Indexed: 02/04/2023] Open
Abstract
Mutations in the BIN1 (Bridging Interactor 1) gene, encoding the membrane remodeling protein amphiphysin 2, cause centronuclear myopathy (CNM) associated with severe muscle weakness and myofiber disorganization and hypotrophy. There is no available therapy, and the validation of therapeutic proof of concept is impaired by the lack of a faithful and easy-to-handle mammalian model. Here, we generated and characterized the Bin1mck-/- mouse through Bin1 knockout in skeletal muscle. Bin1mck-/- mice were viable, unlike the constitutive Bin1 knockout, and displayed decreased muscle force and most histological hallmarks of CNM, including myofiber hypotrophy and intracellular disorganization. Notably, Bin1mck-/- myofibers presented strong defects in mitochondria and T-tubule networks associated with deficient calcium homeostasis and excitation-contraction coupling at the triads, potentially representing the main pathomechanisms. Systemic injection of antisense oligonucleotides (ASOs) targeting Dnm2 (Dynamin 2), which codes for dynamin 2, a BIN1 binding partner regulating membrane fission and mutated in other forms of CNM, improved muscle force and normalized the histological Bin1mck-/- phenotypes within 5 weeks. Overall, we generated a faithful mammalian model for CNM linked to BIN1 defects and validated Dnm2 ASOs as a first translatable approach to efficiently treat BIN1-CNM.
Collapse
Affiliation(s)
- Roberto Silva-Rojas
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS UMR7104, INSERM U1258, Université de Strasbourg, 67404 Illkirch, France
| | - Vasugi Nattarayan
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS UMR7104, INSERM U1258, Université de Strasbourg, 67404 Illkirch, France
| | - Francisco Jaque-Fernandez
- Univ Lyon, Université Claude Bernard Lyon 1, CNRS UMR5310, INSERM U1217, Institut NeuroMyoGène, 69373 Lyon, France
| | - Raquel Gomez-Oca
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS UMR7104, INSERM U1258, Université de Strasbourg, 67404 Illkirch, France,Dynacure, 67404 Illkirch, France
| | - Alexia Menuet
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS UMR7104, INSERM U1258, Université de Strasbourg, 67404 Illkirch, France
| | - David Reiss
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS UMR7104, INSERM U1258, Université de Strasbourg, 67404 Illkirch, France
| | - Marie Goret
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS UMR7104, INSERM U1258, Université de Strasbourg, 67404 Illkirch, France
| | - Nadia Messaddeq
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS UMR7104, INSERM U1258, Université de Strasbourg, 67404 Illkirch, France
| | - Valentina M. Lionello
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS UMR7104, INSERM U1258, Université de Strasbourg, 67404 Illkirch, France
| | - Christine Kretz
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS UMR7104, INSERM U1258, Université de Strasbourg, 67404 Illkirch, France
| | - Belinda S. Cowling
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS UMR7104, INSERM U1258, Université de Strasbourg, 67404 Illkirch, France,Dynacure, 67404 Illkirch, France
| | - Vincent Jacquemond
- Univ Lyon, Université Claude Bernard Lyon 1, CNRS UMR5310, INSERM U1217, Institut NeuroMyoGène, 69373 Lyon, France
| | - Jocelyn Laporte
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS UMR7104, INSERM U1258, Université de Strasbourg, 67404 Illkirch, France,Corresponding author: Jocelyn Laporte, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS UMR7104, INSERM U1258, Université de Strasbourg, 67404 Illkirch, France.
| |
Collapse
|
18
|
Nusier M, Shah AK, Dhalla NS. Structure-Function Relationships and Modifications of Cardiac Sarcoplasmic Reticulum Ca2+-Transport. Physiol Res 2022; 70:S443-S470. [DOI: 10.33549/physiolres.934805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Sarcoplasmic reticulum (SR) is a specialized tubular network, which not only maintains the intracellular concentration of Ca2+ at a low level but is also known to release and accumulate Ca2+ for the occurrence of cardiac contraction and relaxation, respectively. This subcellular organelle is composed of several phospholipids and different Ca2+-cycling, Ca2+-binding and regulatory proteins, which work in a coordinated manner to determine its function in cardiomyocytes. Some of the major proteins in the cardiac SR membrane include Ca2+-pump ATPase (SERCA2), Ca2+-release protein (ryanodine receptor), calsequestrin (Ca2+-binding protein) and phospholamban (regulatory protein). The phosphorylation of SR Ca2+-cycling proteins by protein kinase A or Ca2+-calmodulin kinase (directly or indirectly) has been demonstrated to augment SR Ca2+-release and Ca2+-uptake activities and promote cardiac contraction and relaxation functions. The activation of phospholipases and proteases as well as changes in different gene expressions under different pathological conditions have been shown to alter the SR composition and produce Ca2+-handling abnormalities in cardiomyocytes for the development of cardiac dysfunction. The post-translational modifications of SR Ca2+ cycling proteins by processes such as oxidation, nitrosylation, glycosylation, lipidation, acetylation, sumoylation, and O GlcNacylation have also been reported to affect the SR Ca2+ release and uptake activities as well as cardiac contractile activity. The SR function in the heart is also influenced in association with changes in cardiac performance by several hormones including thyroid hormones and adiponectin as well as by exercise-training. On the basis of such observations, it is suggested that both Ca2+-cycling and regulatory proteins in the SR membranes are intimately involved in determining the status of cardiac function and are thus excellent targets for drug development for the treatment of heart disease.
Collapse
Affiliation(s)
| | | | - NS Dhalla
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen, Research Centre, 351 Tache Avenue, Winnipeg, MB, R2H 2A6 Canada.
| |
Collapse
|
19
|
Zhou J, Singh N, Monnier C, Marszalec W, Gao L, Jin J, Frisk M, Louch WE, Verma S, Krishnamurthy P, Nico E, Mulla M, Aistrup GL, Kishore R, Wasserstrom JA. Phosphatidylinositol-4,5-Bisphosphate Binding to Amphiphysin-II Modulates T-Tubule Remodeling: Implications for Heart Failure. Front Physiol 2022; 12:782767. [PMID: 35002765 PMCID: PMC8733645 DOI: 10.3389/fphys.2021.782767] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 11/19/2021] [Indexed: 11/13/2022] Open
Abstract
BIN1 (amphyphysin-II) is a structural protein involved in T-tubule (TT) formation and phosphatidylinositol-4,5-bisphosphate (PIP2) is responsible for localization of BIN1 to sarcolemma. The goal of this study was to determine if PIP2-mediated targeting of BIN1 to sarcolemma is compromised during the development of heart failure (HF) and is responsible for TT remodeling. Immunohistochemistry showed co-localization of BIN1, Cav1.2, PIP2, and phospholipase-Cβ1 (PLCβ1) in TTs in normal rat and human ventricular myocytes. PIP2 levels were reduced in spontaneously hypertensive rats during HF progression compared to age-matched controls. A PIP Strip assay of two native mouse cardiac-specific isoforms of BIN1 including the longest (cardiac BIN1 #4) and shortest (cardiac BIN1 #1) isoforms as well human skeletal BIN1 showed that all bound PIP2. In addition, overexpression of all three BIN1 isoforms caused tubule formation in HL-1 cells. A triple-lysine motif in a short loop segment between two helices was mutated and replaced by negative charges which abolished tubule formation, suggesting a possible location for PIP2 interaction aside from known consensus binding sites. Pharmacological PIP2 depletion in rat ventricular myocytes caused TT loss and was associated with changes in Ca2+ release typically found in myocytes during HF, including a higher variability in release along the cell length and a slowing in rise time, time to peak, and decay time in treated myocytes. These results demonstrate that depletion of PIP2 can lead to TT disruption and suggest that PIP2 interaction with cardiac BIN1 is required for TT maintenance and function.
Collapse
Affiliation(s)
- Junlan Zhou
- Department of Medicine (Cardiology), Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Neha Singh
- Department of Medicine (Cardiology), Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Chloe Monnier
- Department of Medicine (Cardiology), Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - William Marszalec
- Department of Medicine (Cardiology), Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Li Gao
- Department of Medicine (Cardiology), Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Jing Jin
- Department of Medicine (Cardiology), Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Michael Frisk
- Institute for Experimental Medical Research (IEMR), Oslo University Hospital, Oslo, Norway.,K. G. Jebsen Cardiac Research Center, University of Oslo, Oslo, Norway
| | - William E Louch
- Institute for Experimental Medical Research (IEMR), Oslo University Hospital, Oslo, Norway.,K. G. Jebsen Cardiac Research Center, University of Oslo, Oslo, Norway
| | - Suresh Verma
- Department of Medicine (Cardiology), Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Prasanna Krishnamurthy
- Department of Medicine (Cardiology), Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Elsa Nico
- Department of Medicine (Cardiology), Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Maaz Mulla
- Department of Medicine (Cardiology), Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Gary L Aistrup
- Department of Medicine (Cardiology), Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Raj Kishore
- Department of Medicine (Cardiology), Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - J Andrew Wasserstrom
- Department of Medicine (Cardiology), Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| |
Collapse
|
20
|
Moulton MJ, Barish S, Ralhan I, Chang J, Goodman LD, Harland JG, Marcogliese PC, Johansson JO, Ioannou MS, Bellen HJ. Neuronal ROS-induced glial lipid droplet formation is altered by loss of Alzheimer's disease-associated genes. Proc Natl Acad Sci U S A 2021; 118:e2112095118. [PMID: 34949639 PMCID: PMC8719885 DOI: 10.1073/pnas.2112095118] [Citation(s) in RCA: 92] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 11/11/2021] [Indexed: 01/02/2023] Open
Abstract
A growing list of Alzheimer's disease (AD) genetic risk factors is being identified, but the contribution of each variant to disease mechanism remains largely unknown. We have previously shown that elevated levels of reactive oxygen species (ROS) induces lipid synthesis in neurons leading to the sequestration of peroxidated lipids in glial lipid droplets (LD), delaying neurotoxicity. This neuron-to-glia lipid transport is APOD/E-dependent. To identify proteins that modulate these neuroprotective effects, we tested the role of AD risk genes in ROS-induced LD formation and demonstrate that several genes impact neuroprotective LD formation, including homologs of human ABCA1, ABCA7, VLDLR, VPS26, VPS35, AP2A, PICALM, and CD2AP Our data also show that ROS enhances Aβ42 phenotypes in flies and mice. Finally, a peptide agonist of ABCA1 restores glial LD formation in a humanized APOE4 fly model, highlighting a potentially therapeutic avenue to prevent ROS-induced neurotoxicity. This study places many AD genetic risk factors in a ROS-induced neuron-to-glia lipid transfer pathway with a critical role in protecting against neurotoxicity.
Collapse
Affiliation(s)
- Matthew J Moulton
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX 77030
| | - Scott Barish
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030
| | - Isha Ralhan
- Department of Physiology, University of Alberta, Edmonton, AB T6G 2R3, Canada
- Group on Molecular and Cell Biology of Lipids, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - Jinlan Chang
- Department of Physiology, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - Lindsey D Goodman
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX 77030
| | - Jake G Harland
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX 77030
| | - Paul C Marcogliese
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX 77030
| | | | - Maria S Ioannou
- Department of Physiology, University of Alberta, Edmonton, AB T6G 2R3, Canada
- Group on Molecular and Cell Biology of Lipids, University of Alberta, Edmonton, AB T6G 2R3, Canada
- Department of Cell Biology, University of Alberta, Edmonton, AB T6G 2R3, Canada
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - Hugo J Bellen
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030;
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX 77030
- Program in Developmental Biology, Baylor College of Medicine, Houston TX 77030
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030
| |
Collapse
|
21
|
Lysine acetylation regulates the interaction between proteins and membranes. Nat Commun 2021; 12:6466. [PMID: 34753925 PMCID: PMC8578602 DOI: 10.1038/s41467-021-26657-2] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Accepted: 09/30/2021] [Indexed: 11/23/2022] Open
Abstract
Lysine acetylation regulates the function of soluble proteins in vivo, yet it remains largely unexplored whether lysine acetylation regulates membrane protein function. Here, we use bioinformatics, biophysical analysis of recombinant proteins, live-cell fluorescent imaging and genetic manipulation of Drosophila to explore lysine acetylation in peripheral membrane proteins. Analysis of 50 peripheral membrane proteins harboring BAR, PX, C2, or EHD membrane-binding domains reveals that lysine acetylation predominates in membrane-interaction regions. Acetylation and acetylation-mimicking mutations in three test proteins, amphiphysin, EHD2, and synaptotagmin1, strongly reduce membrane binding affinity, attenuate membrane remodeling in vitro and alter subcellular localization. This effect is likely due to the loss of positive charge, which weakens interactions with negatively charged membranes. In Drosophila, acetylation-mimicking mutations of amphiphysin cause severe disruption of T-tubule organization and yield a flightless phenotype. Our data provide mechanistic insights into how lysine acetylation regulates membrane protein function, potentially impacting a plethora of membrane-related processes. Lysine acetylation regulates the function of soluble proteins in vivo, yet it remains largely unexplored whether lysine acetylation regulates the function of membrane proteins. Here, the authors map lysine acetylation predominantly in membrane-interaction regions in peripheral membrane proteins and show with three candidate proteins how lysine acetylation is a regulator of membrane protein function.
Collapse
|
22
|
Gómez-Oca R, Cowling BS, Laporte J. Common Pathogenic Mechanisms in Centronuclear and Myotubular Myopathies and Latest Treatment Advances. Int J Mol Sci 2021; 22:11377. [PMID: 34768808 PMCID: PMC8583656 DOI: 10.3390/ijms222111377] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Accepted: 10/18/2021] [Indexed: 01/18/2023] Open
Abstract
Centronuclear myopathies (CNM) are rare congenital disorders characterized by muscle weakness and structural defects including fiber hypotrophy and organelle mispositioning. The main CNM forms are caused by mutations in: the MTM1 gene encoding the phosphoinositide phosphatase myotubularin (myotubular myopathy), the DNM2 gene encoding the mechanoenzyme dynamin 2, the BIN1 gene encoding the membrane curvature sensing amphiphysin 2, and the RYR1 gene encoding the skeletal muscle calcium release channel/ryanodine receptor. MTM1, BIN1, and DNM2 proteins are involved in membrane remodeling and trafficking, while RyR1 directly regulates excitation-contraction coupling (ECC). Several CNM animal models have been generated or identified, which confirm shared pathological anomalies in T-tubule remodeling, ECC, organelle mispositioning, protein homeostasis, neuromuscular junction, and muscle regeneration. Dynamin 2 plays a crucial role in CNM physiopathology and has been validated as a common therapeutic target for three CNM forms. Indeed, the promising results in preclinical models set up the basis for ongoing clinical trials. Another two clinical trials to treat myotubular myopathy by MTM1 gene therapy or tamoxifen repurposing are also ongoing. Here, we review the contribution of the different CNM models to understanding physiopathology and therapy development with a focus on the commonly dysregulated pathways and current therapeutic targets.
Collapse
Affiliation(s)
- Raquel Gómez-Oca
- Department of Translational Medicine and Neurogenetics, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), 67400 Illkirch, France;
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1258, 67400 Illkirch, France
- Centre National de la Recherche Scientifique (CNRS), UMR7104, 67400 Illkirch, France
- Strasbourg University, 67081 Strasbourg, France
- Dynacure, 67400 Illkirch, France;
| | | | - Jocelyn Laporte
- Department of Translational Medicine and Neurogenetics, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), 67400 Illkirch, France;
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1258, 67400 Illkirch, France
- Centre National de la Recherche Scientifique (CNRS), UMR7104, 67400 Illkirch, France
- Strasbourg University, 67081 Strasbourg, France
| |
Collapse
|
23
|
Johnson AE, Orr BO, Fetter RD, Moughamian AJ, Primeaux LA, Geier EG, Yokoyama JS, Miller BL, Davis GW. SVIP is a molecular determinant of lysosomal dynamic stability, neurodegeneration and lifespan. Nat Commun 2021; 12:513. [PMID: 33479240 PMCID: PMC7820495 DOI: 10.1038/s41467-020-20796-8] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 12/17/2020] [Indexed: 12/12/2022] Open
Abstract
Missense mutations in Valosin-Containing Protein (VCP) are linked to diverse degenerative diseases including IBMPFD, amyotrophic lateral sclerosis (ALS), muscular dystrophy and Parkinson's disease. Here, we characterize a VCP-binding co-factor (SVIP) that specifically recruits VCP to lysosomes. SVIP is essential for lysosomal dynamic stability and autophagosomal-lysosomal fusion. SVIP mutations cause muscle wasting and neuromuscular degeneration while muscle-specific SVIP over-expression increases lysosomal abundance and is sufficient to extend lifespan in a context, stress-dependent manner. We also establish multiple links between SVIP and VCP-dependent disease in our Drosophila model system. A biochemical screen identifies a disease-causing VCP mutation that prevents SVIP binding. Conversely, over-expression of an SVIP mutation that prevents VCP binding is deleterious. Finally, we identify a human SVIP mutation and confirm the pathogenicity of this mutation in our Drosophila model. We propose a model for VCP disease based on the differential, co-factor-dependent recruitment of VCP to intracellular organelles.
Collapse
Affiliation(s)
- Alyssa E Johnson
- Department of Biochemistry and Biophysics, Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, San Francisco, CA, 94158, USA
- Department of Biological Sciences, Louisiana State University, Baton Rouge, LA, 70803, USA
| | - Brian O Orr
- Department of Biochemistry and Biophysics, Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, San Francisco, CA, 94158, USA
| | - Richard D Fetter
- Department of Biochemistry and Biophysics, Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, San Francisco, CA, 94158, USA
| | - Armen J Moughamian
- Department of Biochemistry and Biophysics, Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, San Francisco, CA, 94158, USA
- Department of Neurology, University of California, San Francisco, San Francisco, CA, 94158, USA
| | - Logan A Primeaux
- Department of Biological Sciences, Louisiana State University, Baton Rouge, LA, 70803, USA
| | - Ethan G Geier
- Department of Biological Sciences, Louisiana State University, Baton Rouge, LA, 70803, USA
- Department of Neurology, Memory and Aging Center, University of California, San Francisco, San Francisco, CA, 94158, USA
| | - Jennifer S Yokoyama
- Department of Neurology, Memory and Aging Center, University of California, San Francisco, San Francisco, CA, 94158, USA
| | - Bruce L Miller
- Department of Neurology, Memory and Aging Center, University of California, San Francisco, San Francisco, CA, 94158, USA
| | - Graeme W Davis
- Department of Biochemistry and Biophysics, Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, San Francisco, CA, 94158, USA.
| |
Collapse
|
24
|
Kaya-Çopur A, Marchiano F, Hein MY, Alpern D, Russeil J, Luis NM, Mann M, Deplancke B, Habermann BH, Schnorrer F. The Hippo pathway controls myofibril assembly and muscle fiber growth by regulating sarcomeric gene expression. eLife 2021; 10:e63726. [PMID: 33404503 PMCID: PMC7815313 DOI: 10.7554/elife.63726] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 01/05/2021] [Indexed: 12/30/2022] Open
Abstract
Skeletal muscles are composed of gigantic cells called muscle fibers, packed with force-producing myofibrils. During development, the size of individual muscle fibers must dramatically enlarge to match with skeletal growth. How muscle growth is coordinated with growth of the contractile apparatus is not understood. Here, we use the large Drosophila flight muscles to mechanistically decipher how muscle fiber growth is controlled. We find that regulated activity of core members of the Hippo pathway is required to support flight muscle growth. Interestingly, we identify Dlg5 and Slmap as regulators of the STRIPAK phosphatase, which negatively regulates Hippo to enable post-mitotic muscle growth. Mechanistically, we show that the Hippo pathway controls timing and levels of sarcomeric gene expression during development and thus regulates the key components that physically mediate muscle growth. Since Dlg5, STRIPAK and the Hippo pathway are conserved a similar mechanism may contribute to muscle or cardiomyocyte growth in humans.
Collapse
Affiliation(s)
- Aynur Kaya-Çopur
- Aix Marseille University, CNRS, IBDM, Turing Center for Living SystemsMarseilleFrance
- Max Planck Institute of BiochemistryMartinsriedGermany
| | - Fabio Marchiano
- Aix Marseille University, CNRS, IBDM, Turing Center for Living SystemsMarseilleFrance
| | - Marco Y Hein
- Max Planck Institute of BiochemistryMartinsriedGermany
| | - Daniel Alpern
- Institute of Bioengineering, School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL)LausanneSwitzerland
| | - Julie Russeil
- Institute of Bioengineering, School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL)LausanneSwitzerland
| | - Nuno Miguel Luis
- Aix Marseille University, CNRS, IBDM, Turing Center for Living SystemsMarseilleFrance
| | - Matthias Mann
- Max Planck Institute of BiochemistryMartinsriedGermany
| | - Bart Deplancke
- Institute of Bioengineering, School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL)LausanneSwitzerland
| | - Bianca H Habermann
- Aix Marseille University, CNRS, IBDM, Turing Center for Living SystemsMarseilleFrance
| | - Frank Schnorrer
- Aix Marseille University, CNRS, IBDM, Turing Center for Living SystemsMarseilleFrance
- Max Planck Institute of BiochemistryMartinsriedGermany
| |
Collapse
|
25
|
Prokic I, Cowling BS, Kutchukian C, Kretz C, Tasfaout H, Gache V, Hergueux J, Wendling O, Ferry A, Toussaint A, Gavriilidis C, Nattarayan V, Koch C, Lainé J, Combe R, Tiret L, Jacquemond V, Pilot-Storck F, Laporte J. Differential physiological roles for BIN1 isoforms in skeletal muscle development, function and regeneration. Dis Model Mech 2020; 13:dmm044354. [PMID: 32994313 PMCID: PMC7710016 DOI: 10.1242/dmm.044354] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 08/27/2020] [Indexed: 01/09/2023] Open
Abstract
Skeletal muscle development and regeneration are tightly regulated processes. How the intracellular organization of muscle fibers is achieved during these steps is unclear. Here, we focus on the cellular and physiological roles of amphiphysin 2 (BIN1), a membrane remodeling protein mutated in both congenital and adult centronuclear myopathies (CNM), that is ubiquitously expressed and has skeletal muscle-specific isoforms. We created and characterized constitutive muscle-specific and inducible Bin1 homozygous and heterozygous knockout mice targeting either ubiquitous or muscle-specific isoforms. Constitutive Bin1-deficient mice died at birth from lack of feeding due to a skeletal muscle defect. T-tubules and other organelles were misplaced and altered, supporting a general early role for BIN1 in intracellular organization, in addition to membrane remodeling. Although restricted deletion of Bin1 in unchallenged adult muscles had no impact, the forced switch from the muscle-specific isoforms to the ubiquitous isoforms through deletion of the in-frame muscle-specific exon delayed muscle regeneration. Thus, ubiquitous BIN1 function is necessary for muscle development and function, whereas its muscle-specific isoforms fine tune muscle regeneration in adulthood, supporting that BIN1 CNM with congenital onset are due to developmental defects, whereas later onset may be due to regeneration defects.
Collapse
Affiliation(s)
- Ivana Prokic
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Department of Translational Medicine, 67404 Illkirch, France
- Centre National de la Recherche Scientifique (CNRS), UMR7104, 67404 Illkirch, France
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1258, 67404 Illkirch, France
- Université de Strasbourg, 67404 Illkirch, France
| | - Belinda S Cowling
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Department of Translational Medicine, 67404 Illkirch, France
- Centre National de la Recherche Scientifique (CNRS), UMR7104, 67404 Illkirch, France
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1258, 67404 Illkirch, France
- Université de Strasbourg, 67404 Illkirch, France
| | - Candice Kutchukian
- Université Lyon, Université Claude Bernard Lyon 1, CNRS UMR-5310, INSERM U-1217, Institut NeuroMyoGène, 8 Avenue Rockefeller, 69373 Lyon, France
| | - Christine Kretz
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Department of Translational Medicine, 67404 Illkirch, France
- Centre National de la Recherche Scientifique (CNRS), UMR7104, 67404 Illkirch, France
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1258, 67404 Illkirch, France
- Université de Strasbourg, 67404 Illkirch, France
| | - Hichem Tasfaout
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Department of Translational Medicine, 67404 Illkirch, France
- Centre National de la Recherche Scientifique (CNRS), UMR7104, 67404 Illkirch, France
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1258, 67404 Illkirch, France
- Université de Strasbourg, 67404 Illkirch, France
| | - Vincent Gache
- Université Lyon, Université Claude Bernard Lyon 1, CNRS UMR-5310, INSERM U-1217, Institut NeuroMyoGène, 8 Avenue Rockefeller, 69373 Lyon, France
| | - Josiane Hergueux
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Department of Translational Medicine, 67404 Illkirch, France
- Centre National de la Recherche Scientifique (CNRS), UMR7104, 67404 Illkirch, France
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1258, 67404 Illkirch, France
- Université de Strasbourg, 67404 Illkirch, France
| | - Olivia Wendling
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Department of Translational Medicine, 67404 Illkirch, France
- Centre National de la Recherche Scientifique (CNRS), UMR7104, 67404 Illkirch, France
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1258, 67404 Illkirch, France
- Université de Strasbourg, 67404 Illkirch, France
| | - Arnaud Ferry
- Sorbonne Université, INSERM, Institute of Myology, Centre of Research in Myology, Unité Mixte de Recherche (UMRS) 794, 75013 Paris, France
| | - Anne Toussaint
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Department of Translational Medicine, 67404 Illkirch, France
- Centre National de la Recherche Scientifique (CNRS), UMR7104, 67404 Illkirch, France
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1258, 67404 Illkirch, France
- Université de Strasbourg, 67404 Illkirch, France
| | - Christos Gavriilidis
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Department of Translational Medicine, 67404 Illkirch, France
- Centre National de la Recherche Scientifique (CNRS), UMR7104, 67404 Illkirch, France
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1258, 67404 Illkirch, France
- Université de Strasbourg, 67404 Illkirch, France
| | - Vasugi Nattarayan
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Department of Translational Medicine, 67404 Illkirch, France
- Centre National de la Recherche Scientifique (CNRS), UMR7104, 67404 Illkirch, France
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1258, 67404 Illkirch, France
- Université de Strasbourg, 67404 Illkirch, France
| | - Catherine Koch
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Department of Translational Medicine, 67404 Illkirch, France
- Centre National de la Recherche Scientifique (CNRS), UMR7104, 67404 Illkirch, France
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1258, 67404 Illkirch, France
- Université de Strasbourg, 67404 Illkirch, France
| | - Jeanne Lainé
- Sorbonne Université, INSERM, Institute of Myology, Centre of Research in Myology, Department of Physiology, UMRS 974, 75013 Paris, France
- Sorbonne Université, Department of Physiology, Université Paris 06, Pitié-Salpêtrière Hospital, 75013 Paris, France
| | - Roy Combe
- Centre National de la Recherche Scientifique (CNRS), UMR7104, 67404 Illkirch, France
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1258, 67404 Illkirch, France
- Université de Strasbourg, 67404 Illkirch, France
- CELPHEDIA-PHENOMIN, Institut Clinique de la Souris (ICS), 67404 Illkirch, France
| | - Laurent Tiret
- Université Paris Est Creteil, INSERM, EnvA, EFS, AP-HP, IMRB, BNMS Team, 94700 Maisons-Alfort, France
| | - Vincent Jacquemond
- Université Lyon, Université Claude Bernard Lyon 1, CNRS UMR-5310, INSERM U-1217, Institut NeuroMyoGène, 8 Avenue Rockefeller, 69373 Lyon, France
| | - Fanny Pilot-Storck
- Université Paris Est Creteil, INSERM, EnvA, EFS, AP-HP, IMRB, BNMS Team, 94700 Maisons-Alfort, France
| | - Jocelyn Laporte
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Department of Translational Medicine, 67404 Illkirch, France
- Centre National de la Recherche Scientifique (CNRS), UMR7104, 67404 Illkirch, France
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1258, 67404 Illkirch, France
- Université de Strasbourg, 67404 Illkirch, France
| |
Collapse
|
26
|
Hsu IU, Linsley JW, Reid LE, Hume RI, Leflein A, Kuwada JY. Dstac Regulates Excitation-Contraction Coupling in Drosophila Body Wall Muscles. Front Physiol 2020; 11:573723. [PMID: 33123029 PMCID: PMC7573238 DOI: 10.3389/fphys.2020.573723] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 09/15/2020] [Indexed: 12/13/2022] Open
Abstract
Stac3 regulates excitation-contraction coupling (EC coupling) in vertebrate skeletal muscles by regulating the L-type voltage-gated calcium channel (Cav channel). Recently a stac-like gene, Dstac, was identified in Drosophila and found to be expressed by both a subset of neurons and muscles. Here, we show that Dstac and Dmca1D, the Drosophila L-type Cav channel, are necessary for normal locomotion by larvae. Immunolabeling with specific antibodies against Dstac and Dmca1D found that Dstac and Dmca1D are expressed by larval body-wall muscles. Furthermore, Ca2+ imaging of muscles of Dstac and Dmca1D deficient larvae found that Dstac and Dmca1D are required for excitation-contraction coupling. Finally, Dstac appears to be required for normal expression levels of Dmca1D in body-wall muscles. These results suggest that Dstac regulates Dmca1D during EC coupling and thus muscle contraction.
Collapse
Affiliation(s)
- I-Uen Hsu
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, United States
| | - Jeremy W Linsley
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, United States.,Cellular and Molecular Biology Program, University of Michigan, Ann Arbor, MI, United States
| | - Lilly E Reid
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, United States
| | - Richard I Hume
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, United States
| | - Ari Leflein
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, United States
| | - John Y Kuwada
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, United States.,Cellular and Molecular Biology Program, University of Michigan, Ann Arbor, MI, United States
| |
Collapse
|
27
|
Yu Y, Niccoli T, Ren Z, Woodling NS, Aleyakpo B, Szabadkai G, Partridge L. PICALM rescues glutamatergic neurotransmission, behavioural function and survival in a Drosophila model of Aβ42 toxicity. Hum Mol Genet 2020; 29:2420-2434. [PMID: 32592479 PMCID: PMC7424762 DOI: 10.1093/hmg/ddaa125] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 06/10/2020] [Accepted: 06/11/2020] [Indexed: 01/08/2023] Open
Abstract
Alzheimer's disease (AD) is the most common form of dementia and the most prevalent neurodegenerative disease. Genome-wide association studies have linked PICALM to AD risk. PICALM has been implicated in Aβ42 production and turnover, but whether it plays a direct role in modulating Aβ42 toxicity remains unclear. We found that increased expression of the Drosophila PICALM orthologue lap could rescue Aβ42 toxicity in an adult-onset model of AD, without affecting Aβ42 level. Imbalances in the glutamatergic system, leading to excessive, toxic stimulation, have been associated with AD. We found that Aβ42 caused the accumulation of presynaptic vesicular glutamate transporter (VGlut) and increased spontaneous glutamate release. Increased lap expression reversed these phenotypes back to control levels, suggesting that lap may modulate glutamatergic transmission. We also found that lap modulated the localization of amphiphysin (Amph), the homologue of another AD risk factor BIN1, and that Amph itself modulated postsynaptic glutamate receptor (GluRII) localization. We propose a model where PICALM modulates glutamatergic transmission, together with BIN1, to ameliorate synaptic dysfunction and disease progression.
Collapse
Affiliation(s)
- Yifan Yu
- Department of Genetics, Evolution and Environment, Institute of Healthy Ageing, University College London, London WC1E 6BT, UK
| | - Teresa Niccoli
- Department of Genetics, Evolution and Environment, Institute of Healthy Ageing, University College London, London WC1E 6BT, UK
- UK Dementia Research Institute at UCL, London WC1E 6BT, UK
| | - Ziyu Ren
- Department of Cell and Developmental Biology, Consortium for Mitochondrial Research, University College London, London WC1E 6BT, UK
| | - Nathaniel S Woodling
- Department of Genetics, Evolution and Environment, Institute of Healthy Ageing, University College London, London WC1E 6BT, UK
| | - Benjamin Aleyakpo
- Department of Genetics, Evolution and Environment, Institute of Healthy Ageing, University College London, London WC1E 6BT, UK
| | - Gyorgy Szabadkai
- Department of Cell and Developmental Biology, Consortium for Mitochondrial Research, University College London, London WC1E 6BT, UK
- The Francis Crick Institute, London NW1 1AT, UK
- Department of Biomedical Sciences, University of Padua, Padua 35131, Italy
| | - Linda Partridge
- Department of Genetics, Evolution and Environment, Institute of Healthy Ageing, University College London, London WC1E 6BT, UK
- Max Planck Institute for Biology of Ageing, Cologne 50931, Germany
| |
Collapse
|
28
|
Auerswald A, Gruber T, Balbach J, Meister A. Lipid-Dependent Interaction of Human N-BAR Domain Proteins with Sarcolemma Mono- and Bilayers. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2020; 36:8695-8704. [PMID: 32649209 DOI: 10.1021/acs.langmuir.0c00649] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
The N-BAR domain of the human Bin1 protein is indispensable for T-tubule biogenesis in skeletal muscles. It binds to lipid mono- and bilayers that mimic the sarcolemma membrane composition, and it transforms vesicles into uniform tubules by generating a decorating protein scaffold. We found that Δ(1-33)BAR, lacking the N-terminal amphipathic helix (H0), and H0 alone bind to sarcolemma monolayers, although both proteins are not able to tubulate sarcolemma vesicles. By variation of the lipid composition, we elucidated the role of PI(4,5)P2, cholesterol, and an asymmetric sarcolemma composition for Bin1-N-BAR binding and sarcolemma tubulation. Our results indicate that Bin1-N-BAR binding is low in the absence of PI(4,5)P2 and it is affected by additional changes in the negative headgroup charge and lipid acyl chain composition. However, it is not dependent on the cholesterol content. The results from Langmuir monolayer experiments are complementary to lipid bilayer studies using electron microscopy that provides information on membrane curvature generation.
Collapse
Affiliation(s)
- Andrea Auerswald
- Institute of Biochemistry and Biotechnology, Martin-Luther-University Halle-Wittenberg, Kurt-Mothes-Straße 3a, 06120 Halle (Saale), Germany
- HALOmem, Martin-Luther-University Halle-Wittenberg, Kurt-Mothes-Straße 3a, 06120 Halle (Saale), Germany
| | - Tobias Gruber
- Institute of Physics, Martin-Luther-University Halle-Wittenberg, Betty-Heimann-Straße 7, 06120 Halle (Saale), Germany
| | - Jochen Balbach
- HALOmem, Martin-Luther-University Halle-Wittenberg, Kurt-Mothes-Straße 3a, 06120 Halle (Saale), Germany
- Institute of Physics, Martin-Luther-University Halle-Wittenberg, Betty-Heimann-Straße 7, 06120 Halle (Saale), Germany
| | - Annette Meister
- Institute of Biochemistry and Biotechnology, Martin-Luther-University Halle-Wittenberg, Kurt-Mothes-Straße 3a, 06120 Halle (Saale), Germany
- HALOmem, Martin-Luther-University Halle-Wittenberg, Kurt-Mothes-Straße 3a, 06120 Halle (Saale), Germany
| |
Collapse
|
29
|
Lu F, Pu WT. The architecture and function of cardiac dyads. Biophys Rev 2020; 12:1007-1017. [PMID: 32661902 PMCID: PMC7429583 DOI: 10.1007/s12551-020-00729-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Accepted: 07/03/2020] [Indexed: 12/28/2022] Open
Abstract
Cardiac excitation-contraction (EC) coupling, which links plasma membrane depolarization to activation of cardiomyocyte contraction, occurs at dyads, the nanoscopic microdomains formed by apposition of transverse (T)-tubules and junctional sarcoplasmic reticulum (jSR). In a dyadic junction, EC coupling occurs through Ca2+-induced Ca2+ release. Membrane depolarization opens voltage-gated L-type Ca2+ channels (LTCCs) in the T-tubule. The resulting influx of extracellular Ca2+ into the dyadic cleft opens Ca2+ release channels known as ryanodine receptors (RYRs) in the jSR, leading to the rapid increase in cytosolic Ca2+ that triggers sarcomere contraction. The efficacy of LTCC-RYR communication greatly affects a myriad of downstream intracellular signaling events, and it is controlled by many factors, including T-tubule and jSR structure, spatial distribution of ion channels, and regulatory proteins that closely regulate the activities of channels within dyads. Alterations in dyad architecture and/or channel activity are seen in many types of heart disease. This review will focus on the current knowledge regarding cardiac dyad structure and function, their alterations in heart failure, and new approaches to study the composition and function of dyads.
Collapse
Affiliation(s)
- Fujian Lu
- Department of Cardiology, Boston Children's Hospital, 300 Longwood Ave, Boston, MA, 02115, USA
| | - William T Pu
- Department of Cardiology, Boston Children's Hospital, 300 Longwood Ave, Boston, MA, 02115, USA.
- Harvard Stem Cell Institute, 7 Divinity Avenue, Cambridge, MA, 02138, USA.
| |
Collapse
|
30
|
Poovathumkadavil P, Jagla K. Genetic Control of Muscle Diversification and Homeostasis: Insights from Drosophila. Cells 2020; 9:cells9061543. [PMID: 32630420 PMCID: PMC7349286 DOI: 10.3390/cells9061543] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Revised: 06/19/2020] [Accepted: 06/23/2020] [Indexed: 12/13/2022] Open
Abstract
In the fruit fly, Drosophila melanogaster, the larval somatic muscles or the adult thoracic flight and leg muscles are the major voluntary locomotory organs. They share several developmental and structural similarities with vertebrate skeletal muscles. To ensure appropriate activity levels for their functions such as hatching in the embryo, crawling in the larva, and jumping and flying in adult flies all muscle components need to be maintained in a functionally stable or homeostatic state despite constant strain. This requires that the muscles develop in a coordinated manner with appropriate connections to other cell types they communicate with. Various signaling pathways as well as extrinsic and intrinsic factors are known to play a role during Drosophila muscle development, diversification, and homeostasis. In this review, we discuss genetic control mechanisms of muscle contraction, development, and homeostasis with particular emphasis on the contractile unit of the muscle, the sarcomere.
Collapse
|
31
|
Sun T, Shen J, Achilli A, Chen N, Chen Q, Dang R, Zheng Z, Zhang H, Zhang X, Wang S, Zhang T, Lu H, Ma Y, Jia Y, Capodiferro MR, Huang Y, Lan X, Chen H, Jiang Y, Lei C. Genomic analyses reveal distinct genetic architectures and selective pressures in buffaloes. Gigascience 2020; 9:giz166. [PMID: 32083286 PMCID: PMC7033652 DOI: 10.1093/gigascience/giz166] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Revised: 11/26/2019] [Accepted: 12/27/2019] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND The domestic buffalo (Bubalus bubalis) is an essential farm animal in tropical and subtropical regions, whose genomic diversity is yet to be fully discovered. RESULTS In this study, we describe the demographic events and selective pressures of buffalo by analyzing 121 whole genomes (98 newly reported) from 25 swamp and river buffalo breeds. Both uniparental and biparental markers were investigated to provide the final scenario. The ancestors of swamp and river buffalo diverged ∼0.23 million years ago and then experienced independent demographic histories. They were domesticated in different regions, the swamp buffalo at the border between southwest China and southeast Asia, while the river buffalo in south Asia. The domestic stocks migrated to other regions and further differentiated, as testified by (at least) 2 ancestral components identified in each subspecies. Different signals of selective pressures were also detected in these 2 types of buffalo. The swamp buffalo, historically used as a draft animal, shows selection signatures in genes associated with the nervous system, while in river dairy breeds, genes under selection are related to heat stress and immunity. CONCLUSIONS Our findings substantially expand the catalogue of genetic variants in buffalo and reveal new insights into the evolutionary history and distinct selective pressures in river and swamp buffalo.
Collapse
Affiliation(s)
- Ting Sun
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Jiafei Shen
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Alessandro Achilli
- Dipartimento di Biologia e Biotecnologie “L. Spallanzani,” Università di Pavia, Pavia 27100, Italy
| | - Ningbo Chen
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Qiuming Chen
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Ruihua Dang
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Zhuqing Zheng
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Hucai Zhang
- Key Laboratory of Plateau Lake Ecology and Environment Change, Yunnan University, Kunming 650504, China
| | - Xiaoming Zhang
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China
| | - Shaoqiang Wang
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Tao Zhang
- School of Bioscience and Engineering, Shaanxi University of Technology, Hanzhong, Shaanxi 723000, China
| | - Hongzhao Lu
- School of Bioscience and Engineering, Shaanxi University of Technology, Hanzhong, Shaanxi 723000, China
| | - Yun Ma
- Agricultural College, Ningxia University, Yinchuan 750021, China
| | - Yutang Jia
- Institute of Animal Science and Veterinary Medicine, Anhui Academy of Agriculture Science, Hefei 230001, China
| | | | - Yongzhen Huang
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Xianyong Lan
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Hong Chen
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Yu Jiang
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Chuzhao Lei
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| |
Collapse
|
32
|
Gowrisankaran S, Wang Z, Morgan DG, Milosevic I, Mim C. Cells Control BIN1-Mediated Membrane Tubulation by Altering the Membrane Charge. J Mol Biol 2019; 432:1235-1250. [PMID: 31857086 DOI: 10.1016/j.jmb.2019.12.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 11/18/2019] [Accepted: 12/07/2019] [Indexed: 11/15/2022]
Abstract
The Bridging integrator 1 (BIN1)/Amphiphysin/Rvs (BAR) protein family is an essential part of the cell's machinery to bend membranes. BIN1 is a muscle-enriched BAR protein with an established role in muscle development and skeletal myopathies. Here, we demonstrate that BIN1, on its own, is able to form complex interconnected tubular systems in vitro, reminiscent of t-tubule system in muscle cells. We further describe how BIN1's electrostatic interactions regulate membrane bending: the ratio of negatively charged lipids in the bilayer altered membrane bending and binding properties of BIN1 and so did the manipulation of BIN1's surface charge. We show that the electrostatically mediated BIN1 membrane binding depended on the membrane curvature-it was less affected in liposomes with high curvature. Curiously, BIN1 membrane binding and bending was diminished in cells where the membrane's charge was experimentally reduced. Membrane bending was also reduced in BIN1 mutants where negative or positive charges in the BAR domain have been eliminated. This phenotype, characteristic of BIN1 mutants linked to myopathies, was rescued when the membrane charge was made more negative. The latter findings also show that cells can control tubulation at their membranes by simply altering the membrane charge and through it, the recruitment of BAR proteins and their interaction partners (e.g. dynamin).
Collapse
Affiliation(s)
- Sindhuja Gowrisankaran
- European Neuroscience Institute (ENI), A Joint Initiative of the University Medical Center Göttingen and the Max Planck Society, Synaptic Vesicle Dynamics Group, Göttingen, Germany
| | - Zuoneng Wang
- Royal Technical Institute (KTH), Dept. for Biomedical Engineering and Health Systems, Stockholm, Sweden; Karolinska Institute, Department of Nutrition and Biosciences, Huddinge, Sweden
| | - David G Morgan
- Indiana University, Electron Microscopy Center, Bloomington, IN USA
| | - Ira Milosevic
- European Neuroscience Institute (ENI), A Joint Initiative of the University Medical Center Göttingen and the Max Planck Society, Synaptic Vesicle Dynamics Group, Göttingen, Germany
| | - Carsten Mim
- Royal Technical Institute (KTH), Dept. for Biomedical Engineering and Health Systems, Stockholm, Sweden; Karolinska Institute, Department of Nutrition and Biosciences, Huddinge, Sweden.
| |
Collapse
|
33
|
Hanawa-Suetsugu K, Itoh Y, Ab Fatah M, Nishimura T, Takemura K, Takeshita K, Kubota S, Miyazaki N, Wan Mohamad Noor WNI, Inaba T, Nguyen NTH, Hamada-Nakahara S, Oono-Yakura K, Tachikawa M, Iwasaki K, Kohda D, Yamamoto M, Kitao A, Shimada A, Suetsugu S. Phagocytosis is mediated by two-dimensional assemblies of the F-BAR protein GAS7. Nat Commun 2019; 10:4763. [PMID: 31628328 PMCID: PMC6802115 DOI: 10.1038/s41467-019-12738-w] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Accepted: 09/26/2019] [Indexed: 01/07/2023] Open
Abstract
Phagocytosis is a cellular process for internalization of micron-sized large particles including pathogens. The Bin-Amphiphysin-Rvs167 (BAR) domain proteins, including the FCH-BAR (F-BAR) domain proteins, impose specific morphologies on lipid membranes. Most BAR domain proteins are thought to form membrane invaginations or protrusions by assembling into helical submicron-diameter filaments, such as on clathrin-coated pits, caveolae, and filopodia. However, the mechanism by which BAR domain proteins assemble into micron-scale phagocytic cups was unclear. Here, we show that the two-dimensional sheet-like assembly of Growth Arrest-Specific 7 (GAS7) plays a critical role in phagocytic cup formation in macrophages. GAS7 has the F-BAR domain that possesses unique hydrophilic loops for two-dimensional sheet formation on flat membranes. Super-resolution microscopy reveals the similar assemblies of GAS7 on phagocytic cups and liposomes. The mutations of the loops abolishes both the membrane localization of GAS7 and phagocytosis. Thus, the sheet-like assembly of GAS7 plays a significant role in phagocytosis.
Collapse
Affiliation(s)
- Kyoko Hanawa-Suetsugu
- 0000 0000 9227 2257grid.260493.aNara Institute of Science and Technology, Ikoma, 630-0192 Japan
| | - Yuzuru Itoh
- 0000 0001 2151 536Xgrid.26999.3dUniversity of Tokyo, Tokyo, 113-0032 Japan ,0000 0004 1936 9377grid.10548.38Present Address: Science for Life Laboratory, Department of Biochemistry and Biophysics, Stockholm University, Stockholm, Sweden
| | - Maisarah Ab Fatah
- 0000 0000 9227 2257grid.260493.aNara Institute of Science and Technology, Ikoma, 630-0192 Japan
| | - Tamako Nishimura
- 0000 0000 9227 2257grid.260493.aNara Institute of Science and Technology, Ikoma, 630-0192 Japan
| | - Kazuhiro Takemura
- 0000 0001 2179 2105grid.32197.3eSchool of Life Science and Technology, Tokyo Institute of Technology, Tokyo, 152-8550 Japan
| | | | - Satoru Kubota
- 0000 0000 9227 2257grid.260493.aNara Institute of Science and Technology, Ikoma, 630-0192 Japan
| | - Naoyuki Miyazaki
- 0000 0004 0373 3971grid.136593.bInstitute for Protein Research, Osaka University, Suita, Osaka 565-0871 Japan
| | | | - Takehiko Inaba
- 0000 0000 9227 2257grid.260493.aNara Institute of Science and Technology, Ikoma, 630-0192 Japan
| | - Nhung Thi Hong Nguyen
- 0000 0000 9227 2257grid.260493.aNara Institute of Science and Technology, Ikoma, 630-0192 Japan
| | | | - Kayoko Oono-Yakura
- 0000 0000 9227 2257grid.260493.aNara Institute of Science and Technology, Ikoma, 630-0192 Japan
| | - Masashi Tachikawa
- 0000000094465255grid.7597.cTheoretical Biology Laboratory, RIKEN, Wako, 351-0198 Japan
| | - Kenji Iwasaki
- 0000 0004 0373 3971grid.136593.bInstitute for Protein Research, Osaka University, Suita, Osaka 565-0871 Japan ,0000 0001 2369 4728grid.20515.33Present Address: Tsukuba Advanced Research Alliance, Life Science Center for Survival Dynamics, University of Tsukuba, Tsukuba, Japan
| | - Daisuke Kohda
- RIKEN SPring-8 Center, Sayo, Hyogo 679-5148 Japan ,0000 0001 2242 4849grid.177174.3Division of Structural Biology, Medical Institute of Bioregulation, Kyushu University, Fukuoka, 812-8582 Japan
| | | | - Akio Kitao
- 0000 0001 2179 2105grid.32197.3eSchool of Life Science and Technology, Tokyo Institute of Technology, Tokyo, 152-8550 Japan
| | - Atsushi Shimada
- RIKEN SPring-8 Center, Sayo, Hyogo 679-5148 Japan ,0000 0001 2242 4849grid.177174.3Division of Structural Biology, Medical Institute of Bioregulation, Kyushu University, Fukuoka, 812-8582 Japan
| | - Shiro Suetsugu
- 0000 0000 9227 2257grid.260493.aNara Institute of Science and Technology, Ikoma, 630-0192 Japan
| |
Collapse
|
34
|
Sauerwald J, Backer W, Matzat T, Schnorrer F, Luschnig S. Matrix metalloproteinase 1 modulates invasive behavior of tracheal branches during entry into Drosophila flight muscles. eLife 2019; 8:48857. [PMID: 31577228 PMCID: PMC6795481 DOI: 10.7554/elife.48857] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Accepted: 10/01/2019] [Indexed: 12/30/2022] Open
Abstract
Tubular networks like the vasculature extend branches throughout animal bodies, but how developing vessels interact with and invade tissues is not well understood. We investigated the underlying mechanisms using the developing tracheal tube network of Drosophila indirect flight muscles (IFMs) as a model. Live imaging revealed that tracheal sprouts invade IFMs directionally with growth-cone-like structures at branch tips. Ramification inside IFMs proceeds until tracheal branches fill the myotube. However, individual tracheal cells occupy largely separate territories, possibly mediated by cell-cell repulsion. Matrix metalloproteinase 1 (MMP1) is required in tracheal cells for normal invasion speed and for the dynamic organization of growth-cone-like branch tips. MMP1 remodels the CollagenIV-containing matrix around branch tips, which show differential matrix composition with low CollagenIV levels, while Laminin is present along tracheal branches. Thus, tracheal-derived MMP1 sustains branch invasion by modulating the dynamic behavior of sprouting branches as well as properties of the surrounding matrix.
Collapse
Affiliation(s)
- Julia Sauerwald
- Institute for Zoophysiology, University of Münster, Münster, Germany.,Cluster of Excellence EXC 1003, Cells in Motion (CiM), Münster, Germany
| | - Wilko Backer
- Institute for Zoophysiology, University of Münster, Münster, Germany.,Cluster of Excellence EXC 1003, Cells in Motion (CiM), Münster, Germany
| | - Till Matzat
- Institute for Zoophysiology, University of Münster, Münster, Germany.,Cluster of Excellence EXC 1003, Cells in Motion (CiM), Münster, Germany
| | | | - Stefan Luschnig
- Institute for Zoophysiology, University of Münster, Münster, Germany.,Cluster of Excellence EXC 1003, Cells in Motion (CiM), Münster, Germany
| |
Collapse
|
35
|
Muller MP, Jiang T, Sun C, Lihan M, Pant S, Mahinthichaichan P, Trifan A, Tajkhorshid E. Characterization of Lipid-Protein Interactions and Lipid-Mediated Modulation of Membrane Protein Function through Molecular Simulation. Chem Rev 2019; 119:6086-6161. [PMID: 30978005 PMCID: PMC6506392 DOI: 10.1021/acs.chemrev.8b00608] [Citation(s) in RCA: 173] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
The cellular membrane constitutes one of the most fundamental compartments of a living cell, where key processes such as selective transport of material and exchange of information between the cell and its environment are mediated by proteins that are closely associated with the membrane. The heterogeneity of lipid composition of biological membranes and the effect of lipid molecules on the structure, dynamics, and function of membrane proteins are now widely recognized. Characterization of these functionally important lipid-protein interactions with experimental techniques is however still prohibitively challenging. Molecular dynamics (MD) simulations offer a powerful complementary approach with sufficient temporal and spatial resolutions to gain atomic-level structural information and energetics on lipid-protein interactions. In this review, we aim to provide a broad survey of MD simulations focusing on exploring lipid-protein interactions and characterizing lipid-modulated protein structure and dynamics that have been successful in providing novel insight into the mechanism of membrane protein function.
Collapse
Affiliation(s)
- Melanie P. Muller
- NIH Center for Macromolecular Modeling and Bioinformatics, Beckman Institute for Advanced Science and Technology
- Department of Biochemistry
- Center for Biophysics and Quantitative Biology
- College of Medicine
- University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Tao Jiang
- NIH Center for Macromolecular Modeling and Bioinformatics, Beckman Institute for Advanced Science and Technology
- Department of Biochemistry
- Center for Biophysics and Quantitative Biology
- University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Chang Sun
- NIH Center for Macromolecular Modeling and Bioinformatics, Beckman Institute for Advanced Science and Technology
- Department of Biochemistry
- University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Muyun Lihan
- NIH Center for Macromolecular Modeling and Bioinformatics, Beckman Institute for Advanced Science and Technology
- Department of Biochemistry
- Center for Biophysics and Quantitative Biology
- University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Shashank Pant
- NIH Center for Macromolecular Modeling and Bioinformatics, Beckman Institute for Advanced Science and Technology
- Department of Biochemistry
- Center for Biophysics and Quantitative Biology
- University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Paween Mahinthichaichan
- NIH Center for Macromolecular Modeling and Bioinformatics, Beckman Institute for Advanced Science and Technology
- Department of Biochemistry
- University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Anda Trifan
- NIH Center for Macromolecular Modeling and Bioinformatics, Beckman Institute for Advanced Science and Technology
- Department of Biochemistry
- Center for Biophysics and Quantitative Biology
- University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Emad Tajkhorshid
- NIH Center for Macromolecular Modeling and Bioinformatics, Beckman Institute for Advanced Science and Technology
- Department of Biochemistry
- Center for Biophysics and Quantitative Biology
- College of Medicine
- University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| |
Collapse
|
36
|
Lawless M, Caldwell JL, Radcliffe EJ, Smith CER, Madders GWP, Hutchings DC, Woods LS, Church SJ, Unwin RD, Kirkwood GJ, Becker LK, Pearman CM, Taylor RF, Eisner DA, Dibb KM, Trafford AW. Phosphodiesterase 5 inhibition improves contractile function and restores transverse tubule loss and catecholamine responsiveness in heart failure. Sci Rep 2019; 9:6801. [PMID: 31043634 PMCID: PMC6494852 DOI: 10.1038/s41598-019-42592-1] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Accepted: 03/26/2019] [Indexed: 12/13/2022] Open
Abstract
Heart failure (HF) is characterized by poor survival, a loss of catecholamine reserve and cellular structural remodeling in the form of disorganization and loss of the transverse tubule network. Indeed, survival rates for HF are worse than many common cancers and have not improved over time. Tadalafil is a clinically relevant drug that blocks phosphodiesterase 5 with high specificity and is used to treat erectile dysfunction. Using a sheep model of advanced HF, we show that tadalafil treatment improves contractile function, reverses transverse tubule loss, restores calcium transient amplitude and the heart's response to catecholamines. Accompanying these effects, tadalafil treatment normalized BNP mRNA and prevented development of subjective signs of HF. These effects were independent of changes in myocardial cGMP content and were associated with upregulation of both monomeric and dimerized forms of protein kinase G and of the cGMP hydrolyzing phosphodiesterases 2 and 3. We propose that the molecular switch for the loss of transverse tubules in HF and their restoration following tadalafil treatment involves the BAR domain protein Amphiphysin II (BIN1) and the restoration of catecholamine sensitivity is through reductions in G-protein receptor kinase 2, protein phosphatase 1 and protein phosphatase 2 A abundance following phosphodiesterase 5 inhibition.
Collapse
Affiliation(s)
- Michael Lawless
- Division of Cardiovascular Sciences, Unit of Cardiac Physiology, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, 3.24 Core Technology Facility, 46 Grafton Street, Manchester, M13 9NT, United Kingdom
| | - Jessica L Caldwell
- Division of Cardiovascular Sciences, Unit of Cardiac Physiology, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, 3.24 Core Technology Facility, 46 Grafton Street, Manchester, M13 9NT, United Kingdom
| | - Emma J Radcliffe
- Division of Cardiovascular Sciences, Unit of Cardiac Physiology, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, 3.24 Core Technology Facility, 46 Grafton Street, Manchester, M13 9NT, United Kingdom
| | - Charlotte E R Smith
- Division of Cardiovascular Sciences, Unit of Cardiac Physiology, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, 3.24 Core Technology Facility, 46 Grafton Street, Manchester, M13 9NT, United Kingdom
| | - George W P Madders
- Division of Cardiovascular Sciences, Unit of Cardiac Physiology, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, 3.24 Core Technology Facility, 46 Grafton Street, Manchester, M13 9NT, United Kingdom
| | - David C Hutchings
- Division of Cardiovascular Sciences, Unit of Cardiac Physiology, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, 3.24 Core Technology Facility, 46 Grafton Street, Manchester, M13 9NT, United Kingdom
| | - Lori S Woods
- Division of Cardiovascular Sciences, Unit of Cardiac Physiology, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, 3.24 Core Technology Facility, 46 Grafton Street, Manchester, M13 9NT, United Kingdom
| | - Stephanie J Church
- Division of Cardiovascular Sciences, Centre for Advanced Discovery and Experimental Therapeutics, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, 3.24 Core Technology Facility, 46 Grafton Street, Manchester, M13 9NT, United Kingdom
| | - Richard D Unwin
- Division of Cardiovascular Sciences, Centre for Advanced Discovery and Experimental Therapeutics, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, 3.24 Core Technology Facility, 46 Grafton Street, Manchester, M13 9NT, United Kingdom
| | - Graeme J Kirkwood
- Division of Cardiovascular Sciences, Unit of Cardiac Physiology, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, 3.24 Core Technology Facility, 46 Grafton Street, Manchester, M13 9NT, United Kingdom
| | - Lorenz K Becker
- Division of Cardiovascular Sciences, Unit of Cardiac Physiology, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, 3.24 Core Technology Facility, 46 Grafton Street, Manchester, M13 9NT, United Kingdom
| | - Charles M Pearman
- Division of Cardiovascular Sciences, Unit of Cardiac Physiology, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, 3.24 Core Technology Facility, 46 Grafton Street, Manchester, M13 9NT, United Kingdom
| | - Rebecca F Taylor
- Division of Cardiovascular Sciences, Unit of Cardiac Physiology, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, 3.24 Core Technology Facility, 46 Grafton Street, Manchester, M13 9NT, United Kingdom
| | - David A Eisner
- Division of Cardiovascular Sciences, Unit of Cardiac Physiology, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, 3.24 Core Technology Facility, 46 Grafton Street, Manchester, M13 9NT, United Kingdom
| | - Katharine M Dibb
- Division of Cardiovascular Sciences, Unit of Cardiac Physiology, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, 3.24 Core Technology Facility, 46 Grafton Street, Manchester, M13 9NT, United Kingdom
| | - Andrew W Trafford
- Division of Cardiovascular Sciences, Unit of Cardiac Physiology, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, 3.24 Core Technology Facility, 46 Grafton Street, Manchester, M13 9NT, United Kingdom.
| |
Collapse
|
37
|
Lionello VM, Nicot AS, Sartori M, Kretz C, Kessler P, Buono S, Djerroud S, Messaddeq N, Koebel P, Prokic I, Hérault Y, Romero NB, Laporte J, Cowling BS. Amphiphysin 2 modulation rescues myotubular myopathy and prevents focal adhesion defects in mice. Sci Transl Med 2019; 11:11/484/eaav1866. [DOI: 10.1126/scitranslmed.aav1866] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Revised: 11/14/2018] [Accepted: 02/28/2019] [Indexed: 12/13/2022]
Abstract
Centronuclear myopathies (CNMs) are severe diseases characterized by muscle weakness and myofiber atrophy. Currently, there are no approved treatments for these disorders. Mutations in the phosphoinositide 3-phosphatase myotubularin (MTM1) are responsible for X-linked CNM (XLCNM), also called myotubular myopathy, whereas mutations in the membrane remodeling Bin/amphiphysin/Rvs protein amphiphysin 2 [bridging integrator 1 (BIN1)] are responsible for an autosomal form of the disease. Here, we investigated the functional relationship between MTM1 and BIN1 in healthy skeletal muscle and in the physiopathology of CNM. Genetic overexpression of human BIN1 efficiently rescued the muscle weakness and life span in a mouse model of XLCNM. Exogenous human BIN1 expression with adeno-associated virus after birth also prevented the progression of the disease, suggesting that human BIN1 overexpression can compensate for the lack of MTM1 expression in this mouse model. Our results showed that MTM1 controls cell adhesion and integrin localization in mammalian muscle. Alterations in this pathway in Mtm1−/y mice were associated with defects in myofiber shape and size. BIN1 expression rescued integrin and laminin alterations and restored myofiber integrity, supporting the idea that MTM1 and BIN1 are functionally linked and necessary for focal adhesions in skeletal muscle. The results suggest that BIN1 modulation might be an effective strategy for treating XLCNM.
Collapse
|
38
|
Kaya-Çopur A, Schnorrer F. RNA Interference Screening for Genes Regulating Drosophila Muscle Morphogenesis. Methods Mol Biol 2019; 1889:331-348. [PMID: 30367424 DOI: 10.1007/978-1-4939-8897-6_20] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
RNA interference (RNAi) is the method of choice to systematically test for gene function in an intact organism. The model organism Drosophila has the advantage that RNAi is cell autonomous, meaning it does not spread from one cell to the next. Hence, RNAi can be performed in a tissue-specific manner by expressing short or long inverted repeat constructs (hairpins) designed to target mRNAs from one specific target gene. This achieves tissue-specific knock-down of a target gene of choice. Here, we detail the methodology to test gene function in Drosophila muscle tissue by expressing hairpins in a muscle-specific manner using the GAL4-UAS system. We further discuss the systematic RNAi resource collections available which also permit large scale screens in a muscle-specific manner. The full power of such screens is revealed by combination of high-throughput assays followed by detailed morphological assays. Together, this chapter should be a practical guide to enable the reader to either test a few candidate genes, or large gene sets for particular functions in Drosophila muscle tissue and provide first insights into the biological process the gene might be important for in muscle.
Collapse
|
39
|
Reuveny A, Shnayder M, Lorber D, Wang S, Volk T. Ma2/d promotes myonuclear positioning and association with the sarcoplasmic reticulum. Development 2018; 145:dev.159558. [PMID: 30093550 DOI: 10.1242/dev.159558] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Accepted: 07/24/2018] [Indexed: 12/24/2022]
Abstract
The cytoplasm of striated myofibers contains a large number of membrane organelles, including sarcoplasmic reticulum (SR), T-tubules and the nuclear membrane. These organelles maintain a characteristic juxtaposition that appears to be essential for efficient inter-membranous exchange of RNA, proteins and ions. We found that the membrane-associated Muscle-specific α2/δ (Ma2/d) subunit of the Ca2+ channel complex localizes to the SR and T-tubules, and accumulates at the myonuclear surfaces. Furthermore, Ma2/d mutant larval muscles exhibit nuclear positioning defects, disruption of the nuclear-SR juxtapositioning, as well as impaired larval locomotion. Ma2/d localization at the nuclear membrane depends on the proper function of the nesprin ortholog Msp300 and the BAR domain protein Amphiphysin (Amph). Importantly, live imaging of muscle contraction in intact Drosophila larvae indicated altered distribution of Sarco/Endoplamic Reticulum Ca2+-ATPase (SERCA) around the myonuclei of Ma2/d mutant larvae. Co-immunoprecipitation analysis supports association between Ma2/d and Amph, and indirectly with Msp300. We therefore suggest that Ma2/d, in association with Msp300 and Amph, mediates interactions between the SR and the nuclear membrane.
Collapse
Affiliation(s)
- Adriana Reuveny
- Department of Molecular Genetics, Weizmann Institute, Rehovot 76100, Israel
| | - Marina Shnayder
- Department of Molecular Genetics, Weizmann Institute, Rehovot 76100, Israel
| | - Dana Lorber
- Department of Molecular Genetics, Weizmann Institute, Rehovot 76100, Israel
| | - Shuoshuo Wang
- Department of Molecular Genetics, Weizmann Institute, Rehovot 76100, Israel
| | - Talila Volk
- Department of Molecular Genetics, Weizmann Institute, Rehovot 76100, Israel
| |
Collapse
|
40
|
Sheng C, Javed U, Gibbs M, Long C, Yin J, Qin B, Yuan Q. Experience-dependent structural plasticity targets dynamic filopodia in regulating dendrite maturation and synaptogenesis. Nat Commun 2018; 9:3362. [PMID: 30135566 PMCID: PMC6105721 DOI: 10.1038/s41467-018-05871-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Accepted: 07/27/2018] [Indexed: 11/21/2022] Open
Abstract
Highly motile dendritic protrusions are hallmarks of developing neurons. These exploratory filopodia sample the environment and initiate contacts with potential synaptic partners. To understand the role for dynamic filopodia in dendrite morphogenesis and experience-dependent structural plasticity, we analyzed dendrite dynamics, synapse formation, and dendrite volume expansion in developing ventral lateral neurons (LNvs) of the Drosophila larval visual circuit. Our findings reveal the temporal coordination between heightened dendrite dynamics with synaptogenesis in LNvs and illustrate the strong influence imposed by sensory experience on the prevalence of dendritic filopodia, which regulate the formation of synapses and the expansion of dendritic arbors. Using genetic analyses, we further identified Amphiphysin (Amph), a BAR (Bin/Amphiphysin/Rvs) domain-containing protein as a required component for tuning the dynamic state of LNv dendrites and promoting dendrite maturation. Taken together, our study establishes dynamic filopodia as the key cellular target for experience-dependent regulation of dendrite development. During development, dendrites display structural plasticity, as reflected in the appearance of long, thin and highly motile dendritic filopodia. Here, the authors examine dendritic dynamics of ventral lateral neurons in the developing Drosophila larva, and identify Amphiphysin as an important regulator of this process.
Collapse
Affiliation(s)
- Chengyu Sheng
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Uzma Javed
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Mary Gibbs
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Caixia Long
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Jun Yin
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Bo Qin
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Quan Yuan
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, 20892, USA.
| |
Collapse
|
41
|
Gan Q, Watanabe S. Synaptic Vesicle Endocytosis in Different Model Systems. Front Cell Neurosci 2018; 12:171. [PMID: 30002619 PMCID: PMC6031744 DOI: 10.3389/fncel.2018.00171] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2018] [Accepted: 06/01/2018] [Indexed: 11/13/2022] Open
Abstract
Neurotransmission in complex animals depends on a choir of functionally distinct synapses releasing neurotransmitters in a highly coordinated manner. During synaptic signaling, vesicles fuse with the plasma membrane to release their contents. The rate of vesicle fusion is high and can exceed the rate at which synaptic vesicles can be re-supplied by distant sources. Thus, local compensatory endocytosis is needed to replenish the synaptic vesicle pools. Over the last four decades, various experimental methods and model systems have been used to study the cellular and molecular mechanisms underlying synaptic vesicle cycle. Clathrin-mediated endocytosis is thought to be the predominant mechanism for synaptic vesicle recycling. However, recent studies suggest significant contribution from other modes of endocytosis, including fast compensatory endocytosis, activity-dependent bulk endocytosis, ultrafast endocytosis, as well as kiss-and-run. Currently, it is not clear whether a universal model of vesicle recycling exist for all types of synapses. It is possible that each synapse type employs a particular mode of endocytosis. Alternatively, multiple modes of endocytosis operate at the same synapse, and the synapse toggles between different modes depending on its activity level. Here we compile review and research articles based on well-characterized model systems: frog neuromuscular junctions, C. elegans neuromuscular junctions, Drosophila neuromuscular junctions, lamprey reticulospinal giant axons, goldfish retinal ribbon synapses, the calyx of Held, and rodent hippocampal synapses. We will compare these systems in terms of their known modes and kinetics of synaptic vesicle endocytosis, as well as the underlying molecular machineries. We will also provide the future development of this field.
Collapse
Affiliation(s)
- Quan Gan
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Shigeki Watanabe
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD, United States.,Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
42
|
Calpena E, López Del Amo V, Chakraborty M, Llamusí B, Artero R, Espinós C, Galindo MI. The Drosophila junctophilin gene is functionally equivalent to its four mammalian counterparts and is a modifier of a Huntingtin poly-Q expansion and the Notch pathway. Dis Model Mech 2018; 11:dmm.029082. [PMID: 29208631 PMCID: PMC5818072 DOI: 10.1242/dmm.029082] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Accepted: 11/08/2017] [Indexed: 12/13/2022] Open
Abstract
Members of the Junctophilin (JPH) protein family have emerged as key actors in all excitable cells, with crucial implications for human pathophysiology. In mammals, this family consists of four members (JPH1-JPH4) that are differentially expressed throughout excitable cells. The analysis of knockout mice lacking JPH subtypes has demonstrated their essential contribution to physiological functions in skeletal and cardiac muscles and in neurons. Moreover, mutations in the human JPH2 gene are associated with hypertrophic and dilated cardiomyopathies; mutations in JPH3 are responsible for the neurodegenerative Huntington's disease-like-2 (HDL2), whereas JPH1 acts as a genetic modifier in Charcot–Marie–Tooth 2K peripheral neuropathy. Drosophila melanogaster has a single junctophilin (jp) gene, as is the case in all invertebrates, which might retain equivalent functions of the four homologous JPH genes present in mammalian genomes. Therefore, owing to the lack of putatively redundant genes, a jpDrosophila model could provide an excellent platform to model the Junctophilin-related diseases, to discover the ancestral functions of the JPH proteins and to reveal new pathways. By up- and downregulation of Jp in a tissue-specific manner in Drosophila, we show that altering its levels of expression produces a phenotypic spectrum characterized by muscular deficits, dilated cardiomyopathy and neuronal alterations. Importantly, our study has demonstrated that Jp modifies the neuronal degeneration in a Drosophila model of Huntington's disease, and it has allowed us to uncover an unsuspected functional relationship with the Notch pathway. Therefore, this Drosophila model has revealed new aspects of Junctophilin function that can be relevant for the disease mechanisms of their human counterparts. Summary: This work reveals that the Drosophila Junctophilin protein has similar functions to its mammalian homologues and uncovers new interactions of potential biomedical interest with Huntingtin and Notch signalling.
Collapse
Affiliation(s)
- Eduardo Calpena
- Program in Molecular Mechanisms of Disease, Centro de Investigación Príncipe Felipe (CIPF), c/ Eduardo Primo Yúfera no. 3, 46012 Valencia, Spain
| | - Víctor López Del Amo
- Program in Molecular Mechanisms of Disease, Centro de Investigación Príncipe Felipe (CIPF), c/ Eduardo Primo Yúfera no. 3, 46012 Valencia, Spain
| | - Mouli Chakraborty
- Translational Genomics Group, Incliva Health Research Institute, Avda. Menendez Pelayo 4 acc 46010, Valencia, Spain.,Department of Genetics and Estructura de Recerca Interdisciplinar en Biotecnologia i Biomedicina (ERI BIOTECMED), Universitat de València, c/ Dr Moliner 50, 46100 Burjasot, Spain
| | - Beatriz Llamusí
- Translational Genomics Group, Incliva Health Research Institute, Avda. Menendez Pelayo 4 acc 46010, Valencia, Spain.,Department of Genetics and Estructura de Recerca Interdisciplinar en Biotecnologia i Biomedicina (ERI BIOTECMED), Universitat de València, c/ Dr Moliner 50, 46100 Burjasot, Spain
| | - Rubén Artero
- Translational Genomics Group, Incliva Health Research Institute, Avda. Menendez Pelayo 4 acc 46010, Valencia, Spain.,Department of Genetics and Estructura de Recerca Interdisciplinar en Biotecnologia i Biomedicina (ERI BIOTECMED), Universitat de València, c/ Dr Moliner 50, 46100 Burjasot, Spain
| | - Carmen Espinós
- Program in Molecular Mechanisms of Disease, Centro de Investigación Príncipe Felipe (CIPF), c/ Eduardo Primo Yúfera no. 3, 46012 Valencia, Spain.,UPV-CIPF Joint Unit Disease Mechanisms and Nanomedicine, 46012 Valencia, Spain
| | - Máximo I Galindo
- Program in Molecular Mechanisms of Disease, Centro de Investigación Príncipe Felipe (CIPF), c/ Eduardo Primo Yúfera no. 3, 46012 Valencia, Spain .,UPV-CIPF Joint Unit Disease Mechanisms and Nanomedicine, 46012 Valencia, Spain.,Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico (IDM), Universitat Politècnica de València, Universitat de València, 46022 Valencia, Spain
| |
Collapse
|
43
|
Calafate S, Flavin W, Verstreken P, Moechars D. Loss of Bin1 Promotes the Propagation of Tau Pathology. Cell Rep 2017; 17:931-940. [PMID: 27760323 DOI: 10.1016/j.celrep.2016.09.063] [Citation(s) in RCA: 184] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Revised: 08/25/2016] [Accepted: 09/20/2016] [Indexed: 10/20/2022] Open
Abstract
Tau pathology propagates within synaptically connected neuronal circuits, but the underlying mechanisms are unclear. BIN1-amphiphysin2 is the second most prevalent genetic risk factor for late-onset Alzheimer's disease. In diseased brains, the BIN1-amphiphysin2 neuronal isoform is downregulated. Here, we show that lowering BIN1-amphiphysin2 levels in neurons promotes Tau pathology propagation whereas overexpression of neuronal BIN1-amphiphysin2 inhibits the process in two in vitro models. Increased Tau propagation is caused by increased endocytosis, given our finding that BIN1-amphiphysin2 negatively regulates endocytic flux. Furthermore, blocking endocytosis by inhibiting dynamin also reduces Tau pathology propagation. Using a galectin-3-binding assay, we show that internalized Tau aggregates damage the endosomal membrane, allowing internalized aggregates to leak into the cytoplasm to propagate pathology. Our work indicates that lower BIN1 levels promote the propagation of Tau pathology by efficiently increasing aggregate internalization by endocytosis and endosomal trafficking.
Collapse
Affiliation(s)
- Sara Calafate
- Discovery Neuroscience, Janssen Research and Development, a Division of Janssen Pharmaceutica NV, 2340 Beerse, Belgium; VIB Center for Brain and Disease Research, 3000 Leuven, Belgium; KU Leuven Department for Human Genetics, 3000 Leuven, Belgium
| | - William Flavin
- Integrative Cell Biology Program, Stritch School of Medicine, Loyola University Chicago, Maywood, IL 90270, USA
| | - Patrik Verstreken
- VIB Center for Brain and Disease Research, 3000 Leuven, Belgium; KU Leuven Department for Human Genetics, 3000 Leuven, Belgium.
| | - Diederik Moechars
- Discovery Neuroscience, Janssen Research and Development, a Division of Janssen Pharmaceutica NV, 2340 Beerse, Belgium.
| |
Collapse
|
44
|
Cowling BS, Prokic I, Tasfaout H, Rabai A, Humbert F, Rinaldi B, Nicot AS, Kretz C, Friant S, Roux A, Laporte J. Amphiphysin (BIN1) negatively regulates dynamin 2 for normal muscle maturation. J Clin Invest 2017; 127:4477-4487. [PMID: 29130937 DOI: 10.1172/jci90542] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Accepted: 10/03/2017] [Indexed: 01/25/2023] Open
Abstract
Regulation of skeletal muscle development and organization is a complex process that is not fully understood. Here, we focused on amphiphysin 2 (BIN1, also known as bridging integrator-1) and dynamin 2 (DNM2), two ubiquitous proteins implicated in membrane remodeling and mutated in centronuclear myopathies (CNMs). We generated Bin1-/- Dnm2+/- mice to decipher the physiological interplay between BIN1 and DNM2. While Bin1-/- mice die perinatally from a skeletal muscle defect, Bin1-/- Dnm2+/- mice survived at least 18 months, and had normal muscle force and intracellular organization of muscle fibers, supporting BIN1 as a negative regulator of DNM2. We next characterized muscle-specific isoforms of BIN1 and DNM2. While BIN1 colocalized with and partially inhibited DNM2 activity during muscle maturation, BIN1 had no effect on the isoform of DNM2 found in adult muscle. Together, these results indicate that BIN1 and DNM2 regulate muscle development and organization, function through a common pathway, and define BIN1 as a negative regulator of DNM2 in vitro and in vivo during muscle maturation. Our data suggest that DNM2 modulation has potential as a therapeutic approach for patients with CNM and BIN1 defects. As BIN1 is implicated in cancers, arrhythmia, and late-onset Alzheimer disease, these findings may trigger research directions and therapeutic development for these common diseases.
Collapse
Affiliation(s)
- Belinda S Cowling
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France.,Institut National de la Santé et de la Recherche Médicale, U964, Illkirch, France.,Centre National de la Recherche Scientifique, UMR7104, Illkirch, France.,Université de Strasbourg, Illkirch, France
| | - Ivana Prokic
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France.,Institut National de la Santé et de la Recherche Médicale, U964, Illkirch, France.,Centre National de la Recherche Scientifique, UMR7104, Illkirch, France.,Université de Strasbourg, Illkirch, France
| | - Hichem Tasfaout
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France.,Institut National de la Santé et de la Recherche Médicale, U964, Illkirch, France.,Centre National de la Recherche Scientifique, UMR7104, Illkirch, France.,Université de Strasbourg, Illkirch, France
| | - Aymen Rabai
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France.,Institut National de la Santé et de la Recherche Médicale, U964, Illkirch, France.,Centre National de la Recherche Scientifique, UMR7104, Illkirch, France.,Université de Strasbourg, Illkirch, France
| | - Frédéric Humbert
- Biochemistry Department, University of Geneva, Geneva, Switzerland
| | - Bruno Rinaldi
- Department of Molecular and Cellular Genetics, UMR7156, Université de Strasbourg and CNRS, Strasbourg, France
| | - Anne-Sophie Nicot
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France.,Institut National de la Santé et de la Recherche Médicale, U964, Illkirch, France.,Centre National de la Recherche Scientifique, UMR7104, Illkirch, France.,Université de Strasbourg, Illkirch, France
| | - Christine Kretz
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France.,Institut National de la Santé et de la Recherche Médicale, U964, Illkirch, France.,Centre National de la Recherche Scientifique, UMR7104, Illkirch, France.,Université de Strasbourg, Illkirch, France
| | - Sylvie Friant
- Department of Molecular and Cellular Genetics, UMR7156, Université de Strasbourg and CNRS, Strasbourg, France
| | - Aurélien Roux
- Biochemistry Department, University of Geneva, Geneva, Switzerland.,Swiss National Centre of Competence in Research Programme Chemical Biology, Geneva, Switzerland
| | - Jocelyn Laporte
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France.,Institut National de la Santé et de la Recherche Médicale, U964, Illkirch, France.,Centre National de la Recherche Scientifique, UMR7104, Illkirch, France.,Université de Strasbourg, Illkirch, France
| |
Collapse
|
45
|
Dräger NM, Nachman E, Winterhoff M, Brühmann S, Shah P, Katsinelos T, Boulant S, Teleman AA, Faix J, Jahn TR. Bin1 directly remodels actin dynamics through its BAR domain. EMBO Rep 2017; 18:2051-2066. [PMID: 28893863 DOI: 10.15252/embr.201744137] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2017] [Revised: 08/07/2017] [Accepted: 08/08/2017] [Indexed: 11/09/2022] Open
Abstract
Endocytic processes are facilitated by both curvature-generating BAR-domain proteins and the coordinated polymerization of actin filaments. Under physiological conditions, the N-BAR protein Bin1 has been shown to sense and curve membranes in a variety of cellular processes. Recent studies have identified Bin1 as a risk factor for Alzheimer's disease, although its possible pathological function in neurodegeneration is currently unknown. Here, we report that Bin1 not only shapes membranes, but is also directly involved in actin binding through its BAR domain. We observed a moderate actin bundling activity by human Bin1 and describe its ability to stabilize actin filaments against depolymerization. Moreover, Bin1 is also involved in stabilizing tau-induced actin bundles, which are neuropathological hallmarks of Alzheimer's disease. We also provide evidence for this effect in vivo, where we observed that downregulation of Bin1 in a Drosophila model of tauopathy significantly reduces the appearance of tau-induced actin inclusions. Together, these findings reveal the ability of Bin1 to modify actin dynamics and provide a possible mechanistic connection between Bin1 and tau-induced pathobiological changes of the actin cytoskeleton.
Collapse
Affiliation(s)
- Nina M Dräger
- Proteostasis in Neurodegenerative Disease (B180), Schaller Research Group at the University of Heidelberg and DKFZ, Heidelberg, Germany
| | - Eliana Nachman
- Proteostasis in Neurodegenerative Disease (B180), Schaller Research Group at the University of Heidelberg and DKFZ, Heidelberg, Germany.,German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance, Center for Molecular Biology of Heidelberg University (ZMBH), Heidelberg, Germany
| | - Moritz Winterhoff
- Institute for Biophysical Chemistry, Hannover Medical School, Hannover, Germany
| | - Stefan Brühmann
- Institute for Biophysical Chemistry, Hannover Medical School, Hannover, Germany
| | - Pranav Shah
- Schaller Research Group at CellNetworks, Department of Infectious Diseases, Virology, Heidelberg University, Heidelberg, Germany.,Cellular polarity and viral infection (F140), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Taxiarchis Katsinelos
- Proteostasis in Neurodegenerative Disease (B180), Schaller Research Group at the University of Heidelberg and DKFZ, Heidelberg, Germany
| | - Steeve Boulant
- Schaller Research Group at CellNetworks, Department of Infectious Diseases, Virology, Heidelberg University, Heidelberg, Germany.,Cellular polarity and viral infection (F140), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Aurelio A Teleman
- Signal Transduction in Cancer and Metabolism (B140), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jan Faix
- Institute for Biophysical Chemistry, Hannover Medical School, Hannover, Germany
| | - Thomas R Jahn
- Proteostasis in Neurodegenerative Disease (B180), Schaller Research Group at the University of Heidelberg and DKFZ, Heidelberg, Germany
| |
Collapse
|
46
|
Fujita N, Huang W, Lin TH, Groulx JF, Jean S, Nguyen J, Kuchitsu Y, Koyama-Honda I, Mizushima N, Fukuda M, Kiger AA. Genetic screen in Drosophila muscle identifies autophagy-mediated T-tubule remodeling and a Rab2 role in autophagy. eLife 2017; 6. [PMID: 28063257 PMCID: PMC5249261 DOI: 10.7554/elife.23367] [Citation(s) in RCA: 83] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Accepted: 12/17/2016] [Indexed: 01/04/2023] Open
Abstract
Transverse (T)-tubules make-up a specialized network of tubulated muscle cell membranes involved in excitation-contraction coupling for power of contraction. Little is known about how T-tubules maintain highly organized structures and contacts throughout the contractile system despite the ongoing muscle remodeling that occurs with muscle atrophy, damage and aging. We uncovered an essential role for autophagy in T-tubule remodeling with genetic screens of a developmentally regulated remodeling program in Drosophila abdominal muscles. Here, we show that autophagy is both upregulated with and required for progression through T-tubule disassembly stages. Along with known mediators of autophagosome-lysosome fusion, our screens uncovered an unexpected shared role for Rab2 with a broadly conserved function in autophagic clearance. Rab2 localizes to autophagosomes and binds to HOPS complex members, suggesting a direct role in autophagosome tethering/fusion. Together, the high membrane flux with muscle remodeling permits unprecedented analysis both of T-tubule dynamics and fundamental trafficking mechanisms. DOI:http://dx.doi.org/10.7554/eLife.23367.001
Collapse
Affiliation(s)
- Naonobu Fujita
- Section of Cell and Developmental Biology, Division of Biological Sciences, University of California, San Diego, La Jolla, United States.,Laboratory of Membrane Trafficking Mechanisms, Department of Developmental Biology and Neurosciences, Graduate School of Life Sciences, Tohoku University, Sendai, Japan
| | - Wilson Huang
- Section of Cell and Developmental Biology, Division of Biological Sciences, University of California, San Diego, La Jolla, United States
| | - Tzu-Han Lin
- Section of Cell and Developmental Biology, Division of Biological Sciences, University of California, San Diego, La Jolla, United States
| | - Jean-Francois Groulx
- Section of Cell and Developmental Biology, Division of Biological Sciences, University of California, San Diego, La Jolla, United States
| | - Steve Jean
- Section of Cell and Developmental Biology, Division of Biological Sciences, University of California, San Diego, La Jolla, United States
| | - Jen Nguyen
- Section of Cell and Developmental Biology, Division of Biological Sciences, University of California, San Diego, La Jolla, United States
| | - Yoshihiko Kuchitsu
- Laboratory of Membrane Trafficking Mechanisms, Department of Developmental Biology and Neurosciences, Graduate School of Life Sciences, Tohoku University, Sendai, Japan
| | - Ikuko Koyama-Honda
- Department of Biochemistry and Molecular Biology, Graduate School and Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Noboru Mizushima
- Department of Biochemistry and Molecular Biology, Graduate School and Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Mitsunori Fukuda
- Laboratory of Membrane Trafficking Mechanisms, Department of Developmental Biology and Neurosciences, Graduate School of Life Sciences, Tohoku University, Sendai, Japan
| | - Amy A Kiger
- Section of Cell and Developmental Biology, Division of Biological Sciences, University of California, San Diego, La Jolla, United States
| |
Collapse
|
47
|
Walmsley GL, Blot S, Venner K, Sewry C, Laporte J, Blondelle J, Barthélémy I, Maurer M, Blanchard-Gutton N, Pilot-Storck F, Tiret L, Piercy RJ. Progressive Structural Defects in Canine Centronuclear Myopathy Indicate a Role for HACD1 in Maintaining Skeletal Muscle Membrane Systems. THE AMERICAN JOURNAL OF PATHOLOGY 2016; 187:441-456. [PMID: 27939133 DOI: 10.1016/j.ajpath.2016.10.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Revised: 10/12/2016] [Accepted: 10/17/2016] [Indexed: 11/16/2022]
Abstract
Mutations in HACD1/PTPLA cause recessive congenital myopathies in humans and dogs. Hydroxyacyl-coA dehydratases are required for elongation of very long chain fatty acids, and HACD1 has a role in early myogenesis, but the functions of this striated muscle-specific enzyme in more differentiated skeletal muscle remain unknown. Canine HACD1 deficiency is histopathologically classified as a centronuclear myopathy (CNM). We investigated the hypothesis that muscle from HACD1-deficient dogs has membrane abnormalities in common with CNMs with different genetic causes. We found progressive changes in tubuloreticular and sarcolemmal membranes and mislocalized triads and mitochondria in skeletal muscle from animals deficient in HACD1. Furthermore, comparable membranous abnormalities in cultured HACD1-deficient myotubes provide additional evidence that these defects are a primary consequence of altered HACD1 expression. Our novel findings, including T-tubule dilatation and disorganization, associated with defects in this additional CNM-associated gene provide a definitive pathophysiologic link with these disorders, confirm that dogs deficient in HACD1 are relevant models, and strengthen the evidence for a unifying pathogenesis in CNMs via defective membrane trafficking and excitation-contraction coupling in muscle. These results build on previous work by determining further functional roles of HACD1 in muscle and provide new insight into the pathology and pathogenetic mechanisms of HACD1 CNM. Consequently, alterations in membrane properties associated with HACD1 mutations should be investigated in humans with related phenotypes.
Collapse
Affiliation(s)
- Gemma L Walmsley
- Comparative Neuromuscular Diseases Laboratory, Department of Clinical Sciences and Services, Royal Veterinary College, London, United Kingdom.
| | - Stéphane Blot
- French National Institute of Health and Medical Research (Inserm), Mondor Institute of Biomedical Research (IMRB) U955-E10 Biology of the Neuromuscular System, Créteil, France; University of Paris East, Alfort School of Veterinary Medicine (EnvA), Maisons-Alfort, France
| | - Kerrie Venner
- Electron Microscopy Unit, Institute of Neurology, London, United Kingdom
| | - Caroline Sewry
- Dubowitz Neuromuscular Centre, University College London Institute of Child Health and Great Ormond Street Hospital, London, United Kingdom
| | - Jocelyn Laporte
- Department of Translational Medicine and Neurogenetics, Institute of Genetics and Cellular and Molecular Biology (IGBMC), Inserm U964, CNRS UMR7104, Strasbourg University, Illkirch, France
| | - Jordan Blondelle
- French National Institute of Health and Medical Research (Inserm), Mondor Institute of Biomedical Research (IMRB) U955-E10 Biology of the Neuromuscular System, Créteil, France; University of Paris East, Alfort School of Veterinary Medicine (EnvA), Maisons-Alfort, France
| | - Inès Barthélémy
- French National Institute of Health and Medical Research (Inserm), Mondor Institute of Biomedical Research (IMRB) U955-E10 Biology of the Neuromuscular System, Créteil, France; University of Paris East, Alfort School of Veterinary Medicine (EnvA), Maisons-Alfort, France
| | - Marie Maurer
- French National Institute of Health and Medical Research (Inserm), Mondor Institute of Biomedical Research (IMRB) U955-E10 Biology of the Neuromuscular System, Créteil, France; University of Paris East, Alfort School of Veterinary Medicine (EnvA), Maisons-Alfort, France
| | - Nicolas Blanchard-Gutton
- French National Institute of Health and Medical Research (Inserm), Mondor Institute of Biomedical Research (IMRB) U955-E10 Biology of the Neuromuscular System, Créteil, France; University of Paris East, Alfort School of Veterinary Medicine (EnvA), Maisons-Alfort, France
| | - Fanny Pilot-Storck
- French National Institute of Health and Medical Research (Inserm), Mondor Institute of Biomedical Research (IMRB) U955-E10 Biology of the Neuromuscular System, Créteil, France; University of Paris East, Alfort School of Veterinary Medicine (EnvA), Maisons-Alfort, France
| | - Laurent Tiret
- French National Institute of Health and Medical Research (Inserm), Mondor Institute of Biomedical Research (IMRB) U955-E10 Biology of the Neuromuscular System, Créteil, France; University of Paris East, Alfort School of Veterinary Medicine (EnvA), Maisons-Alfort, France
| | - Richard J Piercy
- Comparative Neuromuscular Diseases Laboratory, Department of Clinical Sciences and Services, Royal Veterinary College, London, United Kingdom
| |
Collapse
|
48
|
Fraysse B, Guicheney P, Bitoun M. Calcium homeostasis alterations in a mouse model of the Dynamin 2-related centronuclear myopathy. Biol Open 2016; 5:1691-1696. [PMID: 27870637 PMCID: PMC5155535 DOI: 10.1242/bio.020263] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Autosomal dominant centronuclear myopathy (CNM) is a rare congenital myopathy characterized by centrally located nuclei in muscle fibers. CNM results from mutations in the gene encoding dynamin 2 (DNM2), a large GTPase involved in endocytosis, intracellular membrane trafficking, and cytoskeleton regulation. We developed a knock-in mouse model expressing the most frequent DNM2-CNM mutation; i.e. the KI-Dnm2R465W model. Heterozygous (HTZ) KI-Dnm2 mice progressively develop muscle atrophy, impairment of contractile properties, histopathological abnormalities, and elevated cytosolic calcium concentration. Here, we aim at better characterizing the calcium homeostasis impairment in extensor digitorum longus (EDL) and soleus muscles from adult HTZ KI-Dnm2 mice. We demonstrate abnormal contractile properties and cytosolic Ca2+ concentration in EDL but not soleus muscles showing that calcium impairment is correlated with muscle weakness and might be a determinant factor of the spatial muscle involvement. In addition, the elevated cytosolic Ca2+ concentration in EDL muscles is associated with an increased sarcolemmal permeability to Ca2+ and releasable Ca2+ content from the sarcoplasmic reticulum. However, amplitude and kinetics characteristics of the calcium transient appear unchanged. This suggests that calcium defect is probably not a primary cause of decreased force generation by compromised sarcomere shortening but may be involved in long-term deleterious consequences on muscle physiology. Our results highlight the first pathomechanism which may explain the spatial muscle involvement occurring in DNM2-related CNM and open the way toward development of a therapeutic approach to normalize calcium content. Summary: Dynamin 2 mutations cause centronuclear myopathy via unclear mechanisms. We show in a mouse model that changes in cytosolic calcium via incorrect membrane permeability correlate with muscle weakness.
Collapse
Affiliation(s)
- Bodvaël Fraysse
- Atlantic Gene Therapies, INSERM UMR 1089, Université de Nantes, CHU de Nantes, Nantes 44200, France
| | - Pascale Guicheney
- INSERM, UMR_S1166, Sorbonne Universités, UPMC Univ Paris 06, UMR_S1166, Institute of Cardiometabolism and Nutrition (ICAN), Paris 75013, France
| | - Marc Bitoun
- Research Center for Myology, UPMC Univ Paris 06 and INSERM UMR_S974, CNRS FRE 3617, Institute of Myology, Paris 75013, France
| |
Collapse
|
49
|
Stanishneva-Konovalova T, Derkacheva N, Polevova S, Sokolova O. The Role of BAR Domain Proteins in the Regulation of Membrane Dynamics. Acta Naturae 2016; 8:60-69. [PMID: 28050267 PMCID: PMC5199207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Indexed: 11/09/2022] Open
Abstract
Many cellular processes are associated with membrane remodeling. The BAR domain protein family plays a key role in the formation and detection of local membrane curvatures and in attracting other proteins, including the regulators of actin dynamics. Based on their structural and phylogenetic properties, BAR domains are divided into several groups which affect membrane in various ways and perform different functions in cells. However, recent studies have uncovered evidence of functional differences even within the same group. This review discusses the principles underlying the interactions of different groups of BAR domains, and their individual representatives ,with membranes.
Collapse
Affiliation(s)
| | - N.I. Derkacheva
- A.I. Evdokimov Moscow State University of Medicine and Dentistry, Department of Biochemistry, Delegatskaya Str. 20, Bld 1, Moscow, 127473, Russia
| | - S.V. Polevova
- Lomonosov Moscow State University, Faculty of Biology, Leninskie Gory 1, Bld 12, Moscow, 119234 , Russia
| | - O.S. Sokolova
- Lomonosov Moscow State University, Faculty of Biology, Leninskie Gory 1, Bld 12, Moscow, 119234 , Russia
| |
Collapse
|
50
|
De Rossi P, Buggia-Prévot V, Clayton BLL, Vasquez JB, van Sanford C, Andrew RJ, Lesnick R, Botté A, Deyts C, Salem S, Rao E, Rice RC, Parent A, Kar S, Popko B, Pytel P, Estus S, Thinakaran G. Predominant expression of Alzheimer's disease-associated BIN1 in mature oligodendrocytes and localization to white matter tracts. Mol Neurodegener 2016; 11:59. [PMID: 27488240 PMCID: PMC4973113 DOI: 10.1186/s13024-016-0124-1] [Citation(s) in RCA: 84] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2016] [Accepted: 07/27/2016] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Genome-wide association studies have identified BIN1 within the second most significant susceptibility locus in late-onset Alzheimer's disease (AD). BIN1 undergoes complex alternative splicing to generate multiple isoforms with diverse functions in multiple cellular processes including endocytosis and membrane remodeling. An increase in BIN1 expression in AD and an interaction between BIN1 and Tau have been reported. However, disparate descriptions of BIN1 expression and localization in the brain previously reported in the literature and the lack of clarity on brain BIN1 isoforms present formidable challenges to our understanding of how genetic variants in BIN1 increase the risk for AD. METHODS In this study, we analyzed BIN1 mRNA and protein levels in human brain samples from individuals with or without AD. In addition, we characterized the BIN1 expression and isoform diversity in human and rodent tissue by immunohistochemistry and immunoblotting using a panel of BIN1 antibodies. RESULTS Here, we report on BIN1 isoform diversity in the human brain and document alterations in the levels of select BIN1 isoforms in individuals with AD. In addition, we report striking BIN1 localization to white matter tracts in rodent and the human brain, and document that the large majority of BIN1 is expressed in mature oligodendrocytes whereas neuronal BIN1 represents a minor fraction. This predominant non-neuronal BIN1 localization contrasts with the strict neuronal expression and presynaptic localization of the BIN1 paralog, Amphiphysin 1. We also observe upregulation of BIN1 at the onset of postnatal myelination in the brain and during differentiation of cultured oligodendrocytes. Finally, we document that the loss of BIN1 significantly correlates with the extent of demyelination in multiple sclerosis lesions. CONCLUSION Our study provides new insights into the brain distribution and cellular expression of an important risk factor associated with late-onset AD. We propose that efforts to define how genetic variants in BIN1 elevate the risk for AD would behoove to consider BIN1 function in the context of its main expression in mature oligodendrocytes and the potential for a role of BIN1 in the membrane remodeling that accompanies the process of myelination.
Collapse
Affiliation(s)
- Pierre De Rossi
- Department of Neurobiology, The University of Chicago, JFK R212, 924 East 57th Street, Chicago, IL 60637 USA
| | - Virginie Buggia-Prévot
- Department of Neurobiology, The University of Chicago, JFK R212, 924 East 57th Street, Chicago, IL 60637 USA
| | | | - Jared B. Vasquez
- Sanders-Brown Center on Aging and Department of Physiology, University of Kentucky, Lexington, KY 40536 USA
| | - Carson van Sanford
- Sanders-Brown Center on Aging and Department of Physiology, University of Kentucky, Lexington, KY 40536 USA
| | - Robert J. Andrew
- Department of Neurobiology, The University of Chicago, JFK R212, 924 East 57th Street, Chicago, IL 60637 USA
| | - Ruben Lesnick
- Department of Neurobiology, The University of Chicago, JFK R212, 924 East 57th Street, Chicago, IL 60637 USA
| | - Alexandra Botté
- Department of Neurobiology, The University of Chicago, JFK R212, 924 East 57th Street, Chicago, IL 60637 USA
| | - Carole Deyts
- Department of Neurobiology, The University of Chicago, JFK R212, 924 East 57th Street, Chicago, IL 60637 USA
| | - Someya Salem
- Department of Neurobiology, The University of Chicago, JFK R212, 924 East 57th Street, Chicago, IL 60637 USA
| | - Eshaan Rao
- Department of Neurobiology, The University of Chicago, JFK R212, 924 East 57th Street, Chicago, IL 60637 USA
| | - Richard C. Rice
- Department of Neurobiology, The University of Chicago, JFK R212, 924 East 57th Street, Chicago, IL 60637 USA
| | - Angèle Parent
- Department of Neurobiology, The University of Chicago, JFK R212, 924 East 57th Street, Chicago, IL 60637 USA
| | - Satyabrata Kar
- Centre for prions and protein folding diseases, University of Alberta, Edmonton, AB T6G 2B7 Canada
| | - Brian Popko
- Department of Neurology, The University of Chicago, Chicago, IL 60637 USA
| | - Peter Pytel
- Department of Pathology, The University of Chicago, Chicago, IL 60637 USA
| | - Steven Estus
- Sanders-Brown Center on Aging and Department of Physiology, University of Kentucky, Lexington, KY 40536 USA
| | - Gopal Thinakaran
- Department of Neurobiology, The University of Chicago, JFK R212, 924 East 57th Street, Chicago, IL 60637 USA
- Department of Neurology, The University of Chicago, Chicago, IL 60637 USA
- Department of Pathology, The University of Chicago, Chicago, IL 60637 USA
| |
Collapse
|