1
|
Wu XS, Zhang Z, Jin Y, Mushtaheed A, Wu LG. Actin maintains synaptic transmission by restraining vesicle release probability. iScience 2025; 28:112000. [PMID: 40109375 PMCID: PMC11919605 DOI: 10.1016/j.isci.2025.112000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 10/22/2024] [Accepted: 02/07/2025] [Indexed: 03/22/2025] Open
Abstract
Despite decades of pharmacological studies, how the ubiquitous cytoskeletal actin regulates synaptic transmission remains poorly understood. We addressed this issue with a tissue-specific knockout of actin β-isoform or γ-isoform, combined with recordings of postsynaptic EPSCs, presynaptic capacitance jumps or fluorescent synaptophysin-pHluorin changes, and electron microscopy in large calyx-type and small conventional hippocampal synapses. We found that actin restrains basal synaptic transmission during single action potential firings by lowering the readily releasable vesicle's release probability. Such an inhibition of basal synaptic transmission is turned into facilitation during repetitive firings by slowing down depletion of the readily releasable vesicle pool and, thus, short-term synaptic depression, leading to more effective synaptic transmission for a longer time. These mechanisms, together with the previous finding that actin promotes vesicle replenishment to the readily releasable pool, may control synaptic transmission and short-term synaptic plasticity at many synapses, contributing to neurological disorders caused by actin cytoskeleton impairment.
Collapse
Affiliation(s)
- Xin-Sheng Wu
- National Institute of Neurological Disorders and Stroke, 35 Convent Dr., Bethesda, MD 20892, USA
| | - Zhen Zhang
- National Institute of Neurological Disorders and Stroke, 35 Convent Dr., Bethesda, MD 20892, USA
- Office of Genetic Drugs, Center for Drug Evaluation and Research, Food and Drug Administration, 10903 New Hampshire Avenue, Silver Spring, MD 20993, USA
| | - Yinghui Jin
- National Institute of Neurological Disorders and Stroke, 35 Convent Dr., Bethesda, MD 20892, USA
| | - Afreen Mushtaheed
- National Institute of Neurological Disorders and Stroke, 35 Convent Dr., Bethesda, MD 20892, USA
| | - Ling-Gang Wu
- National Institute of Neurological Disorders and Stroke, 35 Convent Dr., Bethesda, MD 20892, USA
| |
Collapse
|
2
|
Sundby LJ, Hawbaker KM, Powers J, Southern WM, Johnson EE, Patrinostro X, Perrin BJ, Ervasti JM. The complete absence of cytoplasmic γ-actin results in no discernible phenotype in mice or primary fibroblasts. FEBS J 2025. [PMID: 40109120 DOI: 10.1111/febs.70075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Revised: 12/26/2024] [Accepted: 02/06/2025] [Indexed: 03/22/2025]
Abstract
Mice and primary fibroblasts derived from mouse embryos completely lacking cytoplasmic β-actin, because the Actb gene was engineered to instead express γ-actin protein, have previously been found to be virtually devoid of phenotype. Here, we report the characterization of mice and mouse embryonic fibroblasts homozygous for an Actg1 allele edited to translate β-actin instead of γ-actin (Actg1-coding beta; Actg1c-b/c-b), which resulted in mice and fibroblasts that are devoid of γ-actin. We demonstrate that these Actg1c-b/c-b mice present with no measurable phenotype in survival, body mass, activity, muscle contractility, or auditory function. Primary fibroblasts derived from Actg1c-b/c-b mouse embryos were still proliferative, with several measured parameters of cell motility not different from wild type. From these and previous data, we conclude that β- and γ-actin proteins are redundant in primary embryonic fibroblasts and during normal mouse development.
Collapse
Affiliation(s)
- Lauren J Sundby
- Program in Molecular, Cellular, Developmental Biology, and Genetics, University of Minnesota, Minneapolis, MN, USA
| | - Katelin M Hawbaker
- Department of Biology, Indiana University - Indianapolis, Indianapolis, IN, USA
| | - Jacob Powers
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN, USA
| | - William M Southern
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN, USA
| | - Erynn E Johnson
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN, USA
| | - Xiaobai Patrinostro
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN, USA
| | - Benjamin J Perrin
- Department of Biology, Indiana University - Indianapolis, Indianapolis, IN, USA
| | - James M Ervasti
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
3
|
Christophers B, Leahy SN, Soffar DB, von Saucken VE, Broadie K, Baylies MK. Muscle cofilin alters neuromuscular junction postsynaptic development to strengthen functional neurotransmission. Development 2024; 151:dev202558. [PMID: 38869008 PMCID: PMC11266751 DOI: 10.1242/dev.202558] [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: 11/21/2023] [Accepted: 06/05/2024] [Indexed: 06/14/2024]
Abstract
Cofilin, an actin-severing protein, plays key roles in muscle sarcomere addition and maintenance. Our previous work found that Drosophila cofilin (DmCFL) knockdown in muscle causes progressive deterioration of muscle structure and function and produces features seen in nemaline myopathy caused by cofilin mutations. We hypothesized that disruption of actin cytoskeleton dynamics by DmCFL knockdown would impact other aspects of muscle development, and, thus, conducted an RNA-sequencing analysis that unexpectedly revealed upregulated expression of numerous neuromuscular junction (NMJ) genes. We found that DmCFL is enriched in the muscle postsynaptic compartment and that DmCFL muscle knockdown causes F-actin disorganization in this subcellular domain prior to the sarcomere defects observed later in development. Despite NMJ gene expression changes, we found no significant changes in gross presynaptic Bruchpilot active zones or total postsynaptic glutamate receptor levels. However, DmCFL knockdown resulted in mislocalization of GluRIIA class glutamate receptors in more deteriorated muscles and strongly impaired NMJ transmission strength. These findings expand our understanding of the roles of cofilin in muscle to include NMJ structural development and suggest that NMJ defects may contribute to the pathophysiology of nemaline myopathy.
Collapse
Affiliation(s)
- Briana Christophers
- Weill Cornell–Rockefeller–Sloan Kettering Tri-Institutional MD-PhD Program, New York, NY 10065, USA
- Biochemistry, Cell & Developmental Biology, and Molecular Biology (BCMB) program, Weill Cornell Graduate School of Medical Sciences, New York, NY 10065, USA
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering, Cancer Center, New York, NY 10065, USA
| | - Shannon N. Leahy
- Department of Biological Sciences, Vanderbilt University and Medical Center, Nashville, TN 37235, USA
| | - David B. Soffar
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering, Cancer Center, New York, NY 10065, USA
| | - Victoria E. von Saucken
- Weill Cornell–Rockefeller–Sloan Kettering Tri-Institutional MD-PhD Program, New York, NY 10065, USA
- Biochemistry, Cell & Developmental Biology, and Molecular Biology (BCMB) program, Weill Cornell Graduate School of Medical Sciences, New York, NY 10065, USA
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering, Cancer Center, New York, NY 10065, USA
| | - Kendal Broadie
- Department of Biological Sciences, Vanderbilt University and Medical Center, Nashville, TN 37235, USA
- Kennedy Center for Research on Human Development, Vanderbilt University and Medical Center, Nashville, TN 37235, USA
- Vanderbilt Brain Institute, Vanderbilt University and Medical Center, Nashville, TN 37235, USA
| | - Mary K. Baylies
- Biochemistry, Cell & Developmental Biology, and Molecular Biology (BCMB) program, Weill Cornell Graduate School of Medical Sciences, New York, NY 10065, USA
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering, Cancer Center, New York, NY 10065, USA
| |
Collapse
|
4
|
Ghiselli S, Parmeggiani G, Zambonini G, Cuda D. Hearing Loss in Baraitser-Winter Syndrome: Case Reports and Review of the Literature. J Clin Med 2024; 13:1500. [PMID: 38592426 PMCID: PMC10935159 DOI: 10.3390/jcm13051500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 03/01/2024] [Accepted: 03/02/2024] [Indexed: 04/10/2024] Open
Abstract
Background: Baraitser-Winter Syndrome (BRWS) is a rare autosomal dominant condition associated with hearing loss (HL). In the literature, two types of this condition are reported, Baraitser-Winter type 1 (BRWS1) and type 2 (BRWS2) produced by specific pathogenetic variants of two different genes, ACTB for BRWS1 and ACTG1 for BRWS2. In addition to syndromic BRWS2, some pathogenic variants in ACTG1 are associated also to another pathologic entity, the "Autosomal dominant non-syndromic hearing loss 20/26". In these syndromes, typical craniofacial features, sensory impairment (vision and hearing) and intellectual disabilities are frequently present. Heart anomalies, renal and gastrointestinal involvement and seizure are also common. Wide inter- and intra-familial variety in the phenotypic spectrum is reported. Some phenotypic aspects of these syndromes are not yet fully described, such as the degree and progression of HL, and better knowledge of them could be useful for correct follow-up and treatment. Methods and Results: In this study, we report two cases of children with HL and diagnosis of BRWS and a review of the current literature on HL in these syndromes.
Collapse
Affiliation(s)
- Sara Ghiselli
- Department of Otorhinolaryngology, AUSL Piacenza, 29121 Piacenza, Italy; (G.Z.); (D.C.)
| | | | - Giulia Zambonini
- Department of Otorhinolaryngology, AUSL Piacenza, 29121 Piacenza, Italy; (G.Z.); (D.C.)
| | - Domenico Cuda
- Department of Otorhinolaryngology, AUSL Piacenza, 29121 Piacenza, Italy; (G.Z.); (D.C.)
- Department of Medicine and Surgery, University of Parma, 43121 Parma, Italy
| |
Collapse
|
5
|
Christophers B, Leahy SN, Soffar DB, von Saucken VE, Broadie K, Baylies MK. Muscle cofilin alters neuromuscular junction postsynaptic development to strengthen functional neurotransmission. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.21.568166. [PMID: 38045306 PMCID: PMC10690168 DOI: 10.1101/2023.11.21.568166] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/05/2023]
Abstract
Cofilin, an actin severing protein, plays critical roles in muscle sarcomere addition and maintenance. Our previous work has shown Drosophila cofilin (DmCFL) knockdown causes progressive deterioration of muscle structure and function and produces features seen in nemaline myopathy (NM) caused by cofilin mutations. We hypothesized that disruption of actin cytoskeleton dynamics by DmCFL knockdown would impact other aspects of muscle development, and, thus, conducted an RNA sequencing analysis which unexpectedly revealed upregulated expression of numerous neuromuscular junction (NMJ) genes. We found that DmCFL is enriched in the muscle postsynaptic compartment and that DmCFL deficiency causes F-actin disorganization in this subcellular domain prior to the sarcomere defects observed later in development. Despite NMJ gene expression changes, we found no significant changes in gross presynaptic Bruchpilot active zones or total postsynaptic glutamate receptor levels. However, DmCFL knockdown results in mislocalization of glutamate receptors containing the GluRIIA subunit in more deteriorated muscles and neurotransmission strength is strongly impaired. These findings expand our understanding of cofilin's roles in muscle to include NMJ structural development and suggest that NMJ defects may contribute to NM pathophysiology.
Collapse
Affiliation(s)
- Briana Christophers
- Weill Cornell–Rockefeller–Sloan Kettering Tri-Institutional MD-PhD Program, New York, NY, 10065, USA
- Biochemistry, Cell & Developmental Biology, and Molecular Biology (BCMB) program, Weill Cornell Graduate School of Medical Sciences, New York, NY 10065, USA
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering, Cancer Center, New York, NY 10065, USA
| | - Shannon N. Leahy
- Department of Biological Sciences, Vanderbilt University and Medical Center, Nashville, TN 37235, USA
| | - David B. Soffar
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering, Cancer Center, New York, NY 10065, USA
| | - Victoria E. von Saucken
- Weill Cornell–Rockefeller–Sloan Kettering Tri-Institutional MD-PhD Program, New York, NY, 10065, USA
- Biochemistry, Cell & Developmental Biology, and Molecular Biology (BCMB) program, Weill Cornell Graduate School of Medical Sciences, New York, NY 10065, USA
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering, Cancer Center, New York, NY 10065, USA
| | - Kendal Broadie
- Department of Biological Sciences, Vanderbilt University and Medical Center, Nashville, TN 37235, USA
- Kennedy Center for Research on Human Development, Vanderbilt University and Medical Center, Nashville, TN 37235, USA
- Vanderbilt Brain Institute, Vanderbilt University and Medical Center, Nashville, TN 37235, USA
| | - Mary K. Baylies
- Biochemistry, Cell & Developmental Biology, and Molecular Biology (BCMB) program, Weill Cornell Graduate School of Medical Sciences, New York, NY 10065, USA
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering, Cancer Center, New York, NY 10065, USA
| |
Collapse
|
6
|
Yun Y, Wu R, He X, Qin X, Chen L, Sha L, Yun X, Nishiumi T, Borjigin G. Integrated Transcriptome Analysis of miRNAs and mRNAs in the Skeletal Muscle of Wuranke Sheep. Genes (Basel) 2023; 14:2034. [PMID: 38002977 PMCID: PMC10671749 DOI: 10.3390/genes14112034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Revised: 10/26/2023] [Accepted: 10/30/2023] [Indexed: 11/26/2023] Open
Abstract
MicroRNAs (miRNAs) are regarded as important regulators in skeletal muscle development. To reveal the regulatory roles of miRNAs and their target mRNAs underlying the skeletal muscle development of Wuranke sheep, we investigated the miRNA and mRNA expression profiles in the biceps femoris of these sheep at the fetal (3 months of gestation) and 3- and 15-month-old postnatal stages. Consequently, a total of 1195 miRNAs and 24,959 genes were identified. Furthermore, 474, 461, and 54 differentially expressed miRNAs (DEMs) and 6783, 7407, and 78 differentially expressed genes (DEGs) were detected among three comparative groups. Functional analysis demonstrated that the target mRNAs of the DEMs were enriched in multiple pathways related to muscle development. Moreover, the interactions among several predicted miRNA-mRNA pairs (oar-miR-133-HDAC1, oar-miR-1185-5p-MYH1/HADHA/OXCT1, and PC-5p-3703_578-INSR/ACTG1) that potentially affect skeletal muscle development were verified using dual-luciferase reporter assays. In this study, we identified the miRNA and mRNA differences in the skeletal muscle of Wuranke sheep at different developmental stages and revealed that a series of candidate miRNA-mRNA pairs may act as modulators of muscle development. These results will contribute to future studies on the function of miRNAs and their target mRNAs during skeletal muscle development in Wuranke sheep.
Collapse
Affiliation(s)
- Yueying Yun
- College of Food Science and Engineering, Inner Mongolia Agricultural University, Hohhot 010018, China; (Y.Y.); (X.H.); (X.Q.); (L.C.); (L.S.); (X.Y.)
- School of Life Science and Technology, Inner Mongolia University of Science and Technology, Baotou 014010, China
| | - Rihan Wu
- College of Biochemistry and Engineering, Hohhot Vocational College, Hohhot 010051, China;
| | - Xige He
- College of Food Science and Engineering, Inner Mongolia Agricultural University, Hohhot 010018, China; (Y.Y.); (X.H.); (X.Q.); (L.C.); (L.S.); (X.Y.)
| | - Xia Qin
- College of Food Science and Engineering, Inner Mongolia Agricultural University, Hohhot 010018, China; (Y.Y.); (X.H.); (X.Q.); (L.C.); (L.S.); (X.Y.)
| | - Lu Chen
- College of Food Science and Engineering, Inner Mongolia Agricultural University, Hohhot 010018, China; (Y.Y.); (X.H.); (X.Q.); (L.C.); (L.S.); (X.Y.)
| | - Lina Sha
- College of Food Science and Engineering, Inner Mongolia Agricultural University, Hohhot 010018, China; (Y.Y.); (X.H.); (X.Q.); (L.C.); (L.S.); (X.Y.)
| | - Xueyan Yun
- College of Food Science and Engineering, Inner Mongolia Agricultural University, Hohhot 010018, China; (Y.Y.); (X.H.); (X.Q.); (L.C.); (L.S.); (X.Y.)
| | - Tadayuki Nishiumi
- Division of Life and Food Science, Graduate School of Science and Technology, Niigata University, Niigata 950-2181, Japan
| | - Gerelt Borjigin
- College of Food Science and Engineering, Inner Mongolia Agricultural University, Hohhot 010018, China; (Y.Y.); (X.H.); (X.Q.); (L.C.); (L.S.); (X.Y.)
| |
Collapse
|
7
|
Coscarella IL, Landim-Vieira M, Rastegarpouyani H, Chase PB, Irianto J, Pinto JR. Nucleus Mechanosensing in Cardiomyocytes. Int J Mol Sci 2023; 24:13341. [PMID: 37686151 PMCID: PMC10487505 DOI: 10.3390/ijms241713341] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 08/20/2023] [Accepted: 08/22/2023] [Indexed: 09/10/2023] Open
Abstract
Cardiac muscle contraction is distinct from the contraction of other muscle types. The heart continuously undergoes contraction-relaxation cycles throughout an animal's lifespan. It must respond to constantly varying physical and energetic burdens over the short term on a beat-to-beat basis and relies on different mechanisms over the long term. Muscle contractility is based on actin and myosin interactions that are regulated by cytoplasmic calcium ions. Genetic variants of sarcomeric proteins can lead to the pathophysiological development of cardiac dysfunction. The sarcomere is physically connected to other cytoskeletal components. Actin filaments, microtubules and desmin proteins are responsible for these interactions. Therefore, mechanical as well as biochemical signals from sarcomeric contractions are transmitted to and sensed by other parts of the cardiomyocyte, particularly the nucleus which can respond to these stimuli. Proteins anchored to the nuclear envelope display a broad response which remodels the structure of the nucleus. In this review, we examine the central aspects of mechanotransduction in the cardiomyocyte where the transmission of mechanical signals to the nucleus can result in changes in gene expression and nucleus morphology. The correlation of nucleus sensing and dysfunction of sarcomeric proteins may assist the understanding of a wide range of functional responses in the progress of cardiomyopathic diseases.
Collapse
Affiliation(s)
| | - Maicon Landim-Vieira
- Department of Biomedical Sciences, Florida State University, Tallahassee, FL 32306, USA
| | - Hosna Rastegarpouyani
- Department of Biological Science, Florida State University, Tallahassee, FL 32306, USA
- Institute for Molecular Biophysics, Florida State University, Tallahassee, FL 32306, USA
| | - Prescott Bryant Chase
- Department of Biological Science, Florida State University, Tallahassee, FL 32306, USA
| | - Jerome Irianto
- Department of Biomedical Sciences, Florida State University, Tallahassee, FL 32306, USA
| | - Jose Renato Pinto
- Department of Biomedical Sciences, Florida State University, Tallahassee, FL 32306, USA
| |
Collapse
|
8
|
Mathur PD, Zou J, Neiswanger G, Zhu D, Wang Y, Almishaal AA, Vashist D, Hammond HK, Park AH, Yang J. Adenylyl cyclase 6 plays a minor role in the mouse inner ear and retina. Sci Rep 2023; 13:7075. [PMID: 37127773 PMCID: PMC10151359 DOI: 10.1038/s41598-023-34361-y] [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: 12/24/2022] [Accepted: 04/28/2023] [Indexed: 05/03/2023] Open
Abstract
Adenylyl cyclase 6 (AC6) synthesizes second messenger cAMP in G protein-coupled receptor (GPCR) signaling. In cochlear hair cells, AC6 distribution relies on an adhesion GPCR, ADGRV1, which is associated with Usher syndrome (USH), a condition of combined hearing and vision loss. ADGRV1 is a component of the USH type 2 (USH2) protein complex in hair cells and photoreceptors. However, the role of AC6 in the inner ear and retina has not been explored. Here, we found that AC6 distribution in hair cells depends on the USH2 protein complex integrity. Several known AC6 regulators and effectors, which were previously reported to participate in ADGRV1 signaling in vitro, are localized to the stereociliary compartments that overlap with AC6 distribution in hair cells. In young AC6 knockout (Adcy6-/-) mice, the activity of cAMP-dependent protein kinase, but not Akt kinase, is altered in cochleas, while both kinases are normal in vestibular organs. Adult Adcy6-/- mice however exhibit normal hearing function. AC6 is expressed in mouse retinas but rarely in photoreceptors. Adcy6-/- mice have slightly enhanced photopic but normal scotopic vision. Therefore, AC6 may participate in the ADGRV1 signaling in hair cells but AC6 is not essential for cochlear and retinal development and maintenance.
Collapse
Affiliation(s)
- Pranav Dinesh Mathur
- Department of Ophthalmology and Visual Sciences, Moran Eye Center, University of Utah, Salt Lake City, UT, 84132, USA
- Department of Neurobiology, University of Utah, Salt Lake City, UT, 84132, USA
- Vecprobio Inc., San Diego, CA, 92126, USA
| | - Junhuang Zou
- Department of Ophthalmology and Visual Sciences, Moran Eye Center, University of Utah, Salt Lake City, UT, 84132, USA
| | - Grace Neiswanger
- Department of Ophthalmology and Visual Sciences, Moran Eye Center, University of Utah, Salt Lake City, UT, 84132, USA
| | - Daniel Zhu
- Department of Ophthalmology and Visual Sciences, Moran Eye Center, University of Utah, Salt Lake City, UT, 84132, USA
| | - Yong Wang
- Division of Otolaryngology, Department of Surgery, University of Utah, Salt Lake City, UT, 84132, USA
| | - Ali A Almishaal
- Department of Communication Sciences and Disorders, University of Utah, Salt Lake City, UT, 84112, USA
- Department of Speech-Language Pathology and Audiology, College of Applied Medical Sciences, University of Hail, Hail, 81451, Saudi Arabia
| | - Deepti Vashist
- Department of Ophthalmology and Visual Sciences, Moran Eye Center, University of Utah, Salt Lake City, UT, 84132, USA
| | - H Kirk Hammond
- Division of Cardiovascular Medicine, Department of Medicine, University of California, San Diego, VA San Diego Healthcare System, San Diego, CA, 92161, USA
| | - Albert H Park
- Division of Otolaryngology, Department of Surgery, University of Utah, Salt Lake City, UT, 84132, USA
| | - Jun Yang
- Department of Ophthalmology and Visual Sciences, Moran Eye Center, University of Utah, Salt Lake City, UT, 84132, USA.
- Department of Neurobiology, University of Utah, Salt Lake City, UT, 84132, USA.
- Division of Otolaryngology, Department of Surgery, University of Utah, Salt Lake City, UT, 84132, USA.
| |
Collapse
|
9
|
Jeruzalska E, Mazur AJ. The Role of non-muscle actin paralogs in cell cycle progression and proliferation. Eur J Cell Biol 2023; 102:151315. [PMID: 37099935 DOI: 10.1016/j.ejcb.2023.151315] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 04/14/2023] [Accepted: 04/17/2023] [Indexed: 04/28/2023] Open
Abstract
Uncontrolled cell proliferation leads to several pathologies, including cancer. Thus, this process must be tightly regulated. The cell cycle accounts for cell proliferation, and its progression is coordinated with changes in cell shape, for which cytoskeleton reorganization is responsible. Rearrangement of the cytoskeleton allows for its participation in the precise division of genetic material and cytokinesis. One of the main cytoskeletal components is filamentous actin-based structures. Mammalian cells have at least six actin paralogs, four of which are muscle-specific, while two, named β- and γ-actin, are abundantly present in all types of cells. This review summarizes the findings that establish the role of non-muscle actin paralogs in regulating cell cycle progression and proliferation. We discuss studies showing that the level of a given non-muscle actin paralog in a cell influences the cell's ability to progress through the cell cycle and, thus, proliferation. Moreover, we elaborate on the non-muscle actins' role in regulating gene transcription, interactions of actin paralogs with proteins involved in controlling cell proliferation, and the contribution of non-muscle actins to different structures in a dividing cell. The data cited in this review show that non-muscle actins regulate the cell cycle and proliferation through varying mechanisms. We point to the need for further studies addressing these mechanisms.
Collapse
Affiliation(s)
- Estera Jeruzalska
- Department of Cell Pathology, Faculty of Biotechnology, University of Wroclaw, Poland
| | - Antonina J Mazur
- Department of Cell Pathology, Faculty of Biotechnology, University of Wroclaw, Poland.
| |
Collapse
|
10
|
Castillo-Lopez E, Pacífico C, Sener-Aydemir A, Hummel K, Nöbauer K, Ricci S, Rivera-Chacon R, Reisinger N, Razzazi-Fazeli E, Zebeli Q, Kreuzer-Redmer S. Diet and phytogenic supplementation substantially modulate the salivary proteome in dairy cows. J Proteomics 2023; 273:104795. [PMID: 36535624 DOI: 10.1016/j.jprot.2022.104795] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 11/30/2022] [Accepted: 12/11/2022] [Indexed: 12/23/2022]
Abstract
Phytogenic compounds may influence salivation or salivary properties. However, their effects on the bovine salivary proteome have not been evaluated. We investigated changes in the bovine salivary proteome due to transition from forage to high-concentrate diet, with and without supplementation with a phytogenic feed additive. Eight non-lactating cows were fed forage, then transitioned to a 65% concentrate diet (DM basis) over a week. Cows were control (n = 4, CON) or supplemented with a phytogenic feed additive (n = 4, PHY). Proteomic analysis was conducted using liquid chromatography coupled with mass spectrometry. We identified 1233 proteins; 878 were bovine proteins, 189 corresponded to bacteria, and 166 were plant proteins. Between forage and high-concentrate, 139 proteins were differentially abundant (P < 0.05), with 48 proteins having a log2FC difference > |2|. The salivary proteome reflected shifts in processes involving nutrient utilization, body tissue accretion, and immune response. Between PHY and CON, 195 proteins were differently abundant (P < 0.05), with 37 having a log2FC difference > |2|; 86 proteins were increased by PHY, including proteins involved in smell recognition. Many differentially abundant proteins correlated (r > |0.70|) with salivary bicarbonate, total mucins or pH. Results provide novel insights into the bovine salivary proteome using a non-invasive approach, and the association of specific proteins with major salivary properties influencing rumen homeostasis. SIGNIFICANCE: Phytogenic compounds may stimulate salivation due to their olfactory properties, but their effects on the salivary proteome have not been investigated. We investigated the effect of high-concentrate diets and supplementation with a phytogenic additive on the salivary proteome of cows. We show that analysis of cows' saliva can be a non-invasive approach to detect effects occurring not only in the gut, but also systemically including indications for gut health and immune response. Thus, results provide unique insights into the bovine salivary proteome, and will have a crucial contribution to further understand animal response in terms of nutrient utilization and immune activity due to the change from forage to a high-energy diet. Additionally, our findings reveal changes due to supplementation with a phytogenic feed additive with regard to health and olfactory stimulation. Furthermore, findings suggest an association between salivary proteins and other components like bicarbonate content.
Collapse
Affiliation(s)
- Ezequias Castillo-Lopez
- University of Veterinary Medicine Vienna, Institute of Animal Nutrition and Functional Plant Compounds, Christian Doppler Laboratory for Innovative Gut Health Concepts of Livestock, Vienna, Austria.
| | - Cátia Pacífico
- University of Veterinary Medicine Vienna, Institute of Animal Nutrition and Functional Plant Compounds, Christian Doppler Laboratory for Innovative Gut Health Concepts of Livestock, Vienna, Austria
| | - Arife Sener-Aydemir
- University of Veterinary Medicine Vienna, Institute of Animal Nutrition and Functional Plant Compounds, Christian Doppler Laboratory for Innovative Gut Health Concepts of Livestock, Vienna, Austria
| | - Karin Hummel
- University of Veterinary Medicine Vienna, VetCore Facility (Proteomics), Vienna, Austria
| | - Katharina Nöbauer
- University of Veterinary Medicine Vienna, VetCore Facility (Proteomics), Vienna, Austria
| | - Sara Ricci
- University of Veterinary Medicine Vienna, Institute of Animal Nutrition and Functional Plant Compounds, Christian Doppler Laboratory for Innovative Gut Health Concepts of Livestock, Vienna, Austria
| | - Raul Rivera-Chacon
- University of Veterinary Medicine Vienna, Institute of Animal Nutrition and Functional Plant Compounds, Christian Doppler Laboratory for Innovative Gut Health Concepts of Livestock, Vienna, Austria
| | | | - Ebrahim Razzazi-Fazeli
- University of Veterinary Medicine Vienna, VetCore Facility (Proteomics), Vienna, Austria
| | - Qendrim Zebeli
- University of Veterinary Medicine Vienna, Institute of Animal Nutrition and Functional Plant Compounds, Christian Doppler Laboratory for Innovative Gut Health Concepts of Livestock, Vienna, Austria
| | - Susanne Kreuzer-Redmer
- University of Veterinary Medicine Vienna, Institute of Animal Nutrition and Functional Plant Compounds, Nutrigenomics Unit, Vienna, Austria.
| |
Collapse
|
11
|
Sundby LJ, Southern WM, Hawbaker KM, Trujillo JM, Perrin BJ, Ervasti JM. Nucleotide- and Protein-Dependent Functions of Actg1. Mol Biol Cell 2022; 33:ar77. [PMID: 35594181 PMCID: PMC9582642 DOI: 10.1091/mbc.e22-02-0054] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 05/10/2022] [Accepted: 05/11/2022] [Indexed: 11/11/2022] Open
Abstract
Cytoplasmic β- and γ-actin proteins are 99% identical but support unique organismal functions. The cytoplasmic actin nucleotide sequences Actb and Actg1, respectively, are more divergent but still 89% similar. Actb-/- mice are embryonic lethal and Actb-/- cells fail to proliferate, but editing the Actb gene to express γ-actin (Actbc-g) resulted in none of the overt phenotypes of the knockout revealing protein-independent functions for Actb. To determine if Actg1 has a protein-independent function, we crossed Actbc-g and Actg1-/- mice to generate the bG/0 line, where the only cytoplasmic actin expressed is γ-actin from Actbc-g. The bG/0 mice were viable but showed a survival defect despite expressing γ-actin protein at levels no different from bG/gG with normal survival. A unique myopathy phenotype was also observed in bG/0 mice. We conclude that impaired survival and myopathy in bG/0 mice are due to loss of Actg1 nucleotide-dependent function(s). On the other hand, the bG/0 genotype rescued functions impaired by Actg1-/-, including cell proliferation and auditory function, suggesting a role for γ-actin protein in both fibroblasts and hearing. Together, these results identify nucleotide-dependent functions for Actg1 while implicating γ-actin protein in more cell-/tissue-specific functions.
Collapse
Affiliation(s)
- Lauren J. Sundby
- Program in Molecular, Cellular, Developmental Biology, and Genetics, and
| | - William M. Southern
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN 55455
| | - Katelin M. Hawbaker
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis, IN 46022
| | - Jesús M. Trujillo
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN 55455
| | - Benjamin J. Perrin
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis, IN 46022
| | - James M. Ervasti
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN 55455
| |
Collapse
|
12
|
Zhang Z, Zhang W, Blakes R, Sundby LJ, Shi Z, Rockey DC, Ervasti JM, Nam YJ. Fibroblast fate determination during cardiac reprogramming by remodeling of actin filaments. Stem Cell Reports 2022; 17:1604-1619. [PMID: 35688153 PMCID: PMC9287671 DOI: 10.1016/j.stemcr.2022.05.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 05/12/2022] [Accepted: 05/13/2022] [Indexed: 11/23/2022] Open
Abstract
Fibroblasts can be reprogrammed into induced cardiomyocyte-like cells (iCMs) by forced expression of cardiogenic transcription factors. However, it remains unknown how fibroblasts adopt a cardiomyocyte (CM) fate during their spontaneous ongoing transdifferentiation toward myofibroblasts (MFs). By tracing fibroblast lineages following cardiac reprogramming in vitro, we found that most mature iCMs are derived directly from fibroblasts without transition through the MF state. This direct conversion is attributable to mutually exclusive induction of cardiac sarcomeres and MF cytoskeletal structures in the cytoplasm of fibroblasts during reprogramming. For direct fate switch from fibroblasts to iCMs, significant remodeling of actin isoforms occurs in fibroblasts, including induction of α-cardiac actin and decrease of the actin isoforms predominant in MFs. Accordingly, genetic or pharmacological ablation of MF-enriched actin isoforms significantly enhances cardiac reprogramming. Our results demonstrate that remodeling of actin isoforms is required for fibroblast to CM fate conversion by cardiac reprogramming.
Collapse
Affiliation(s)
- Zhentao Zhang
- Department of Medicine, Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN, USA; Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, USA; Vanderbilt Center for Stem Cell Biology, Vanderbilt University, Nashville, TN, USA
| | - Wenhui Zhang
- Department of Medicine, Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN, USA; Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, USA; Vanderbilt Center for Stem Cell Biology, Vanderbilt University, Nashville, TN, USA
| | - Robert Blakes
- Department of Medicine, Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN, USA; Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, USA; Vanderbilt Center for Stem Cell Biology, Vanderbilt University, Nashville, TN, USA
| | - Lauren J Sundby
- Program in Molecular, Cellular, Developmental Biology, and Genetics, University of Minnesota, Minneapolis, MN, USA
| | - Zengdun Shi
- Department of Internal Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - Don C Rockey
- Department of Internal Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - James M Ervasti
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, USA; Program in Molecular, Cellular, Developmental Biology, and Genetics, University of Minnesota, Minneapolis, MN, USA
| | - Young-Jae Nam
- Department of Medicine, Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN, USA; Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, USA; Vanderbilt Center for Stem Cell Biology, Vanderbilt University, Nashville, TN, USA.
| |
Collapse
|
13
|
Knudsen JR, Madsen AB, Li Z, Andersen NR, Schjerling P, Jensen TE. Gene deletion of γ-actin impairs insulin-stimulated skeletal muscle glucose uptake in growing mice but not in mature adult mice. Physiol Rep 2022; 10:e15183. [PMID: 35224890 PMCID: PMC8882697 DOI: 10.14814/phy2.15183] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Revised: 12/16/2021] [Accepted: 12/17/2021] [Indexed: 04/14/2023] Open
Abstract
The cortical cytoskeleton, consisting of the cytoplasmic actin isoforms β and/or γ-actin, has been implicated in insulin-stimulated GLUT4 translocation and glucose uptake in muscle and adipose cell culture. Furthermore, transgenic inhibition of multiple actin-regulating proteins in muscle inhibits insulin-stimulated muscle glucose uptake. The current study tested if γ-actin was required for insulin-stimulated glucose uptake in mouse skeletal muscle. Based on our previously reported age-dependent phenotype in muscle-specific β-actin gene deletion (-/- ) mice, we included cohorts of growing 8-14 weeks old and mature 18-32 weeks old muscle-specific γ-actin-/- mice or wild-type littermates. In growing mice, insulin significantly increased the glucose uptake in slow-twitch oxidative soleus and fast-twitch glycolytic EDL muscles from wild-type mice, but not γ-actin-/- . In relative values, the maximal insulin-stimulated glucose uptake was reduced by ~50% in soleus and by ~70% in EDL muscles from growing γ-actin-/- mice compared to growing wild-type mice. In contrast, the insulin-stimulated glucose uptake responses in mature adult γ-actin-/- soleus and EDL muscles were indistinguishable from the responses in wild-type muscles. Mature adult insulin-stimulated phosphorylations on Akt, p70S6K, and ULK1 were not significantly affected by genotype. Hence, insulin-stimulated muscle glucose uptake shows an age-dependent impairment in young growing but not in fully grown γ-actin-/- mice, bearing phenotypic resemblance to β-actin-/- mice. Overall, γ-actin does not appear required for insulin-stimulated muscle glucose uptake in adulthood. Furthermore, our data emphasize the need to consider the rapid growth of young mice as a potential confounder in transgenic mouse phenotyping studies.
Collapse
Affiliation(s)
- Jonas R. Knudsen
- Section for Molecular PhysiologyDepartment of Nutrition, Exercise and SportsUniversity of CopenhagenCopenhagenDenmark
| | - Agnete B. Madsen
- Section for Molecular PhysiologyDepartment of Nutrition, Exercise and SportsUniversity of CopenhagenCopenhagenDenmark
| | - Zhencheng Li
- Section for Molecular PhysiologyDepartment of Nutrition, Exercise and SportsUniversity of CopenhagenCopenhagenDenmark
| | - Nicoline R. Andersen
- Section for Molecular PhysiologyDepartment of Nutrition, Exercise and SportsUniversity of CopenhagenCopenhagenDenmark
| | - Peter Schjerling
- Department of Orthopedic Surgery MInstitute of Sports Medicine CopenhagenBispebjerg HospitalCopenhagenDenmark
| | - Thomas E. Jensen
- Section for Molecular PhysiologyDepartment of Nutrition, Exercise and SportsUniversity of CopenhagenCopenhagenDenmark
| |
Collapse
|
14
|
DFNA20/26 and Other ACTG1-Associated Phenotypes: A Case Report and Review of the Literature. Audiol Res 2021; 11:582-593. [PMID: 34698053 PMCID: PMC8544197 DOI: 10.3390/audiolres11040052] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 10/10/2021] [Accepted: 10/13/2021] [Indexed: 02/08/2023] Open
Abstract
Since the early 2000s, an ever-increasing subset of missense pathogenic variants in the ACTG1 gene has been associated with an autosomal-dominant, progressive, typically post-lingual non-syndromic hearing loss (NSHL) condition designed as DFNA20/26. ACTG1 gene encodes gamma actin, the predominant actin protein in the cytoskeleton of auditory hair cells; its normal expression and function are essential for the stereocilia maintenance. Different gain-of-function pathogenic variants of ACTG1 have been associated with two major phenotypes: DFNA20/26 and Baraitser-Winter syndrome, a multiple congenital anomaly disorder. Here, we report a novel ACTG1 variant [c.625G>A (p. Val209Met)] in an adult patient with moderate-severe NSHL characterized by a downsloping audiogram. The patient, who had a clinical history of slowly progressive NSHL and tinnitus, was referred to our laboratory for the analysis of a large panel of NSHL-associated genes by next generation sequencing. An extensive review of previously reported ACTG1 variants and their associated phenotypes was also performed.
Collapse
|
15
|
Garcia-Pelagio KP, Bloch RJ. Biomechanical Properties of the Sarcolemma and Costameres of Skeletal Muscle Lacking Desmin. Front Physiol 2021; 12:706806. [PMID: 34489727 PMCID: PMC8416993 DOI: 10.3389/fphys.2021.706806] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Accepted: 07/13/2021] [Indexed: 01/23/2023] Open
Abstract
Intermediate filaments (IFs), composed primarily by desmin and keratins, link the myofibrils to each other, to intracellular organelles, and to the sarcolemma. There they may play an important role in transfer of contractile force from the Z-disks and M-lines of neighboring myofibrils to costameres at the membrane, across the membrane to the extracellular matrix, and ultimately to the tendon (“lateral force transmission”). We measured the elasticity of the sarcolemma and the connections it makes at costameres with the underlying contractile apparatus of individual fast twitch muscle fibers of desmin-null mice. By positioning a suction pipet to the surface of the sarcolemma and applying increasing pressure, we determined the pressure at which the sarcolemma separated from nearby sarcomeres, Pseparation, and the pressure at which the isolated sarcolemma burst, Pbursting. We also examined the time required for the intact sarcolemma-costamere-sarcomere complex to reach equilibrium at lower pressures. All measurements showed the desmin-null fibers to have slower equilibrium times and lower Pseparation and Pbursting than controls, suggesting that the sarcolemma and its costameric links to nearby contractile structures were weaker in the absence of desmin. Comparisons to earlier values determined for muscles lacking dystrophin or synemin suggest that the desmin-null phenotype is more stable than the former and less stable than the latter. Our results are consistent with the moderate myopathy seen in desmin-null muscles and support the idea that desmin contributes significantly to sarcolemmal stability and lateral force transmission.
Collapse
Affiliation(s)
- Karla P Garcia-Pelagio
- Departamento de Fisica, Facultad de Ciencias, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Robert J Bloch
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, United States
| |
Collapse
|
16
|
Lechuga S, Naydenov NG, Feygin A, Cruise M, Ervasti JM, Ivanov AI. Loss of β-Cytoplasmic Actin in the Intestinal Epithelium Increases Gut Barrier Permeability in vivo and Exaggerates the Severity of Experimental Colitis. Front Cell Dev Biol 2020; 8:588836. [PMID: 33195251 PMCID: PMC7644907 DOI: 10.3389/fcell.2020.588836] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Accepted: 10/06/2020] [Indexed: 12/21/2022] Open
Abstract
Intestinal epithelial barrier is critical for the maintenance of normal gut homeostasis and disruption of this barrier may trigger or exaggerate mucosal inflammation. The actin cytoskeleton is a key regulator of barrier structure and function, controlling the assembly and permeability of epithelial adherens and tight junctions. Epithelial cells express two actin isoforms: a β-cytoplasmic actin and γ-cytoplasmic actin. Our previous in vitro studies demonstrated that these actin isoforms play distinctive roles in establishing the intestinal epithelial barrier, by controlling the organization of different junctional complexes. It remains unknown, whether β-actin and γ-actin have unique or redundant functions in regulating the gut barrier in vivo. To address this question, we selectively knocked out β-actin expression in mouse intestinal epithelium. Mice with intestinal epithelial knockout of β-actin do not display gastrointestinal abnormalities or gross alterations of colonic mucosal architecture. This could be due to compensatory upregulation of γ-actin expression. Despite such compensation, β-actin knockout mice demonstrate increased intestinal permeability. Furthermore, these animals show more severe clinical symptoms during dextran sodium sulfate induced colitis, compared to control littermates. Such exaggerated colitis is associated with the higher expression of inflammatory cytokines, increased macrophage infiltration in the gut, and accelerated mucosal cell death. Consistently, intestinal organoids generated from β-actin knockout mice are more sensitive to tumor necrosis factor induced cell death, ex vivo. Overall, our data suggests that β-actin functions as an essential regulator of gut barrier integrity in vivo, and plays a tissue protective role during mucosal injury and inflammation.
Collapse
Affiliation(s)
- Susana Lechuga
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, United States
| | - Nayden G Naydenov
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, United States
| | - Alex Feygin
- School of Nursing, Virginia Commonwealth University School of Nursing, Richmond, VA, United States
| | - Michael Cruise
- Department of Pathology, Cleveland Clinic Foundation, Cleveland, OH, United States
| | - James M Ervasti
- Department of Biochemistry and Molecular Biology, University of Minnesota Medical School, Minneapolis, MN, United States
| | - Andrei I Ivanov
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, United States
| |
Collapse
|
17
|
Nelson DM, Fasbender EK, Jakubiak MC, Lindsay A, Lowe DA, Ervasti JM. Rapid, redox-mediated mechanical susceptibility of the cortical microtubule lattice in skeletal muscle. Redox Biol 2020; 37:101730. [PMID: 33002761 PMCID: PMC7527753 DOI: 10.1016/j.redox.2020.101730] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 09/12/2020] [Accepted: 09/12/2020] [Indexed: 01/25/2023] Open
Abstract
The highly ordered cortical microtubule lattice of skeletal muscle is disorganized in dystrophin-deficient mdx mice. Implicated mechanisms include loss of dystrophin binding, altered α-tubulin posttranslational modification, expression of a β-tubulin involved in regeneration, and reactive oxygen species (ROS). Here we show that the transverse microtubules in mdx muscle expressing miniaturized dystrophins are rapidly lost after eccentric contraction. Analysis of mdx lines expressing different dystrophin constructs demonstrate that spectrin-like repeats R4-15 and R20-23 were required for mechanically stable microtubules. Microtubule loss was prevented by the non-specific antioxidant N-acetylcysteine while inhibition of NADPH oxidase 2 had only a partial effect, suggesting that ROS from multiple sources mediate the rapid loss of transverse microtubules after eccentric contraction. Finally, ablation of α-dystrobrevin, β- or γ-cytoplasmic actin phenocopied the transverse microtubule instability of miniaturized dystrophins. Our data demonstrate that multiple dystrophin domains, α-dystrobrevin and cytoplasmic actins are necessary for mechanically stable microtubules.
Collapse
Affiliation(s)
- D'anna M Nelson
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, USA
| | - Elizabeth K Fasbender
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, USA; College of Biological Sciences, University of Minnesota, Minneapolis, MN, USA
| | - Margurite C Jakubiak
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, USA; College of Biological Sciences, University of Minnesota, Minneapolis, MN, USA
| | - Angus Lindsay
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, USA; Department of Rehabilitation Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Dawn A Lowe
- Department of Rehabilitation Medicine, University of Minnesota, Minneapolis, MN, USA
| | - James M Ervasti
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
18
|
Alvarez-Romero J, Voisin S, Eynon N, Hiam D. Mapping Robust Genetic Variants Associated with Exercise Responses. Int J Sports Med 2020; 42:3-18. [PMID: 32693428 DOI: 10.1055/a-1198-5496] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
This review summarised robust and consistent genetic variants associated with aerobic-related and resistance-related phenotypes. In total we highlight 12 SNPs and 7 SNPs that are robustly associated with variance in aerobic-related and resistance-related phenotypes respectively. To date, there is very little literature ascribed to understanding the interplay between genes and environmental factors and the development of physiological traits. We discuss future directions, including large-scale exercise studies to elucidate the functional relevance of the discovered genomic markers. This approach will allow more rigour and reproducible research in the field of exercise genomics.
Collapse
Affiliation(s)
| | - Sarah Voisin
- Institute for Health and Sport, Victoria University, Melbourne, Australia
| | - Nir Eynon
- Institute for Health and Sport, Victoria University, Melbourne, Australia.,MCRI, Murdoch Childrens Research Institute, Parkville, Australia
| | - Danielle Hiam
- Institute for Health and Sport, Victoria University, Melbourne, Australia
| |
Collapse
|
19
|
Ferry A, Messéant J, Parlakian A, Lemaitre M, Roy P, Delacroix C, Lilienbaum A, Hovhannisyan Y, Furling D, Klein A, Li Z, Agbulut O. Desmin prevents muscle wasting, exaggerated weakness and fragility, and fatigue in dystrophic mdx mouse. J Physiol 2020; 598:3667-3689. [PMID: 32515007 DOI: 10.1113/jp279282] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Accepted: 06/05/2020] [Indexed: 01/21/2023] Open
Abstract
KEY POINTS Desmin, similar to dystrophin, is associated with costameric structures bridging sarcomeres to the extracellular matrix. Deletion of the desmin gene in mdx mice [double knockout (DKO) mice] induces marked muscle weakness and fatigue resistance compared to mdx mice. Muscle fragility (higher susceptibility to contraction-induced injury) was also aggravated in DKO mice compared to mdx mice. By contrast to mdx mice, the DKO mice did not undergo muscle hypertrophy. Desmin cDNA transfer with adeno-associated virus in newborn mdx mice reduced muscle weakness. Overall, desmin plays important and beneficial roles in muscle wasting, performance and fragility in dystrophic muscle. ABSTRACT Duchenne muscular dystrophy (DMD) is a severe neuromuscular disease caused by dystrophin deficiency. Desmin, similar to dystrophin, is associated with costameric structures bridging sarcomeres to the extracellular matrix that contributes to muscle function. In the present study, we attempted to provide further insight into the roles of desmin, for which the expression is increased in the muscle from the mouse mdx DMD model. We show that a deletion of the desmin gene (Des) in mdx mice [double knockout (DKO) mice, mdx:desmin-/-] induces a marked muscle weakness; namely, a reduced absolute maximal force production and increased fatigue compared to that in mdx mice. Fragility (i.e. higher susceptibility to contraction-induced injury) was also aggravated in DKO mice compared to mdx mice, despite the promotion of supposedly less fragile muscle fibres in DKO mice, and this worsening of fragility was related to a decreased muscle excitability. Moreover, in contrast to mdx mice, the DKO mice did not undergo muscle hypertrophy, as indicated by smaller and fewer fibres, with a reduced percentage of centronucleated fibres, potentially explaining the severe muscle weakness. Notably, Desmin cDNA transfer with adeno-associated virus in newborn mdx mice improved specific maximal force normalized to muscle weight. Overall, desmin plays important and beneficial roles in muscle wasting, performance and fragility in dystrophic mdx mice, which differ, at least in part, from those observed in healthy muscle.
Collapse
Affiliation(s)
- Arnaud Ferry
- Sorbonne Université, Centre de recherche en myologie, INSERM U974, Institut de Myologie, Paris, France.,Université de Paris, Institut des Sciences du Sport Santé de Paris, UFRSTAPS, Paris, France
| | - Julien Messéant
- Sorbonne Université, Centre de recherche en myologie, INSERM U974, Institut de Myologie, Paris, France
| | - Ara Parlakian
- Sorbonne Université, Institut de Biologie Paris-Seine (IBPS), UMR CNRS 8256, INSERM ERL U1164, Biological Adaptation and Ageing, Paris, France
| | - Mégane Lemaitre
- Sorbonne Université, Centre de recherche en myologie, INSERM U974, Institut de Myologie, Paris, France
| | - Pauline Roy
- Sorbonne Université, Centre de recherche en myologie, INSERM U974, Institut de Myologie, Paris, France
| | - Clément Delacroix
- Sorbonne Université, Centre de recherche en myologie, INSERM U974, Institut de Myologie, Paris, France
| | - Alain Lilienbaum
- Université de Paris, Unité de Biologie Fonctionnelle et Adaptative, CNRS UMR 8251, Paris, France
| | - Yeranuhi Hovhannisyan
- Sorbonne Université, Institut de Biologie Paris-Seine (IBPS), UMR CNRS 8256, INSERM ERL U1164, Biological Adaptation and Ageing, Paris, France
| | - Denis Furling
- Sorbonne Université, Centre de recherche en myologie, INSERM U974, Institut de Myologie, Paris, France
| | - Arnaud Klein
- Sorbonne Université, Centre de recherche en myologie, INSERM U974, Institut de Myologie, Paris, France
| | - Zhenlin Li
- Sorbonne Université, Institut de Biologie Paris-Seine (IBPS), UMR CNRS 8256, INSERM ERL U1164, Biological Adaptation and Ageing, Paris, France
| | - Onnik Agbulut
- Sorbonne Université, Institut de Biologie Paris-Seine (IBPS), UMR CNRS 8256, INSERM ERL U1164, Biological Adaptation and Ageing, Paris, France
| |
Collapse
|
20
|
Báez-Matus X, Figueroa-Cares C, Gónzalez-Jamett AM, Almarza-Salazar H, Arriagada C, Maldifassi MC, Guerra MJ, Mouly V, Bigot A, Caviedes P, Cárdenas AM. Defects in G-Actin Incorporation into Filaments in Myoblasts Derived from Dysferlinopathy Patients Are Restored by Dysferlin C2 Domains. Int J Mol Sci 2019; 21:ijms21010037. [PMID: 31861684 PMCID: PMC6981584 DOI: 10.3390/ijms21010037] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Revised: 12/13/2019] [Accepted: 12/16/2019] [Indexed: 12/23/2022] Open
Abstract
Dysferlin is a transmembrane C-2 domain-containing protein involved in vesicle trafficking and membrane remodeling in skeletal muscle cells. However, the mechanism by which dysferlin regulates these cellular processes remains unclear. Since actin dynamics is critical for vesicle trafficking and membrane remodeling, we studied the role of dysferlin in Ca2+-induced G-actin incorporation into filaments in four different immortalized myoblast cell lines (DYSF2, DYSF3, AB320, and ER) derived from patients harboring mutations in the dysferlin gene. As compared with immortalized myoblasts obtained from a control subject, dysferlin expression and G-actin incorporation were significantly decreased in myoblasts from dysferlinopathy patients. Stable knockdown of dysferlin with specific shRNA in control myoblasts also significantly reduced G-actin incorporation. The impaired G-actin incorporation was restored by the expression of full-length dysferlin as well as dysferlin N-terminal or C-terminal regions, both of which contain three C2 domains. DYSF3 myoblasts also exhibited altered distribution of annexin A2, a dysferlin partner involved in actin remodeling. However, dysferlin N-terminal and C-terminal regions appeared to not fully restore such annexin A2 mislocation. Then, our results suggest that dysferlin regulates actin remodeling by a mechanism that does to not involve annexin A2.
Collapse
Affiliation(s)
- Ximena Báez-Matus
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile; (X.B.-M.); (C.F.-C.); (A.M.G.-J.); (M.C.M.); (M.J.G.)
| | - Cindel Figueroa-Cares
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile; (X.B.-M.); (C.F.-C.); (A.M.G.-J.); (M.C.M.); (M.J.G.)
| | - Arlek M. Gónzalez-Jamett
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile; (X.B.-M.); (C.F.-C.); (A.M.G.-J.); (M.C.M.); (M.J.G.)
| | - Hugo Almarza-Salazar
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile; (X.B.-M.); (C.F.-C.); (A.M.G.-J.); (M.C.M.); (M.J.G.)
| | - Christian Arriagada
- Departamento de Anatomía y Medicina Legal, Facultad de Medicina, Universidad de Chile, Santiago 8389100, Chile
| | - María Constanza Maldifassi
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile; (X.B.-M.); (C.F.-C.); (A.M.G.-J.); (M.C.M.); (M.J.G.)
| | - María José Guerra
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile; (X.B.-M.); (C.F.-C.); (A.M.G.-J.); (M.C.M.); (M.J.G.)
| | - Vincent Mouly
- Sorbonne Université, Inserm, Institut de Myologie, UMRS 974, Center for Research in Myology, 75013 Paris, France; (V.M.); (A.B.)
| | - Anne Bigot
- Sorbonne Université, Inserm, Institut de Myologie, UMRS 974, Center for Research in Myology, 75013 Paris, France; (V.M.); (A.B.)
| | - Pablo Caviedes
- Programa de Farmacología Molecular y Clínica, ICBM, Facultad de Medicina, Universidad de Chile, Santiago 8389100, Chile;
- Centro de Biotecnología y Bioingeniería (CeBiB), Departamento de Ingeniería Química, Biotecnología y Materiales, Facultad de Ciencias Físicas y Matemáticas, Universidad de Chile, Santiago 8370456, Chile
| | - Ana M. Cárdenas
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile; (X.B.-M.); (C.F.-C.); (A.M.G.-J.); (M.C.M.); (M.J.G.)
- Correspondence: ; Tel.: +56-322-508-052
| |
Collapse
|
21
|
A new evolutionary model for the vertebrate actin family including two novel groups. Mol Phylogenet Evol 2019; 141:106632. [DOI: 10.1016/j.ympev.2019.106632] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 09/19/2019] [Accepted: 09/23/2019] [Indexed: 02/06/2023]
|
22
|
How the central domain of dystrophin acts to bridge F-actin to sarcolemmal lipids. J Struct Biol 2019; 209:107411. [PMID: 31689503 DOI: 10.1016/j.jsb.2019.107411] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 10/07/2019] [Accepted: 10/29/2019] [Indexed: 01/08/2023]
Abstract
Dystrophin is a large intracellular protein that prevents sarcolemmal ruptures by providing a mechanical link between the intracellular actin cytoskeleton and the transmembrane dystroglycan complex. Dystrophin deficiency leads to the severe muscle wasting disease Duchenne Muscular Dystrophy and the milder allelic variant, Becker Muscular Dystrophy (DMD and BMD). Previous work has shown that concomitant interaction of the actin binding domain 2 (ABD2) comprising spectrin like repeats 11 to 15 (R11-15) of the central domain of dystrophin, with both actin and membrane lipids, can greatly increase membrane stiffness. Based on a combination of SAXS and SANS measurements, mass spectrometry analysis of cross-linked complexes and interactive low-resolution simulations, we explored in vitro the molecular properties of dystrophin that allow the formation of ABD2-F-actin and ABD2-membrane model complexes. In dystrophin we identified two subdomains interacting with F-actin, one located in R11 and a neighbouring region in R12 and another one in R15, while a single lipid binding domain was identified at the C-terminal end of R12. Relative orientations of the dystrophin central domain with F-actin and a membrane model were obtained from docking simulation under experimental constraints. SAXS-based models were then built for an extended central subdomain from R4 to R19, including ABD2. Overall results are compatible with a potential F-actin/dystrophin/membrane lipids ternary complex. Our description of this selected part of the dystrophin associated complex bridging muscle cell membrane and cytoskeleton opens the way to a better understanding of how cell muscle scaffolding is maintained through this essential protein.
Collapse
|
23
|
Ross JA, Levy Y, Ripolone M, Kolb JS, Turmaine M, Holt M, Lindqvist J, Claeys KG, Weis J, Monforte M, Tasca G, Moggio M, Figeac N, Zammit PS, Jungbluth H, Fiorillo C, Vissing J, Witting N, Granzier H, Zanoteli E, Hardeman EC, Wallgren-Pettersson C, Ochala J. Impairments in contractility and cytoskeletal organisation cause nuclear defects in nemaline myopathy. Acta Neuropathol 2019; 138:477-495. [PMID: 31218456 PMCID: PMC6689292 DOI: 10.1007/s00401-019-02034-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 05/28/2019] [Accepted: 06/05/2019] [Indexed: 02/07/2023]
Abstract
Nemaline myopathy (NM) is a skeletal muscle disorder caused by mutations in genes that are generally involved in muscle contraction, in particular those related to the structure and/or regulation of the thin filament. Many pathogenic aspects of this disease remain largely unclear. Here, we report novel pathological defects in skeletal muscle fibres of mouse models and patients with NM: irregular spacing and morphology of nuclei; disrupted nuclear envelope; altered chromatin arrangement; and disorganisation of the cortical cytoskeleton. Impairments in contractility are the primary cause of these nuclear defects. We also establish the role of microtubule organisation in determining nuclear morphology, a phenomenon which is likely to contribute to nuclear alterations in this disease. Our results overlap with findings in diseases caused directly by mutations in nuclear envelope or cytoskeletal proteins. Given the important role of nuclear shape and envelope in regulating gene expression, and the cytoskeleton in maintaining muscle fibre integrity, our findings are likely to explain some of the hallmarks of NM, including contractile filament disarray, altered mechanical properties and broad transcriptional alterations.
Collapse
|
24
|
Lindsay A, Southern WM, McCourt PM, Larson AA, Hodges JS, Lowe DA, Ervasti JM. Variable cytoplasmic actin expression impacts the sensitivity of different dystrophin-deficient mdx skeletal muscles to eccentric contraction. FEBS J 2019; 286:2562-2576. [PMID: 30942954 PMCID: PMC6613979 DOI: 10.1111/febs.14831] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 01/24/2019] [Accepted: 04/01/2019] [Indexed: 11/29/2022]
Abstract
Eccentric contractions (ECCs) induce force loss in several skeletal muscles of dystrophin-deficient mice (mdx), with the exception of the soleus (Sol). The eccentric force : isometric force (ECC : ISO), expression level of utrophin, fiber type distribution, and sarcoendoplasmic reticulum calcium ATPase expression are factors that differ between muscles and may contribute to the sensitivity of mdx skeletal muscle to ECC. Here, we confirm that the Sol of mdx mice loses only 13% force compared to 87% in the extensor digitorum longus (EDL) following 10 ECC of isolated muscles. The Sol has a greater proportion of fibers expressing Type I myosin heavy chain (MHC) and expresses 2.3-fold more utrophin compared to the EDL. To examine the effect of ECC : ISO, we show that the mdx Sol is insensitive to ECC at ECC : ISO up to 230 ± 15%. We show that the peroneus longus (PL) muscle presents with similar ECC : ISO compared to the EDL, intermediate force loss (68%) following 10 ECC, and intermediate fiber type distribution and utrophin expression relative to EDL and Sol. The combined absence of utrophin and dystrophin in mdx/utrophin-/- mice rendered the Sol only partially susceptible to ECC and exacerbated force loss in the EDL and PL. Most interestingly, the expression levels of cytoplasmic β- and γ-actins correlate inversely with a given muscle's sensitivity to ECC; EDL < PL < Sol. Our data indicate that fiber type, utrophin, and cytoplasmic actin expression all contribute to the differential sensitivities of mdxEDL, PL, and Sol muscles to ECC.
Collapse
Affiliation(s)
- Angus Lindsay
- Division of Rehabilitation Science and Division of Physical Therapy, Department of Rehabilitation Medicine, University of Minnesota, Minneapolis, USA
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, USA
| | - William M. Southern
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, USA
| | - Preston M. McCourt
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, USA
| | - Alexie A. Larson
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, USA
| | - James S. Hodges
- Division of Biostatistics, University of Minnesota, Minneapolis, USA
| | - Dawn A. Lowe
- Division of Rehabilitation Science and Division of Physical Therapy, Department of Rehabilitation Medicine, University of Minnesota, Minneapolis, USA
| | - James M. Ervasti
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, USA
| |
Collapse
|
25
|
Kita AM, Swider ZT, Erofeev I, Halloran MC, Goryachev AB, Bement WM. Spindle-F-actin interactions in mitotic spindles in an intact vertebrate epithelium. Mol Biol Cell 2019; 30:1645-1654. [PMID: 31091161 PMCID: PMC6727749 DOI: 10.1091/mbc.e19-02-0126] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Mitotic spindles are well known to be assembled from and dependent on microtubules. In contrast, whether actin filaments (F-actin) are required for or are even present in mitotic spindles has long been controversial. Here we have developed improved methods for simultaneously preserving F-actin and microtubules in fixed samples and exploited them to demonstrate that F-actin is indeed associated with mitotic spindles in intact Xenopus laevis embryonic epithelia. We also find that there is an “F-actin cycle,” in which the distribution and organization of spindle F-actin changes over the course of the cell cycle. Live imaging using a probe for F-actin reveals that at least two pools of F-actin are associated with mitotic spindles: a relatively stable internal network of cables that moves in concert with and appears to be linked to spindles, and F-actin “fingers” that rapidly extend from the cell cortex toward the spindle and make transient contact with the spindle poles. We conclude that there is a robust endoplasmic F-actin network in normal vertebrate epithelial cells and that this network is also a component of mitotic spindles. More broadly, we conclude that there is far more internal F-actin in epithelial cells than is commonly believed.
Collapse
Affiliation(s)
- Angela M Kita
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI 53706.,Laboratory of Cell and Molecular Biology, University of Wisconsin-Madison, Madison, WI 53706
| | - Zachary T Swider
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI 53706.,Laboratory of Cell and Molecular Biology, University of Wisconsin-Madison, Madison, WI 53706
| | - Ivan Erofeev
- Centre for Synthetic and Systems Biology, University of Edinburgh, Edinburgh EH9 3JD, United Kingdom
| | - Mary C Halloran
- Department of Integrative Biology, University of Wisconsin-Madison, Madison, WI 53706.,Department of Neuroscience, University of Wisconsin-Madison, Madison, WI 53706
| | - Andrew B Goryachev
- Centre for Synthetic and Systems Biology, University of Edinburgh, Edinburgh EH9 3JD, United Kingdom
| | - William M Bement
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI 53706.,Laboratory of Cell and Molecular Biology, University of Wisconsin-Madison, Madison, WI 53706.,Department of Integrative Biology, University of Wisconsin-Madison, Madison, WI 53706
| |
Collapse
|
26
|
Abstract
PURPOSE OF REVIEW To summarize the evidence from recent studies on the shared genetics between bone and muscle in humans. RECENT FINDINGS Genome-wide association studies (GWAS) have successfully identified a multitude of loci influencing the variability of different bone or muscle parameters, with multiple loci overlapping between the traits. In addition, joint analyses of multiple correlated musculoskeletal traits (i.e., multivariate GWAS) have underscored several genes with possible pleiotropic effects on both bone and muscle including MEF2C and SREBF1. Notably, several of the proposed pleiotropic genes have been validated using human cells or animal models. It is clear that the study of pleiotropy may provide novel insights into disease pathophysiology potentially leading to the identification of new treatment strategies that simultaneously prevent or treat both osteoporosis and sarcopenia. However, the role of muscle factors (myokines) that stimulate bone metabolism, as well as osteokines that affect muscles, is in its earliest stage of understanding.
Collapse
Affiliation(s)
- Katerina Trajanoska
- Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Fernando Rivadeneira
- Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands.
| | - Douglas P Kiel
- Hebrew SeniorLife, Institute for Aging Research, Boston, MA, USA
- Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
- Broad Institute of Harvard and Massachusetts Institute of Technology, Boston, MA, USA
| | - David Karasik
- Hebrew SeniorLife, Institute for Aging Research, Boston, MA, USA.
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel.
| |
Collapse
|
27
|
Abstract
Dynamin 2 (DNM2) belongs to a family of large GTPases that are well known for mediating membrane fission by oligomerizing at the neck of membrane invaginations. Autosomal dominant mutations in the ubiquitously expressed DNM2 cause 2 discrete neuromuscular diseases: autosomal dominant centronuclear myopathy (ADCNM) and dominant intermediate Charcot-Marie-Tooth neuropathy (CMT). CNM and CMT mutations may affect DNM2 in distinct manners: CNM mutations may cause protein hyperactivity with elevated GTPase and fission activities, while CMT mutations could impair DNM2 lipid binding and activity. DNM2 is also a modifier of the X-linked and autosomal recessive forms of CNM, as DNM2 protein levels are upregulated in animal models and patient muscle samples. Strikingly, reducing DNM2 has been shown to revert muscle phenotypes in preclinical models of CNM. As DNM2 emerges as the key player in CNM pathogenesis, the role(s) of DNM2 in skeletal muscle remains unclear. This review aims to provide insights into potential pathomechanisms related to DNM2-CNM mutations, and discuss exciting outcomes of current and future therapeutic approaches targeting DNM2 hyperactivity.
Collapse
Affiliation(s)
- Mo Zhao
- Genetics and Genome Biology Program, Hospital for Sick Children, Toronto, ON, M5G 0A4, Canada
| | - Nika Maani
- Genetics and Genome Biology Program, Hospital for Sick Children, Toronto, ON, M5G 0A4, Canada
| | - James J Dowling
- Genetics and Genome Biology Program, Hospital for Sick Children, Toronto, ON, M5G 0A4, Canada.
- Division of Neurology, Hospital for Sick Children, Toronto, ON, M5G 1X8, Canada.
- Department of Pediatrics, University of Toronto, Toronto, ON, M5G 1X8, Canada.
- Department of Molecular Genetics, University of Toronto, Toronto, ON, M5S 1A8, Canada.
| |
Collapse
|
28
|
Madsen AB, Knudsen JR, Henriquez-Olguin C, Angin Y, Zaal KJ, Sylow L, Schjerling P, Ralston E, Jensen TE. β-Actin shows limited mobility and is required only for supraphysiological insulin-stimulated glucose transport in young adult soleus muscle. Am J Physiol Endocrinol Metab 2018; 315. [PMID: 29533739 PMCID: PMC6087721 DOI: 10.1152/ajpendo.00392.2017] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Studies in skeletal muscle cell cultures suggest that the cortical actin cytoskeleton is a major requirement for insulin-stimulated glucose transport, implicating the β-actin isoform, which in many cell types is the main actin isoform. However, it is not clear that β-actin plays such a role in mature skeletal muscle. Neither dependency of glucose transport on β-actin nor actin reorganization upon glucose transport have been tested in mature muscle. To investigate the role of β-actin in fully differentiated muscle, we performed a detailed characterization of wild type and muscle-specific β-actin knockout (KO) mice. The effects of the β-actin KO were subtle; however, we confirmed the previously reported decline in running performance of β-actin KO mice compared with wild type during repeated maximal running tests. We also found insulin-stimulated glucose transport into incubated muscles reduced in soleus but not in extensor digitorum longus muscle of young adult mice. Contraction-stimulated glucose transport trended toward the same pattern, but the glucose transport phenotype disappeared in soleus muscles from mature adult mice. No genotype-related differences were found in body composition or glucose tolerance or by indirect calorimetry measurements. To evaluate β-actin mobility in mature muscle, we electroporated green fluorescent protein (GFP)-β-actin into flexor digitorum brevis muscle fibers and measured fluorescence recovery after photobleaching. GFP-β-actin showed limited unstimulated mobility and no changes after insulin stimulation. In conclusion, β-actin is not required for glucose transport regulation in mature mouse muscle under the majority of the tested conditions. Thus, our work reveals fundamental differences in the role of the cortical β-actin cytoskeleton in mature muscle compared with cell culture.
Collapse
Affiliation(s)
- Agnete B Madsen
- Department of Nutrition, Exercise and Sports, University of Copenhagen , Copenhagen , Denmark
| | - Jonas R Knudsen
- Department of Nutrition, Exercise and Sports, University of Copenhagen , Copenhagen , Denmark
| | - Carlos Henriquez-Olguin
- Department of Nutrition, Exercise and Sports, University of Copenhagen , Copenhagen , Denmark
- Centro de Estudios Moleculares de la Célula, Instituto de Ciencias Biomédicas, Universidad de Chile ; Laboratory of Exercise Sciences, Clínica MEDS, Santiago , Chile
| | - Yeliz Angin
- Department of Nutrition, Exercise and Sports, University of Copenhagen , Copenhagen , Denmark
| | - Kristien J Zaal
- Light Imaging Section, Office of Science and Technology, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health , Bethesda, Maryland
| | - Lykke Sylow
- Department of Nutrition, Exercise and Sports, University of Copenhagen , Copenhagen , Denmark
| | - Peter Schjerling
- Institute of Sports Medicine, Department of Orthopedic Surgery, Bispebjerg Hospital , Copenhagen , Denmark
- Center of Healthy Aging, Faculty of Health and Medical Sciences, University of Copenhagen , Copenhagen , Denmark
| | - Evelyn Ralston
- Light Imaging Section, Office of Science and Technology, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health , Bethesda, Maryland
| | - Thomas E Jensen
- Department of Nutrition, Exercise and Sports, University of Copenhagen , Copenhagen , Denmark
| |
Collapse
|
29
|
Vedula P, Kashina A. The makings of the 'actin code': regulation of actin's biological function at the amino acid and nucleotide level. J Cell Sci 2018; 131:131/9/jcs215509. [PMID: 29739859 DOI: 10.1242/jcs.215509] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
The actin cytoskeleton plays key roles in every eukaryotic cell and is essential for cell adhesion, migration, mechanosensing, and contractility in muscle and non-muscle tissues. In higher vertebrates, from birds through to mammals, actin is represented by a family of six conserved genes. Although these genes have evolved independently for more than 100 million years, they encode proteins with ≥94% sequence identity, which are differentially expressed in different tissues, and tightly regulated throughout embryogenesis and adulthood. It has been previously suggested that the existence of such similar actin genes is a fail-safe mechanism to preserve the essential function of actin through redundancy. However, knockout studies in mice and other organisms demonstrate that the different actins have distinct biological roles. The mechanisms maintaining this distinction have been debated in the literature for decades. This Review summarizes data on the functional regulation of different actin isoforms, and the mechanisms that lead to their different biological roles in vivo We focus here on recent studies demonstrating that at least some actin functions are regulated beyond the amino acid level at the level of the actin nucleotide sequence.
Collapse
Affiliation(s)
- Pavan Vedula
- Department of Biomedical Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Anna Kashina
- Department of Biomedical Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
30
|
O'Rourke AR, Lindsay A, Tarpey MD, Yuen S, McCourt P, Nelson DM, Perrin BJ, Thomas DD, Spangenburg EE, Lowe DA, Ervasti JM. Impaired muscle relaxation and mitochondrial fission associated with genetic ablation of cytoplasmic actin isoforms. FEBS J 2018; 285:481-500. [PMID: 29265728 DOI: 10.1111/febs.14367] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Revised: 11/06/2017] [Accepted: 12/13/2017] [Indexed: 12/28/2022]
Abstract
While α-actin isoforms predominate in adult striated muscle, skeletal muscle-specific knockouts (KOs) of nonmuscle cytoplasmic βcyto - or γcyto -actin each cause a mild, but progressive myopathy effected by an unknown mechanism. Using transmission electron microscopy, we identified morphological abnormalities in both the mitochondria and the sarcoplasmic reticulum (SR) in aged muscle-specific βcyto - and γcyto -actin KO mice. We found βcyto - and γcyto -actin proteins to be enriched in isolated mitochondrial-associated membrane preparations, which represent the interface between mitochondria and sarco-endoplasmic reticulum important in signaling and mitochondrial dynamics. We also measured significantly elongated and interconnected mitochondrial morphologies associated with a significant decrease in mitochondrial fission events in primary mouse embryonic fibroblasts lacking βcyto - and/or γcyto -actin. Interestingly, mitochondrial respiration in muscle was not measurably affected as oxygen consumption was similar in skeletal muscle fibers from 12 month-old muscle-specific βcyto - and γcyto -actin KO mice. Instead, we found that the maximal rate of relaxation after isometric contraction was significantly slowed in muscles of 12-month-old βcyto - and γcyto -actin muscle-specific KO mice. Our data suggest that impaired Ca2+ re-uptake may presage development of the observed SR morphological changes in aged mice while providing a potential pathological mechanism for the observed myopathy.
Collapse
Affiliation(s)
- Allison R O'Rourke
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, MN, USA
| | - Angus Lindsay
- Department of Rehabilitation Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Michael D Tarpey
- Department of Physiology, East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC, USA
| | - Samantha Yuen
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN, USA
| | - Preston McCourt
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN, USA
| | - D'anna M Nelson
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN, USA
| | - Benjamin J Perrin
- Department of Biology, Indiana University-Purdue University Indianapolis, IN, USA
| | - David D Thomas
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN, USA
| | - Espen E Spangenburg
- Department of Physiology, East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC, USA
| | - Dawn A Lowe
- Department of Rehabilitation Medicine, University of Minnesota, Minneapolis, MN, USA
| | - James M Ervasti
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
31
|
Roman W, Martins JP, Carvalho FA, Voituriez R, Abella JV, Santos NC, Cadot B, Way M, Gomes ER. Myofibril contraction and crosslinking drive nuclear movement to the periphery of skeletal muscle. Nat Cell Biol 2017; 19:1189-1201. [PMID: 28892082 PMCID: PMC5675053 DOI: 10.1038/ncb3605] [Citation(s) in RCA: 85] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Accepted: 08/04/2017] [Indexed: 12/17/2022]
Abstract
Nuclear movements are important for multiple cellular functions, and are driven by polarized forces generated by motor proteins and the cytoskeleton. During skeletal myofibre formation or regeneration, nuclei move from the centre to the periphery of the myofibre for proper muscle function. Centrally located nuclei are also found in different muscle disorders. Using theoretical and experimental approaches, we demonstrate that nuclear movement to the periphery of myofibres is mediated by centripetal forces around the nucleus. These forces arise from myofibril contraction and crosslinking that 'zip' around the nucleus in combination with tight regulation of nuclear stiffness by lamin A/C. In addition, an Arp2/3 complex containing Arpc5L together with γ-actin is required to organize desmin to crosslink myofibrils for nuclear movement. Our work reveals that centripetal forces exerted by myofibrils squeeze the nucleus to the periphery of myofibres.
Collapse
Affiliation(s)
- William Roman
- Sorbonne Universités, UPMC Univ Paris 06, INSERM UMRS974, CNRS FRE3617, Center for Research in Myology, GH Pitié-Salpêtrière, 47 Boulevard de l'hôpital, 75013 Paris, France; Centre de Référence de Pathologie Neuromusculaire Paris-Est, Institut de Myologie, GHU La Pitié-Salpêtrière, Assistance Publique-Hôpitaux de Paris, Paris, France
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Avenida Professor Egas Moniz, 1649-028, Lisboa, Portugal
| | - Joao P. Martins
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Avenida Professor Egas Moniz, 1649-028, Lisboa, Portugal
| | - Filomena A. Carvalho
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Avenida Professor Egas Moniz, 1649-028, Lisboa, Portugal
| | - Raphael Voituriez
- Laboratoire de Physique Théorique de la Matière Condensée; CNRS UMR 7600; Université Pierre et Marie Curie, Paris , France
- Laboratoire Jean Perrin; CNRS FRE 3231, Université Pierre et Marie Curie ; Paris, France
| | - Jasmine V.G. Abella
- Cellular Signalling and Cytoskeletal Function, The Francis Crick Institute, 44 Lincoln’s Inn Fields, London, WC2A 3LY, UK
| | - Nuno C. Santos
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Avenida Professor Egas Moniz, 1649-028, Lisboa, Portugal
| | - Bruno Cadot
- Sorbonne Universités, UPMC Univ Paris 06, INSERM UMRS974, CNRS FRE3617, Center for Research in Myology, GH Pitié-Salpêtrière, 47 Boulevard de l'hôpital, 75013 Paris, France; Centre de Référence de Pathologie Neuromusculaire Paris-Est, Institut de Myologie, GHU La Pitié-Salpêtrière, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Michael Way
- Cellular Signalling and Cytoskeletal Function, The Francis Crick Institute, 44 Lincoln’s Inn Fields, London, WC2A 3LY, UK
| | - Edgar R. Gomes
- Sorbonne Universités, UPMC Univ Paris 06, INSERM UMRS974, CNRS FRE3617, Center for Research in Myology, GH Pitié-Salpêtrière, 47 Boulevard de l'hôpital, 75013 Paris, France; Centre de Référence de Pathologie Neuromusculaire Paris-Est, Institut de Myologie, GHU La Pitié-Salpêtrière, Assistance Publique-Hôpitaux de Paris, Paris, France
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Avenida Professor Egas Moniz, 1649-028, Lisboa, Portugal
| |
Collapse
|
32
|
Willems SM, Wright DJ, Day FR, Trajanoska K, Joshi PK, Morris JA, Matteini AM, Garton FC, Grarup N, Oskolkov N, Thalamuthu A, Mangino M, Liu J, Demirkan A, Lek M, Xu L, Wang G, Oldmeadow C, Gaulton KJ, Lotta LA, Miyamoto-Mikami E, Rivas MA, White T, Loh PR, Aadahl M, Amin N, Attia JR, Austin K, Benyamin B, Brage S, Cheng YC, Cięszczyk P, Derave W, Eriksson KF, Eynon N, Linneberg A, Lucia A, Massidda M, Mitchell BD, Miyachi M, Murakami H, Padmanabhan S, Pandey A, Papadimitriou I, Rajpal DK, Sale C, Schnurr TM, Sessa F, Shrine N, Tobin MD, Varley I, Wain LV, Wray NR, Lindgren CM, MacArthur DG, Waterworth DM, McCarthy MI, Pedersen O, Khaw KT, Kiel DP, Pitsiladis Y, Fuku N, Franks PW, North KN, van Duijn CM, Mather KA, Hansen T, Hansson O, Spector T, Murabito JM, Richards JB, Rivadeneira F, Langenberg C, Perry JRB, Wareham NJ, Scott RA. Large-scale GWAS identifies multiple loci for hand grip strength providing biological insights into muscular fitness. Nat Commun 2017; 8:16015. [PMID: 29313844 PMCID: PMC5510175 DOI: 10.1038/ncomms16015] [Citation(s) in RCA: 135] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Accepted: 05/22/2017] [Indexed: 02/02/2023] Open
Abstract
Hand grip strength is a widely used proxy of muscular fitness, a marker of frailty, and predictor of a range of morbidities and all-cause mortality. To investigate the genetic determinants of variation in grip strength, we perform a large-scale genetic discovery analysis in a combined sample of 195,180 individuals and identify 16 loci associated with grip strength (P<5 × 10-8) in combined analyses. A number of these loci contain genes implicated in structure and function of skeletal muscle fibres (ACTG1), neuronal maintenance and signal transduction (PEX14, TGFA, SYT1), or monogenic syndromes with involvement of psychomotor impairment (PEX14, LRPPRC and KANSL1). Mendelian randomization analyses are consistent with a causal effect of higher genetically predicted grip strength on lower fracture risk. In conclusion, our findings provide new biological insight into the mechanistic underpinnings of grip strength and the causal role of muscular strength in age-related morbidities and mortality.
Collapse
Affiliation(s)
- Sara M. Willems
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Cambridge CB2 0QQ, UK
| | - Daniel J. Wright
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Cambridge CB2 0QQ, UK
| | - Felix R. Day
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Cambridge CB2 0QQ, UK
| | - Katerina Trajanoska
- Department of Internal Medicine, Erasmus Medical Center, 3015 CE Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus Medical Center, 3015 CE Rotterdam, The Netherlands
| | - Peter K. Joshi
- Usher Institute for Population Health Sciences and Informatics, University of Edinburgh, Edinburgh EH8 9AB, UK
| | - John A. Morris
- Centre for Clinical Epidemiology, Lady Davis Institute for Medical Research, Jewish General Hospital, McGill University, Montreal, Quebec, Canada QC H3T 1E2
- Department of Human Genetics, McGill University, Montreal, Quebec, Canada H3G 0B1
| | - Amy M. Matteini
- Division of Geriatric Medicine and Gerontology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287, USA
| | - Fleur C. Garton
- Queensland Brain Institute, University of Queensland, St Lucia, Queensland 4072, Australia
| | - Niels Grarup
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Nikolay Oskolkov
- Lund University Diabetes Center, Department of Clinical Sciences, Diabetes and Endocrinology, Skånes University Hospital, 222 41 Lund, Sweden
| | - Anbupalam Thalamuthu
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, Sydney, New South Wales 2031, Australia
| | - Massimo Mangino
- Department of Twin Research & Genetic Epidemiology, Kings College London, London SE1 7EH, UK
- NIHR Biomedical Research Centre at Guy’s and St. Thomas’ NHS Foundation Trust, London SE1 9RT, UK
| | - Jun Liu
- Department of Epidemiology, Erasmus Medical Center, 3015 CE Rotterdam, The Netherlands
| | - Ayse Demirkan
- Department of Epidemiology, Erasmus Medical Center, 3015 CE Rotterdam, The Netherlands
- Department of Human Genetics, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Monkol Lek
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, Maryland 02114, USA
- Harvard Medical School, Boston, Maryland 02115, USA
| | - Liwen Xu
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, Maryland 02114, USA
- Harvard Medical School, Boston, Maryland 02115, USA
| | - Guan Wang
- Centre for Sport and Exercise Science and Medicine (SESAME), University of Brighton, Eastbourne BN20 7SN, UK
| | | | - Kyle J. Gaulton
- Department of Pediatrics, University of California San Diego, La Jolla, California 92093, USA
| | - Luca A. Lotta
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Cambridge CB2 0QQ, UK
| | - Eri Miyamoto-Mikami
- Japan Society for the Promotion of Science, Tokyo 102-0083, Japan
- Department of Sports and Life Science, National Institute of Fitness and Sports, Kanoya, Kagoshima 891-2393, Japan
| | - Manuel A. Rivas
- Department of Biomedical Data Sciences, Stanford University, Stanford, California 94305, USA
- BROAD Institute of the Massachusetts Institute of Technology and Harvard University, Cambridge, Massachusetts 02142, USA
| | - Tom White
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Cambridge CB2 0QQ, UK
| | - Po-Ru Loh
- BROAD Institute of the Massachusetts Institute of Technology and Harvard University, Cambridge, Massachusetts 02142, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts 02115, USA
| | - Mette Aadahl
- Research Centre for Prevention and Health, Capital Region of Denmark, Glostrup University Hospital, DK-2600 Glostrup, Denmark
| | - Najaf Amin
- Department of Epidemiology, Erasmus Medical Center, 3015 CE Rotterdam, The Netherlands
| | - John R. Attia
- Hunter Medical Research Institute, Newcastle, New South Wales 2305, Australia
- Faculty of Health and Medicine, University of Newcastle, Newcastle, New South Wales 2308, Australia
- John Hunter Hospital, New Lambton, New South Wales 2305, Australia
| | - Krista Austin
- Centre for Sport and Exercise Science and Medicine (SESAME), University of Brighton, Eastbourne BN20 7SN, UK
| | - Beben Benyamin
- Queensland Brain Institute, University of Queensland, St Lucia, Queensland 4072, Australia
- Institute for Molecular Bioscience, University of Queensland, St Lucia, Queensland 4072, Australia
| | - Søren Brage
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Cambridge CB2 0QQ, UK
| | - Yu-Ching Cheng
- Division of Endocrinology Diabetes and Nutrition, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland 21201, USA
| | - Paweł Cięszczyk
- Faculty of Physical Education, Gdańsk University of Physical Education and Sport, 80-336 Gdańsk, Poland
| | - Wim Derave
- Department of Movement and Sports Sciences, Ghent University, 9000 Ghent, Belgium
| | - Karl-Fredrik Eriksson
- Lund University Diabetes Center, Department of Clinical Sciences, Diabetes and Endocrinology, Skånes University Hospital, 222 41 Lund, Sweden
| | - Nir Eynon
- Institute of Sport, Exercise & Active Living (ISEAL), Victoria University, Melbourne, Victoria 8001, Australia
- Murdoch Children’s Research Institute, Royal Children’s Hospital, Melbourne, Victoria 3052, Australia
| | - Allan Linneberg
- Research Centre for Prevention and Health, Capital Region of Denmark, Glostrup University Hospital, DK-2600 Glostrup, Denmark
- Department of Clinical Experimental Research, Rigshospitalet, 2600 Glostrup, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark
| | - Alejandro Lucia
- Universidad Europea de Madrid, 28670 Villaviciosa de Odón, Madrid, Spain
- Research Institute ‘i+12’, Hospital Universitario 12 de Octubre, 28041 Madrid, Spain
| | - Myosotis Massidda
- Department of Life and Environmental Sciences, University of Cagliari, 09124 Cagliari, Italy
| | - Braxton D. Mitchell
- Division of Endocrinology Diabetes and Nutrition, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland 21201, USA
- Geriatrics Research and Education Clinical Center, Baltimore Veterans Administration Medical Center, Baltimore, Maryland 21201, USA
| | - Motohiko Miyachi
- National Institutes of Biomedical Innovation, Health and Nutrition, Tokyo 162-8636, Japan
| | - Haruka Murakami
- National Institutes of Biomedical Innovation, Health and Nutrition, Tokyo 162-8636, Japan
| | - Sandosh Padmanabhan
- British Heart Foundation Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, UK
| | - Ashutosh Pandey
- Target Sciences, GlaxoSmithKline, King of Prussia, Pennsylvania 19406, USA
| | - Ioannis Papadimitriou
- Institute of Sport, Exercise & Active Living (ISEAL), Victoria University, Melbourne, Victoria 8001, Australia
| | - Deepak K. Rajpal
- Target Sciences, GlaxoSmithKline, King of Prussia, Pennsylvania 19406, USA
| | - Craig Sale
- Musculoskeletal Physiology Research Group, Sport, Health and Performance Enhancement (SHAPE) Research Centre, Nottingham Trent University, Nottingham NG1 4FQ, UK
| | - Theresia M. Schnurr
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Francesco Sessa
- Department of Clinical and Experimental Medicine, Medical Genetics, University of Foggia, 71122 Foggia FG, Italy
| | - Nick Shrine
- Department of Health Sciences, University of Leicester, Leicester LE1 7RH, UK
- National Institute for Health Research, Leicester Respiratory Biomedical Research Unit, Glenfield Hospital, Leicester LE3 9QP, UK
| | - Martin D. Tobin
- Department of Health Sciences, University of Leicester, Leicester LE1 7RH, UK
- National Institute for Health Research, Leicester Respiratory Biomedical Research Unit, Glenfield Hospital, Leicester LE3 9QP, UK
| | - Ian Varley
- Musculoskeletal Physiology Research Group, Sport, Health and Performance Enhancement (SHAPE) Research Centre, Nottingham Trent University, Nottingham NG1 4FQ, UK
| | - Louise V. Wain
- Department of Health Sciences, University of Leicester, Leicester LE1 7RH, UK
- National Institute for Health Research, Leicester Respiratory Biomedical Research Unit, Glenfield Hospital, Leicester LE3 9QP, UK
| | - Naomi R. Wray
- Queensland Brain Institute, University of Queensland, St Lucia, Queensland 4072, Australia
- Institute for Molecular Bioscience, University of Queensland, St Lucia, Queensland 4072, Australia
| | - Cecilia M. Lindgren
- BROAD Institute of the Massachusetts Institute of Technology and Harvard University, Cambridge, Massachusetts 02142, USA
- The Big Data Institute, University of Oxford, Oxford OX3 7BN, UK
- Wellcome Trust Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7BN, UK
| | - Daniel G. MacArthur
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, Maryland 02114, USA
- BROAD Institute of the Massachusetts Institute of Technology and Harvard University, Cambridge, Massachusetts 02142, USA
| | - Dawn M. Waterworth
- Target Sciences, GlaxoSmithKline, King of Prussia, Pennsylvania 19406, USA
| | - Mark I. McCarthy
- Wellcome Trust Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7BN, UK
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford OX3 7LE, UK
- NIHR Oxford Biomedical Research Centre, Oxford OX3 7LE, UK
| | - Oluf Pedersen
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Kay-Tee Khaw
- Department of Public Health and Primary Care, University of Cambridge, Cambridge CB2 0SR, UK
| | - Douglas P. Kiel
- Harvard Medical School, Boston, Maryland 02115, USA
- Institute for Aging Research, Hebrew SeniorLife, Boston, Massachusetts 02131, USA
- Department of Medicine, Beth Israel Deaconess Medical Centre, Boston, Massachusetts 02215, USA
| | - Yannis Pitsiladis
- Centre for Sport and Exercise Science and Medicine (SESAME), University of Brighton, Eastbourne BN20 7SN, UK
| | - Noriyuki Fuku
- Graduate School of Health and Sports Science, Juntendo University, Chiba 270-1695, Japan
| | - Paul W. Franks
- Genetic and Molecular Epidemiology Unit, Department of Clinical Sciences, Lund University, Skånes University Hospital, 222 41 Lund, Sweden
- Public Health and Clinical Medicine, Section for Medicine, Umeå University, 901 87 Umeå, Sweden
- Biobank Research, Umeå University, 901 87 Umeå, Sweden
| | - Kathryn N. North
- Murdoch Children’s Research Institute, Royal Children’s Hospital, Melbourne, Victoria 3052, Australia
| | - Cornelia M. van Duijn
- Department of Epidemiology, Erasmus Medical Center, 3015 CE Rotterdam, The Netherlands
| | - Karen A. Mather
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, Sydney, New South Wales 2031, Australia
| | - Torben Hansen
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
- Faculty of Health Sciences, University of Southern Denmark, 5230 Odense M, Denmark
| | - Ola Hansson
- Lund University Diabetes Center, Department of Clinical Sciences, Diabetes and Endocrinology, Skånes University Hospital, 222 41 Lund, Sweden
| | - Tim Spector
- Department of Twin Research & Genetic Epidemiology, Kings College London, London SE1 7EH, UK
| | - Joanne M. Murabito
- Boston University School of Medicine, Department of Medicine, Section of General Internal Medicine, Boston, Massachusetts 02118, USA
- National Heart Lung and Blood Institute’s and Boston University’s Framingham Heart Study, Framingham, Massachusetts 01702, USA
| | - J. Brent Richards
- Centre for Clinical Epidemiology, Lady Davis Institute for Medical Research, Jewish General Hospital, McGill University, Montreal, Quebec, Canada QC H3T 1E2
- Department of Human Genetics, McGill University, Montreal, Quebec, Canada H3G 0B1
- Department of Twin Research & Genetic Epidemiology, Kings College London, London SE1 7EH, UK
- Department of Medicine, McGill University, Montreal, Quebec, Canada H3G 1A4
| | - Fernando Rivadeneira
- Department of Internal Medicine, Erasmus Medical Center, 3015 CE Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus Medical Center, 3015 CE Rotterdam, The Netherlands
| | - Claudia Langenberg
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Cambridge CB2 0QQ, UK
| | - John R. B. Perry
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Cambridge CB2 0QQ, UK
| | - Nick J. Wareham
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Cambridge CB2 0QQ, UK
| | - Robert A. Scott
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Cambridge CB2 0QQ, UK
| |
Collapse
|
33
|
González-Jamett AM, Baez-Matus X, Olivares MJ, Hinostroza F, Guerra-Fernández MJ, Vasquez-Navarrete J, Bui MT, Guicheney P, Romero NB, Bevilacqua JA, Bitoun M, Caviedes P, Cárdenas AM. Dynamin-2 mutations linked to Centronuclear Myopathy impair actin-dependent trafficking in muscle cells. Sci Rep 2017; 7:4580. [PMID: 28676641 PMCID: PMC5496902 DOI: 10.1038/s41598-017-04418-w] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Accepted: 05/16/2017] [Indexed: 12/20/2022] Open
Abstract
Dynamin-2 is a ubiquitously expressed GTP-ase that mediates membrane remodeling. Recent findings indicate that dynamin-2 also regulates actin dynamics. Mutations in dynamin-2 cause dominant centronuclear myopathy (CNM), a congenital myopathy characterized by progressive weakness and atrophy of skeletal muscles. However, the muscle-specific roles of dynamin-2 affected by these mutations remain elusive. Here we show that, in muscle cells, the GTP-ase activity of dynamin-2 is involved in de novo actin polymerization as well as in actin-mediated trafficking of the glucose transporter GLUT4. Expression of dynamin-2 constructs carrying CNM-linked mutations disrupted the formation of new actin filaments as well as the stimulus-induced translocation of GLUT4 to the plasma membrane. Similarly, mature muscle fibers isolated from heterozygous knock-in mice that harbor the dynamin-2 mutation p.R465W, an animal model of CNM, exhibited altered actin organization, reduced actin polymerization and impaired insulin-induced translocation of GLUT4 to the sarcolemma. Moreover, GLUT4 displayed aberrant perinuclear accumulation in biopsies from CNM patients carrying dynamin-2 mutations, further suggesting trafficking defects. These results suggest that dynamin-2 is a key regulator of actin dynamics and GLUT4 trafficking in muscle cells. Our findings also support a model in which impairment of actin-dependent trafficking contributes to the pathological mechanism in dynamin-2-associated CNM.
Collapse
Affiliation(s)
- Arlek M González-Jamett
- Centro Interdisciplinario de Neurociencia de Valparaíso. Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile. .,Programa de Farmacología Molecular y Clinica, ICBM, Facultad de Medicina, Universidad de Chile, Santiago, Chile.
| | - Ximena Baez-Matus
- Centro Interdisciplinario de Neurociencia de Valparaíso. Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - María José Olivares
- Centro Interdisciplinario de Neurociencia de Valparaíso. Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Fernando Hinostroza
- Centro Interdisciplinario de Neurociencia de Valparaíso. Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile.,Doctorado en Ciencias, mención Neurociencia, Universidad de Valparaíso, Valparaíso, Chile
| | - Maria José Guerra-Fernández
- Centro Interdisciplinario de Neurociencia de Valparaíso. Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Jacqueline Vasquez-Navarrete
- Centro Interdisciplinario de Neurociencia de Valparaíso. Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Mai Thao Bui
- Université Sorbonne, UPMC Univ Paris 06, INSERM UMRS974, CNRS FRE3617, Center for Research in Myology, Paris, France.,Centre de référence de Pathologie Neuromusculaire Paris-Est, Institut de Myologie, GHU Pitié-Salpêtrière, Assistance Publique-Hôpitaux de Paris, GH Pitié-Salpêtrière, Paris, France
| | - Pascale Guicheney
- INSERM, UMR_S1166, Sorbonne Universités, UPMC Univ Paris 06, UMR_S1166, Institute of Cardiometabolism and Nutrition (ICAN), Paris, France
| | - Norma Beatriz Romero
- Université Sorbonne, UPMC Univ Paris 06, INSERM UMRS974, CNRS FRE3617, Center for Research in Myology, Paris, France.,Centre de référence de Pathologie Neuromusculaire Paris-Est, Institut de Myologie, GHU Pitié-Salpêtrière, Assistance Publique-Hôpitaux de Paris, GH Pitié-Salpêtrière, Paris, France
| | - Jorge A Bevilacqua
- Programa de Anatomía y Biología del Desarrollo, ICBM, Facultad de Medicina, Departamento de Neurología y Neurocirugía, Hospital Clínico Universidad de Chile, Universidad de Chile, Santiago, Chile
| | - Marc Bitoun
- Research Center for Myology, UPMC Univ Paris 06 and INSERM UMRS 974, Institute of Myology, Paris, France
| | - Pablo Caviedes
- Programa de Farmacología Molecular y Clinica, ICBM, Facultad de Medicina, Universidad de Chile, Santiago, Chile.
| | - Ana M Cárdenas
- Centro Interdisciplinario de Neurociencia de Valparaíso. Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| |
Collapse
|
34
|
Zhan X, Jiao J, Zhang H, Li C, Zhao J, Liao L, Wu J, Wu B, Wu Z, Wang S, Du Z, Shen J, Zou H, Neufeld G, Xu L, Li E. A three-gene signature from protein-protein interaction network of LOXL2- and actin-related proteins for esophageal squamous cell carcinoma prognosis. Cancer Med 2017; 6:1707-1719. [PMID: 28556501 PMCID: PMC5504325 DOI: 10.1002/cam4.1096] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Revised: 04/06/2017] [Accepted: 04/18/2017] [Indexed: 02/05/2023] Open
Abstract
Current staging is inadequate for predicting clinical outcome of esophageal squamous cell carcinoma (ESCC). Aberrant expression of LOXL2 and actin-related proteins plays important roles in ESCC. Here, we aimed to develop a novel molecular signature that exceeds the power of the current staging system in predicting ESCC prognosis. We found that LOXL2 colocalized with filamentous actin in ESCC cells, and gene set enrichment analysis (GSEA) showed that LOXL2 is related to the actin cytoskeleton. An ESCC-specific protein-protein interaction (PPI) network involving LOXL2 and actin-related proteins was generated based on genome-wide RNA-seq in 15 paired ESCC samples, and the prognostic significance of 14 core genes was analyzed. Using risk score calculation, a three-gene signature comprising LOXL2, CDH1, and FN1 was derived from transcriptome data of patients with ESCC. The high-risk three-gene signature strongly correlated with poor prognosis in a training cohort of 60 patients (P = 0.003). In mRNA and protein levels, the prognostic values of this signature were further validated in 243 patients from a testing cohort (P = 0.001) and two validation cohorts (P = 0.021, P = 0.007). Furthermore, Cox regression analysis revealed that the signature was an independent prognostic factor. Compared with using the signature or TNM stage alone, the combined model significantly enhanced the accuracy in evaluating ESCC prognosis. In conclusion, our data reveal that the tumor-promoting role of LOXL2 in ESCC is mediated by perturbing the architecture of actin cytoskeleton through its PPIs. We generated a novel three-gene signature (PPI interfaces) that robustly predicts poor clinical outcome in ESCC patients.
Collapse
Affiliation(s)
- Xiu‐hui Zhan
- Department of Biochemistry and Molecular BiologyShantou University Medical CollegeShantouGuangdongChina
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan AreaShantou University Medical CollegeShantouGuangdongChina
| | - Ji‐wei Jiao
- Department of Biochemistry and Molecular BiologyShantou University Medical CollegeShantouGuangdongChina
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan AreaShantou University Medical CollegeShantouGuangdongChina
| | - Hai‐feng Zhang
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan AreaShantou University Medical CollegeShantouGuangdongChina
| | - Chun‐quan Li
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan AreaShantou University Medical CollegeShantouGuangdongChina
- College of Medical InformaticsDaqing CampusHarbin Medical UniversityDaqingHeilongjiangChina
| | - Jian‐mei Zhao
- Department of Biochemistry and Molecular BiologyShantou University Medical CollegeShantouGuangdongChina
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan AreaShantou University Medical CollegeShantouGuangdongChina
| | - Lian‐di Liao
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan AreaShantou University Medical CollegeShantouGuangdongChina
- Institute of Oncologic PathologyShantou University Medical CollegeShantouGuangdongChina
| | - Jian‐yi Wu
- Department of Biochemistry and Molecular BiologyShantou University Medical CollegeShantouGuangdongChina
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan AreaShantou University Medical CollegeShantouGuangdongChina
| | - Bing‐li Wu
- Department of Biochemistry and Molecular BiologyShantou University Medical CollegeShantouGuangdongChina
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan AreaShantou University Medical CollegeShantouGuangdongChina
| | - Zhi‐yong Wu
- Department of Tumor SurgeryShantou Central HospitalAffiliated Shantou Hospital of Sun Yat‐sen UniversityShantouGuangdongChina
| | - Shao‐hong Wang
- Department of PathologyShantou Central HospitalAffiliated Shantou Hospital of Sun Yat‐sen UniversityShantouGuangdongChina
| | - Ze‐peng Du
- Department of PathologyShantou Central HospitalAffiliated Shantou Hospital of Sun Yat‐sen UniversityShantouGuangdongChina
| | - Jin‐hui Shen
- Department of PathologyShantou Central HospitalAffiliated Shantou Hospital of Sun Yat‐sen UniversityShantouGuangdongChina
| | - Hai‐ying Zou
- Department of Biochemistry and Molecular BiologyShantou University Medical CollegeShantouGuangdongChina
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan AreaShantou University Medical CollegeShantouGuangdongChina
| | - Gera Neufeld
- Cancer Research and Vascular Biology CenterThe Bruce Rappaport Faculty of MedicineTechnionIsrael Institute of TechnologyHaifaIsrael
| | - Li‐yan Xu
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan AreaShantou University Medical CollegeShantouGuangdongChina
- Institute of Oncologic PathologyShantou University Medical CollegeShantouGuangdongChina
| | - En‐min Li
- Department of Biochemistry and Molecular BiologyShantou University Medical CollegeShantouGuangdongChina
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan AreaShantou University Medical CollegeShantouGuangdongChina
| |
Collapse
|
35
|
Abstract
Cardiac and skeletal striated muscles are intricately designed machines responsible for muscle contraction. Coordination of the basic contractile unit, the sarcomere, and the complex cytoskeletal networks are critical for contractile activity. The sarcomere is comprised of precisely organized individual filament systems that include thin (actin), thick (myosin), titin, and nebulin. Connecting the sarcomere to other organelles (e.g., mitochondria and nucleus) and serving as the scaffold to maintain cellular integrity are the intermediate filaments. The costamere, on the other hand, tethers the sarcomere to the cell membrane. Unique structures like the intercalated disc in cardiac muscle and the myotendinous junction in skeletal muscle help synchronize and transmit force. Intense investigation has been done on many of the proteins that make up these cytoskeletal assemblies. Yet the details of their function and how they interconnect have just started to be elucidated. A vast number of human myopathies are contributed to mutations in muscle proteins; thus understanding their basic function provides a mechanistic understanding of muscle disorders. In this review, we highlight the components of striated muscle with respect to their interactions, signaling pathways, functions, and connections to disease. © 2017 American Physiological Society. Compr Physiol 7:891-944, 2017.
Collapse
Affiliation(s)
- Christine A Henderson
- Department of Cellular and Molecular Medicine, The University of Arizona, Tucson, Arizona, USA.,Sarver Molecular Cardiovascular Research Program, The University of Arizona, Tucson, Arizona, USA
| | - Christopher G Gomez
- Department of Cellular and Molecular Medicine, The University of Arizona, Tucson, Arizona, USA.,Sarver Molecular Cardiovascular Research Program, The University of Arizona, Tucson, Arizona, USA
| | - Stefanie M Novak
- Department of Cellular and Molecular Medicine, The University of Arizona, Tucson, Arizona, USA.,Sarver Molecular Cardiovascular Research Program, The University of Arizona, Tucson, Arizona, USA
| | - Lei Mi-Mi
- Department of Cellular and Molecular Medicine, The University of Arizona, Tucson, Arizona, USA.,Sarver Molecular Cardiovascular Research Program, The University of Arizona, Tucson, Arizona, USA
| | - Carol C Gregorio
- Department of Cellular and Molecular Medicine, The University of Arizona, Tucson, Arizona, USA.,Sarver Molecular Cardiovascular Research Program, The University of Arizona, Tucson, Arizona, USA
| |
Collapse
|
36
|
Moradi M, Sivadasan R, Saal L, Lüningschrör P, Dombert B, Rathod RJ, Dieterich DC, Blum R, Sendtner M. Differential roles of α-, β-, and γ-actin in axon growth and collateral branch formation in motoneurons. J Cell Biol 2017; 216:793-814. [PMID: 28246119 PMCID: PMC5346967 DOI: 10.1083/jcb.201604117] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Revised: 11/11/2016] [Accepted: 01/17/2017] [Indexed: 12/17/2022] Open
Abstract
α-, β-, and γ-actin differentially regulate cytoskeletal dynamics and stability in axons of motoneurons. Locally translated α-actin contributes to stable actin filaments in axonal branches, whereas β- and γ-actin give rise to highly dynamic filaments that modulate growth cone dynamics. Axonal branching and terminal arborization are fundamental events during the establishment of synaptic connectivity. They are triggered by assembly of actin filaments along axon shafts giving rise to filopodia. The specific contribution of the three actin isoforms, Actα, Actβ, and Actγ, to filopodia stability and dynamics during this process is not well understood. Here, we report that Actα, Actβ, and Actγ isoforms are expressed in primary mouse motoneurons and their transcripts are translocated into axons. shRNA-mediated depletion of Actα reduces axonal filopodia dynamics and disturbs collateral branch formation. Knockdown of Actβ reduces dynamic movements of growth cone filopodia and impairs presynaptic differentiation. Ablation of Actβ or Actγ leads to compensatory up-regulation of the two other isoforms, which allows maintenance of total actin levels and preserves F-actin polymerization. Collectively, our data provide evidence for specific roles of different actin isoforms in spatial regulation of actin dynamics and stability in axons of developing motoneurons.
Collapse
Affiliation(s)
- Mehri Moradi
- Institute of Clinical Neurobiology, University Hospital Wuerzburg, University of Wuerzburg, 97078 Wuerzburg, Germany
| | - Rajeeve Sivadasan
- Institute of Clinical Neurobiology, University Hospital Wuerzburg, University of Wuerzburg, 97078 Wuerzburg, Germany
| | - Lena Saal
- Institute of Clinical Neurobiology, University Hospital Wuerzburg, University of Wuerzburg, 97078 Wuerzburg, Germany
| | - Patrick Lüningschrör
- Institute of Clinical Neurobiology, University Hospital Wuerzburg, University of Wuerzburg, 97078 Wuerzburg, Germany
| | - Benjamin Dombert
- Institute of Clinical Neurobiology, University Hospital Wuerzburg, University of Wuerzburg, 97078 Wuerzburg, Germany
| | - Reena Jagdish Rathod
- Institute of Clinical Neurobiology, University Hospital Wuerzburg, University of Wuerzburg, 97078 Wuerzburg, Germany
| | - Daniela C Dieterich
- Institute for Pharmacology and Toxicology, Medical Faculty, University of Magdeburg, 39120 Magdeburg, Germany.,Center for Behavioral Brain Sciences, Medical Faculty, University of Magdeburg, 39120 Magdeburg, Germany
| | - Robert Blum
- Institute of Clinical Neurobiology, University Hospital Wuerzburg, University of Wuerzburg, 97078 Wuerzburg, Germany
| | - Michael Sendtner
- Institute of Clinical Neurobiology, University Hospital Wuerzburg, University of Wuerzburg, 97078 Wuerzburg, Germany
| |
Collapse
|
37
|
Patrinostro X, O'Rourke AR, Chamberlain CM, Moriarity BS, Perrin BJ, Ervasti JM. Relative importance of β cyto- and γ cyto-actin in primary mouse embryonic fibroblasts. Mol Biol Cell 2017; 28:771-782. [PMID: 28077619 PMCID: PMC5349784 DOI: 10.1091/mbc.e16-07-0503] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Revised: 11/29/2016] [Accepted: 01/05/2017] [Indexed: 12/14/2022] Open
Abstract
The highly homologous β (βcyto) and γ (γcyto) cytoplasmic actins are hypothesized to carry out both redundant and unique essential functions, but studies using targeted gene knockout and siRNA-mediated transcript knockdown to examine βcyto- and γcyto-isoform--specific functions in various cell types have yielded conflicting data. Here we quantitatively characterized actin transcript and protein levels, as well as cellular phenotypes, in both gene- and transcript-targeted primary mouse embryonic fibroblasts. We found that the smooth muscle αsm-actin isoform was the dominantly expressed actin isoform in WT primary fibroblasts and was also the most dramatically up-regulated in primary βcyto- or β/γcyto-actin double-knockout fibroblasts. Gene targeting of βcyto-actin, but not γcyto-actin, led to greatly decreased cell proliferation, decreased levels of cellular ATP, and increased serum response factor signaling in primary fibroblasts, whereas immortalization induced by SV40 large T antigen supported fibroblast proliferation in the absence of βcyto-actin. Consistent with in vivo gene-targeting studies in mice, both gene- and transcript-targeting approaches demonstrate that the loss of βcyto-actin protein is more disruptive to primary fibroblast function than is the loss of γcyto-actin.
Collapse
Affiliation(s)
- Xiaobai Patrinostro
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN 55455
| | - Allison R O'Rourke
- Program in Molecular, Cellular, Developmental Biology, and Genetics, University of Minnesota, Minneapolis, MN 55455
| | - Christopher M Chamberlain
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN 55455
| | | | - Benjamin J Perrin
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis, IN 46022
| | - James M Ervasti
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN 55455 .,Program in Molecular, Cellular, Developmental Biology, and Genetics, University of Minnesota, Minneapolis, MN 55455
| |
Collapse
|
38
|
Yates TM, Turner CL, Firth HV, Berg J, Pilz DT. Baraitser-Winter cerebrofrontofacial syndrome. Clin Genet 2016; 92:3-9. [PMID: 27625340 DOI: 10.1111/cge.12864] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2016] [Revised: 09/07/2016] [Accepted: 09/07/2016] [Indexed: 01/31/2023]
Abstract
Baraitser-Winter cerebrofrontofacial syndrome (BWCFF) (BRWS; MIM #243310, 614583) is a rare developmental disorder affecting multiple organ systems. It is characterised by intellectual disability (mild to severe) and distinctive facial appearance (metopic ridging/trigonocephaly, bilateral ptosis, hypertelorism). The additional presence of cortical malformations (pachygyria/lissencephaly) and ocular colobomata are also suggestive of this syndrome. Other features include moderate short stature, contractures, congenital cardiac disease and genitourinary malformations. BWCFF is caused by missense mutations in the cytoplasmic beta- and gamma-actin genes ACTB and ACTG1. We provide an overview of the clinical characteristics (including some novel findings in four recently diagnosed patients), diagnosis, management, mutation spectrum and genetic counselling.
Collapse
Affiliation(s)
- T M Yates
- Department of Medical Genetics, University of Glasgow, Glasgow, UK
| | - C L Turner
- Peninsula Clinical Genetics Service, Royal Devon and Exeter Hospital, Exeter, UK
| | - H V Firth
- Addenbrooke's Hospital, Cambridge University Hospitals, Cambridge, UK
| | - J Berg
- Department of Clinical Genetics, Ninewells Hospital, Dundee, UK
| | - D T Pilz
- West of Scotland Genetics Service, Queen Elizabeth University Hospital, Glasgow, UK
| |
Collapse
|
39
|
Actin Is Crucial for All Kinetically Distinguishable Forms of Endocytosis at Synapses. Neuron 2016; 92:1020-1035. [PMID: 27840001 DOI: 10.1016/j.neuron.2016.10.014] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2014] [Revised: 06/16/2016] [Accepted: 10/04/2016] [Indexed: 01/18/2023]
Abstract
Mechanical force is needed to mediate endocytosis. Whether actin, the most abundant force-generating molecule, is essential for endocytosis is highly controversial in mammalian cells, particularly synapses, likely due to the use of actin blockers, the efficiency and specificity of which are often unclear in the studied cell. Here we addressed this issue using a knockout approach combined with measurements of membrane capacitance and fission pore conductance, imaging of vesicular protein endocytosis, and electron microscopy. We found that two actin isoforms, β- and γ-actin, are crucial for slow, rapid, bulk, and overshoot endocytosis at large calyx-type synapses, and for slow endocytosis and bulk endocytosis at small hippocampal synapses. Polymerized actin provides mechanical force to form endocytic pits. Actin also facilitates replenishment of the readily releasable vesicle pool, likely via endocytic clearance of active zones. We conclude that polymerized actin provides mechanical force essential for all kinetically distinguishable forms of endocytosis at synapses.
Collapse
|
40
|
Musner N, Sidoli M, Zambroni D, Del Carro U, Ungaro D, D'Antonio M, Feltri ML, Wrabetz L. Perk Ablation Ameliorates Myelination in S63del-Charcot-Marie-Tooth 1B Neuropathy. ASN Neuro 2016; 8:8/2/1759091416642351. [PMID: 27095827 PMCID: PMC4844932 DOI: 10.1177/1759091416642351] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2015] [Accepted: 02/18/2016] [Indexed: 12/31/2022] Open
Abstract
In peripheral nerves, P0 glycoprotein accounts for more than 20% of myelin protein content. P0 is synthesized by Schwann cells, processed in the endoplasmic reticulum (ER) and enters the secretory pathway. However, the mutant P0 with S63 deleted (P0S63del) accumulates in the ER lumen and induces a demyelinating neuropathy in Charcot–Marie–Tooth disease type 1B (CMT1B)–S63del mice. Accumulation of P0S63del in the ER triggers a persistent unfolded protein response. Protein kinase RNA-like endoplasmic reticulum kinase (PERK) is an ER stress sensor that phosphorylates eukaryotic initiation factor 2 alpha (eIF2alpha) in order to attenuate protein synthesis. We have shown that increasing phosphophorylated-eIF2alpha (P-eIF2alpha) is a potent therapeutic strategy, improving myelination and motor function in S63del mice. Here, we explore the converse experiment: Perk haploinsufficiency reduces P-eIF2alpha in S63del nerves as expected, but surprisingly, ameliorates, rather than worsens S63del neuropathy. Motor performance and myelin abnormalities improved in S63del//Perk+/− compared with S63del mice. These data suggest that mechanisms other than protein translation might be involved in CMT1B/S63del neuropathy. In addition, Perk deficiency in other cells may contribute to demyelination in a non–Schwann-cell autonomous manner.
Collapse
Affiliation(s)
- Nicolò Musner
- Hunter James Kelly Research Institute, University at Buffalo, NY, USA
| | - Mariapaola Sidoli
- Hunter James Kelly Research Institute, University at Buffalo, NY, USA Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, NY, USA
| | - Desireè Zambroni
- Division of Neuroscience, San Raffaele Scientific Institute, DIBIT, Milan, Italy
| | - Ubaldo Del Carro
- Division of Neuroscience, San Raffaele Scientific Institute, DIBIT, Milan, Italy
| | - Daniela Ungaro
- Division of Neuroscience, San Raffaele Scientific Institute, DIBIT, Milan, Italy
| | - Maurizio D'Antonio
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute, DIBIT, Milan, Italy
| | - Maria L Feltri
- Hunter James Kelly Research Institute, University at Buffalo, NY, USA Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, NY, USA Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, NY, USA
| | - Lawrence Wrabetz
- Hunter James Kelly Research Institute, University at Buffalo, NY, USA Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, NY, USA Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, NY, USA
| |
Collapse
|
41
|
Allen DG, Whitehead NP, Froehner SC. Absence of Dystrophin Disrupts Skeletal Muscle Signaling: Roles of Ca2+, Reactive Oxygen Species, and Nitric Oxide in the Development of Muscular Dystrophy. Physiol Rev 2016; 96:253-305. [PMID: 26676145 DOI: 10.1152/physrev.00007.2015] [Citation(s) in RCA: 309] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Dystrophin is a long rod-shaped protein that connects the subsarcolemmal cytoskeleton to a complex of proteins in the surface membrane (dystrophin protein complex, DPC), with further connections via laminin to other extracellular matrix proteins. Initially considered a structural complex that protected the sarcolemma from mechanical damage, the DPC is now known to serve as a scaffold for numerous signaling proteins. Absence or reduced expression of dystrophin or many of the DPC components cause the muscular dystrophies, a group of inherited diseases in which repeated bouts of muscle damage lead to atrophy and fibrosis, and eventually muscle degeneration. The normal function of dystrophin is poorly defined. In its absence a complex series of changes occur with multiple muscle proteins showing reduced or increased expression or being modified in various ways. In this review, we will consider the various proteins whose expression and function is changed in muscular dystrophies, focusing on Ca(2+)-permeable channels, nitric oxide synthase, NADPH oxidase, and caveolins. Excessive Ca(2+) entry, increased membrane permeability, disordered caveolar function, and increased levels of reactive oxygen species are early changes in the disease, and the hypotheses for these phenomena will be critically considered. The aim of the review is to define the early damage pathways in muscular dystrophy which might be appropriate targets for therapy designed to minimize the muscle degeneration and slow the progression of the disease.
Collapse
Affiliation(s)
- David G Allen
- Sydney Medical School & Bosch Institute, University of Sydney, New South Wales, Australia; and Department of Physiology & Biophysics, University of Washington, Seattle, Washington
| | - Nicholas P Whitehead
- Sydney Medical School & Bosch Institute, University of Sydney, New South Wales, Australia; and Department of Physiology & Biophysics, University of Washington, Seattle, Washington
| | - Stanley C Froehner
- Sydney Medical School & Bosch Institute, University of Sydney, New South Wales, Australia; and Department of Physiology & Biophysics, University of Washington, Seattle, Washington
| |
Collapse
|
42
|
Abstract
Actin is the central building block of the actin cytoskeleton, a highly regulated filamentous network enabling dynamic processes of cells and simultaneously providing structure. Mammals have six actin isoforms that are very conserved and thus share common functions. Tissue-specific expression in part underlies their differential roles, but actin isoforms also coexist in various cell types and tissues, suggesting specific functions and preferential interaction partners. Gene deletion models, antibody-based staining patterns, gene silencing effects, and the occurrence of isoform-specific mutations in certain diseases have provided clues for specificity on the subcellular level and its consequences on the organism level. Yet, the differential actin isoform functions are still far from understood in detail. Biochemical studies on the different isoforms in pure form are just emerging, and investigations in cells have to deal with a complex and regulated system, including compensatory actin isoform expression.
Collapse
Affiliation(s)
- Christophe Ampe
- Department of Biochemistry, Faculty of Medicine and Health Sciences, Ghent University, A. Baertsoenkaai 3, 9000, Ghent, Belgium.
| | - Marleen Van Troys
- Department of Biochemistry, Faculty of Medicine and Health Sciences, Ghent University, A. Baertsoenkaai 3, 9000, Ghent, Belgium
| |
Collapse
|
43
|
Falcone S, Roman W, Hnia K, Gache V, Didier N, Lainé J, Auradé F, Marty I, Nishino I, Charlet-Berguerand N, Romero NB, Marazzi G, Sassoon D, Laporte J, Gomes ER. N-WASP is required for Amphiphysin-2/BIN1-dependent nuclear positioning and triad organization in skeletal muscle and is involved in the pathophysiology of centronuclear myopathy. EMBO Mol Med 2015; 6:1455-75. [PMID: 25262827 PMCID: PMC4237471 DOI: 10.15252/emmm.201404436] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Mutations in amphiphysin-2/BIN1, dynamin 2, and myotubularin are associated with centronuclear myopathy (CNM), a muscle disorder characterized by myofibers with atypical central nuclear positioning and abnormal triads. Mis-splicing of amphiphysin-2/BIN1 is also associated with myotonic dystrophy that shares histopathological hallmarks with CNM. How amphiphysin-2 orchestrates nuclear positioning and triad organization and how CNM-associated mutations lead to muscle dysfunction remains elusive. We find that N-WASP interacts with amphiphysin-2 in myofibers and that this interaction and N-WASP distribution are disrupted by amphiphysin-2 CNM mutations. We establish that N-WASP functions downstream of amphiphysin-2 to drive peripheral nuclear positioning and triad organization during myofiber formation. Peripheral nuclear positioning requires microtubule/Map7/Kif5b-dependent distribution of nuclei along the myofiber and is driven by actin and nesprins. In adult myofibers, N-WASP and amphiphysin-2 are only involved in the maintenance of triad organization but not in the maintenance of peripheral nuclear positioning. Importantly, we confirmed that N-WASP distribution is disrupted in CNM and myotonic dystrophy patients. Our results support a role for N-WASP in amphiphysin-2-dependent nuclear positioning and triad organization and in CNM and myotonic dystrophy pathophysiology.
Collapse
Affiliation(s)
- Sestina Falcone
- Myology Group, UMR S 787 INSERM, Université Pierre et Marie Curie Paris 6, Paris, France Institut de Myologie, Groupe Hospitalier Pitié-Salpêtrière, Paris, France
| | - William Roman
- Myology Group, UMR S 787 INSERM, Université Pierre et Marie Curie Paris 6, Paris, France
| | - Karim Hnia
- IGBMC-CNRS, UMR 7104 INSERM U964, Illkirch, France
| | - Vincent Gache
- Myology Group, UMR S 787 INSERM, Université Pierre et Marie Curie Paris 6, Paris, France Institut de Myologie, Groupe Hospitalier Pitié-Salpêtrière, Paris, France
| | - Nathalie Didier
- Myology Group, UMR S 787 INSERM, Université Pierre et Marie Curie Paris 6, Paris, France
| | - Jeanne Lainé
- Institut de Myologie, Groupe Hospitalier Pitié-Salpêtrière, Paris, France
| | - Frederic Auradé
- Myology Group, UMR S 787 INSERM, Université Pierre et Marie Curie Paris 6, Paris, France
| | - Isabelle Marty
- INSERM U836, Grenoble Institut des Neurosciences, Equipe Muscle et Pathologies, Grenoble, France
| | - Ichizo Nishino
- National Center of Neurology and Psychiatry, Tokyo, Japan
| | | | | | - Giovanna Marazzi
- Myology Group, UMR S 787 INSERM, Université Pierre et Marie Curie Paris 6, Paris, France
| | - David Sassoon
- Myology Group, UMR S 787 INSERM, Université Pierre et Marie Curie Paris 6, Paris, France
| | | | - Edgar R Gomes
- Myology Group, UMR S 787 INSERM, Université Pierre et Marie Curie Paris 6, Paris, France Institut de Myologie, Groupe Hospitalier Pitié-Salpêtrière, Paris, France Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, Lisboa, Portugal
| |
Collapse
|
44
|
WNT/β-Catenin Signaling Regulates Multiple Steps of Myogenesis by Regulating Step-Specific Targets. Mol Cell Biol 2015; 35:1763-76. [PMID: 25755281 DOI: 10.1128/mcb.01180-14] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2014] [Accepted: 02/27/2015] [Indexed: 12/23/2022] Open
Abstract
Molecules involved in WNT/β-catenin signaling show specific spatiotemporal expression and play vital roles in myogenesis; however, it is still largely unknown how WNT/β-catenin signaling regulates each step of myogenesis. Here, we show that WNT/β-catenin signaling can control diverse biological processes of myogenesis by regulating step-specific molecules. In order to identify the temporally specific roles of WNT/β-catenin signaling molecules in muscle development and homeostasis, we used in vitro culture systems for both primary mouse myoblasts and C2C12 cells, which can differentiate into myofibers. We found that a blockade of WNT/β-catenin signaling in the proliferating cells decreases proliferation activity, but does not induce cell death, through the regulation of genes cyclin A2 (Ccna2) and cell division cycle 25C (Cdc25c). During muscle differentiation, the inhibition of WNT/β-catenin signaling blocks myoblast fusion through the inhibition of the Fermitin family homolog 2 (Fermt2) gene. Blocking WNT/β-catenin signaling in the well-differentiated myofibers results in the failure of maintenance of their structure by disruption of cadherin/β-catenin/actin complex formation, which plays a crucial role in connecting a myofiber's cytoskeleton to the surrounding extracellular matrix. Thus, our results indicate that WNT/β-catenin signaling can regulate multiple steps of myogenesis, including cell proliferation, myoblast fusion, and homeostasis, by targeting step-specific molecules.
Collapse
|
45
|
Wei Q, Zhu H, Qian X, Chen Z, Yao J, Lu Y, Cao X, Xing G. Targeted genomic capture and massively parallel sequencing to identify novel variants causing Chinese hereditary hearing loss. J Transl Med 2014; 12:311. [PMID: 25388789 PMCID: PMC4234825 DOI: 10.1186/s12967-014-0311-1] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2014] [Accepted: 10/23/2014] [Indexed: 11/15/2022] Open
Abstract
Background Hereditary hearing loss is genetically heterogeneous, and hundreds of mutations in than 60 genes are involved in this disease. Therefore, it is difficult to identify the causative gene mutations involved. In this study, we combined targeted genomic capture and massively parallel sequencing (MPS) to address this issue. Methods Using targeted genomic capture and MPS, 104 genes and three microRNA regions were selected and simultaneously sequenced in 23 unrelated probands of Chinese families with nonsyndromic hearing loss. The results were validated by Sanger sequencing for all available members of the probands’ families. To analyze the possible pathogenic functional effects of the variants, three types of prediction programs (Mutation Taster, PROVEAN and SIFT) were used. A total of 195 healthy Chinese Han individuals were compared as controls to verify the novel causative mutations. Results Of the 23 probands, six had mutations in DFNA genes [WFS1 (n = 2), COCH, ACTG1, TMC1, and POU4F3] known to cause autosomal dominant nonsyndromic hearing loss. These included one novel in-frame indel mutation, three novel missense mutations and two reported missense mutations. Furthermore, one proband from a family with recessive DFNB carried two monoallelic mutations in the GJB2 and USH2A genes. All of these mutations co-segregated with the hearing loss phenotype in 36 affected individuals from 7 families and were predicted to be pathogenic. Conclusions Mutations in uncommon deafness genes contribute to a portion of nonsyndromic deafness cases. In the future, critical gene mutations may be accurately and quickly identified in families with hereditary hearing loss by targeted genomic capture and MPS. Electronic supplementary material The online version of this article (doi:10.1186/s12967-014-0311-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Qinjun Wei
- Department of Biotechnology, School of Basic Medical Science, Nanjing Medical University, Nanjing, 210029, PR China.
| | - Hongmei Zhu
- Department of Otolaryngology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, PR China.
| | - Xuli Qian
- Department of Biotechnology, School of Basic Medical Science, Nanjing Medical University, Nanjing, 210029, PR China.
| | - Zhibin Chen
- Department of Otolaryngology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, PR China.
| | - Jun Yao
- Department of Biotechnology, School of Basic Medical Science, Nanjing Medical University, Nanjing, 210029, PR China.
| | - Yajie Lu
- Department of Biotechnology, School of Basic Medical Science, Nanjing Medical University, Nanjing, 210029, PR China.
| | - Xin Cao
- Department of Biotechnology, School of Basic Medical Science, Nanjing Medical University, Nanjing, 210029, PR China.
| | - Guangqian Xing
- Department of Otolaryngology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, PR China.
| |
Collapse
|
46
|
Baraitser-Winter cerebrofrontofacial syndrome: delineation of the spectrum in 42 cases. Eur J Hum Genet 2014; 23:292-301. [PMID: 25052316 DOI: 10.1038/ejhg.2014.95] [Citation(s) in RCA: 103] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2013] [Revised: 04/05/2014] [Accepted: 04/30/2014] [Indexed: 11/08/2022] Open
Abstract
Baraitser-Winter, Fryns-Aftimos and cerebrofrontofacial syndrome types 1 and 3 have recently been associated with heterozygous gain-of-function mutations in one of the two ubiquitous cytoplasmic actin-encoding genes ACTB and ACTG1 that encode β- and γ-actins. We present detailed phenotypic descriptions and neuroimaging on 36 patients analyzed by our group and six cases from the literature with a molecularly proven actinopathy (9 ACTG1 and 33 ACTB). The major clinical anomalies are striking dysmorphic facial features with hypertelorism, broad nose with large tip and prominent root, congenital non-myopathic ptosis, ridged metopic suture and arched eyebrows. Iris or retinal coloboma is present in many cases, as is sensorineural deafness. Cleft lip and palate, hallux duplex, congenital heart defects and renal tract anomalies are seen in some cases. Microcephaly may develop with time. Nearly all patients with ACTG1 mutations, and around 60% of those with ACTB mutations have some degree of pachygyria with anteroposterior severity gradient, rarely lissencephaly or neuronal heterotopia. Reduction of shoulder girdle muscle bulk and progressive joint stiffness is common. Early muscular involvement, occasionally with congenital arthrogryposis, may be present. Progressive, severe dystonia was seen in one family. Intellectual disability and epilepsy are variable in severity and largely correlate with CNS anomalies. One patient developed acute lymphocytic leukemia, and another a cutaneous lymphoma, indicating that actinopathies may be cancer-predisposing disorders. Considering the multifaceted role of actins in cell physiology, we hypothesize that some clinical manifestations may be partially mutation specific. Baraitser-Winter cerebrofrontofacial syndrome is our suggested designation for this clinical entity.
Collapse
|
47
|
Vassilopoulos S, Gentil C, Lainé J, Buclez PO, Franck A, Ferry A, Précigout G, Roth R, Heuser JE, Brodsky FM, Garcia L, Bonne G, Voit T, Piétri-Rouxel F, Bitoun M. Actin scaffolding by clathrin heavy chain is required for skeletal muscle sarcomere organization. ACTA ACUST UNITED AC 2014; 205:377-93. [PMID: 24798732 PMCID: PMC4018784 DOI: 10.1083/jcb.201309096] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Clathrin heavy chain contributes to the formation and maintenance of the contractile apparatus in skeletal muscle through interactions with costameric proteins. The ubiquitous clathrin heavy chain (CHC), the main component of clathrin-coated vesicles, is well characterized for its role in intracellular membrane traffic and endocytosis from the plasma membrane (PM). Here, we demonstrate that in skeletal muscle CHC regulates the formation and maintenance of PM–sarcomere attachment sites also known as costameres. We show that clathrin forms large coated lattices associated with actin filaments and the muscle-specific isoform of α-actinin at the PM of differentiated myotubes. Depletion of CHC in myotubes induced a loss of actin and α-actinin sarcomeric organization, whereas CHC depletion in vivo induced a loss of contractile force due to the detachment of sarcomeres from the PM. Our results suggest that CHC contributes to the formation and maintenance of the contractile apparatus through interactions with costameric proteins and highlight an unconventional role for CHC in skeletal muscle that may be relevant to pathophysiology of neuromuscular disorders.
Collapse
Affiliation(s)
- Stéphane Vassilopoulos
- Institut National de la Santé et de la Recherche Médicale (INSERM) U974, 2 Centre National de la Recherche Scientifique (CNRS) UMR 7215, and 3 Université Pierre et Marie Curie-Paris 6, UM 76, Paris F-75013, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Rubenstein PA, Wen KK. Insights into the effects of disease-causing mutations in human actins. Cytoskeleton (Hoboken) 2014; 71:211-29. [PMID: 24574087 DOI: 10.1002/cm.21169] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2013] [Revised: 02/13/2013] [Accepted: 02/19/2014] [Indexed: 01/04/2023]
Abstract
Mutations in all six actins in humans have now been shown to cause diseases. However, a number of factors have made it difficult to gain insight into how the changes in actin functions brought about by these pathogenic mutations result in the disease phenotype. These include the presence of multiple actins in the same cell, limited accessibility to pure mutant material, and complexities associated with the structures and their component cells that manifest the diseases. To try to circumvent these difficulties, investigators have turned to the use of model systems. This review describes these various approaches, the initial results obtained using them, and the insight they have provided into allosteric mechanisms that govern actin function. Although results so far have not explained a particular disease phenotype at the molecular level, they have provided valuable insight into actin function at the mechanistic level which can be utilized in the future to delineate the molecular bases of these different actinopathies.
Collapse
Affiliation(s)
- Peter A Rubenstein
- Department of Biochemistry, University of Iowa Carver College of Medicine, Iowa City, Iowa
| | | |
Collapse
|
49
|
Huang Z, Fang Z, Zhen H, Zhou L, Amin HM, Shi P. Inhibition of type I insulin-like growth factor receptor tyrosine kinase by picropodophyllin induces apoptosis and cell cycle arrest in T lymphoblastic leukemia/lymphoma. Leuk Lymphoma 2014; 55:1876-83. [PMID: 24206093 DOI: 10.3109/10428194.2013.862241] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
It has been recently shown that the type I insulin-like growth factor receptor (IGF-IR) contributes significantly to the survival of T lymphoblastic leukemia/lymphoma (T-LBL) cells, and it was therefore suggested that IGF-IR could represent a legitimate therapeutic target in this aggressive disease. Picropodophyllin (PPP) is a potent, selective inhibitor of IGF-IR that is currently used with notable success in clinical trials that include patients with aggressive types of epithelial tumors. In the present study, we tested the effects of PPP on Jurkat and Molt-3 cells; two prototype T-LBL cell lines. Our results demonstrate that PPP efficiently induced apoptotic cell death and cell cycle arrest of these two cells. These effects were attributable to alterations of downstream target proteins. By using proteomic analysis, seven different proteins were found to be affected by PPP treatment of Jurkat cells. These proteins are involved in various aspects of cellular metabolism, cytoskeleton organization and signal transduction pathways. The results suggest that PPP affects multiple signaling molecules and inhibits fundamental pathways that control cell growth and survival. Our study also provides novel evidence that PPP could be potentially utilized for the treatment of aggressive T-LBL.
Collapse
Affiliation(s)
- Zhiwei Huang
- College of Chemistry, Chemical Engineering and Biotechnology, Donghua University , Shanghai , China
| | | | | | | | | | | |
Collapse
|
50
|
Luo Y, Kong F, Wang Z, Chen D, Liu Q, Wang T, Xu R, Wang X, Yang JY. Loss of ASAP3 destabilizes cytoskeletal protein ACTG1 to suppress cancer cell migration. Mol Med Rep 2013; 9:387-94. [PMID: 24284654 DOI: 10.3892/mmr.2013.1831] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2013] [Accepted: 10/31/2013] [Indexed: 11/06/2022] Open
Abstract
ArfGAP with SH3 domain, ankyrin repeat and PH domain 3 (ASAP3), previously known as ACAP4, DDEFL1 and UPLC1, is considered to be an important regulator in cancer cell migration/invasion and actin-based cytoskeletal remodeling. However, the underlying mechanisms through which ASAP3 mediates these processes are not well-elucidated. This study reported that in certain types of cancer cells, loss of ASAP3 suppressed cell migration/invasion, in part by destabilizing γ-actin-1 (ACTG1), a cytoskeletal protein considered to be an integral component of the cell migratory machinery, essential for the rearrangement of the dynamic cytoskeletal networks and important in diseases, such as brain malformation, hearing loss and cancer development. The data, for the first time, link ASAP3 with ACTG1 in the regulation of cytoskeletal maintenance and cell motility.
Collapse
Affiliation(s)
- Yu Luo
- School of Nursing, The Third Military Medical University, Chongqing 400038, P.R. China
| | - Fang Kong
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Biology, School of Life Sciences, Xiamen University, Xiang'an, Xiamen, Fujian 361102, P.R. China
| | - Zhen Wang
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Biology, School of Life Sciences, Xiamen University, Xiang'an, Xiamen, Fujian 361102, P.R. China
| | - Dahan Chen
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Biology, School of Life Sciences, Xiamen University, Xiang'an, Xiamen, Fujian 361102, P.R. China
| | - Qiuyan Liu
- School of Biomedical Sciences, Huaqiao University, Quanzhou, Fujian 362021, P.R. China
| | - Tao Wang
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Biology, School of Life Sciences, Xiamen University, Xiang'an, Xiamen, Fujian 361102, P.R. China
| | - Ruian Xu
- School of Biomedical Sciences, Huaqiao University, Quanzhou, Fujian 362021, P.R. China
| | - Xianyuan Wang
- School of Nursing, The Third Military Medical University, Chongqing 400038, P.R. China
| | - James Y Yang
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Biology, School of Life Sciences, Xiamen University, Xiang'an, Xiamen, Fujian 361102, P.R. China
| |
Collapse
|