101
|
Avila-Smirnow D, Gueneau L, Batonnet-Pichon S, Delort F, Bécane HM, Claeys K, Beuvin M, Goudeau B, Jais JP, Nelson I, Richard P, Ben Yaou R, Romero NB, Wahbi K, Mathis S, Voit T, Furst D, van der Ven P, Gil R, Vicart P, Fardeau M, Bonne G, Behin A. Cardiac arrhythmia and late-onset muscle weakness caused by a myofibrillar myopathy with unusual histopathological features due to a novel missense mutation in FLNC. Rev Neurol (Paris) 2016; 172:594-606. [PMID: 27633507 DOI: 10.1016/j.neurol.2016.07.017] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2016] [Revised: 07/16/2016] [Accepted: 07/26/2016] [Indexed: 11/29/2022]
Abstract
Myofibrillar myopathies (MFM) are mostly adult-onset diseases characterized by progressive morphological alterations of the muscle fibers beginning in the Z-disk and the presence of protein aggregates in the sarcoplasm. They are mostly caused by mutations in different genes that encode Z-disk proteins, including DES, CRYAB, LDB3, MYOT, FLNC and BAG3. A large family of French origin, presenting an autosomal dominant pattern, characterized by cardiac arrhythmia associated to late-onset muscle weakness, was evaluated to clarify clinical, morphological and genetic diagnosis. Muscle weakness began during adult life (over 30 years of age), and had a proximal distribution. Histology showed clear signs of a myofibrillar myopathy, but with unusual, large inclusions. Subsequently, genetic testing was performed in MFM genes available for screening at the time of clinical/histological diagnosis, and desmin (DES), αB-crystallin (CRYAB), myotilin (MYOT) and ZASP (LDB3), were excluded. LMNA gene screening found the p.R296C variant which did not co-segregate with the disease. Genome wide scan revealed linkage to 7q.32, containing the FLNC gene. FLNC direct sequencing revealed a heterozygous c.3646T>A p.Tyr1216Asn change, co-segregating with the disease, in a highly conserved amino acid of the protein. Normal filamin C levels were detected by Western-blot analysis in patient muscle biopsies and expression of the mutant protein in NIH3T3 showed filamin C aggregates. This is an original FLNC mutation in a MFM family with an atypical clinical and histopathological presentation, given the presence of significantly focal lesions and prominent sarcoplasmic masses in muscle biopsies and the constant heart involvement preceding significantly the onset of the myopathy. Though a rare etiology, FLNC gene should not be excluded in early-onset arrhythmia, even in the absence of myopathy, which occurs later in the disease course.
Collapse
Affiliation(s)
- D Avila-Smirnow
- Sorbonne universités, UPMC Paris 06, center of research in myology, Inserm UMRS974, CNRS FRE3617, 75013 Paris, France
| | - L Gueneau
- Sorbonne universités, UPMC Paris 06, center of research in myology, Inserm UMRS974, CNRS FRE3617, 75013 Paris, France
| | - S Batonnet-Pichon
- Sorbonne Paris Cité, université Paris Diderot, CNRS, unité de biologie fonctionnelle et adaptative, UMR 8251, 75013 Paris, France
| | - F Delort
- Sorbonne Paris Cité, université Paris Diderot, CNRS, unité de biologie fonctionnelle et adaptative, UMR 8251, 75013 Paris, France
| | - H-M Bécane
- AP-HP, groupe hospitalier Pitié-Salpêtrière, institut de myologie, centre de référence de pathologie neuromusculaire Paris-Est, 47-83, boulevard de l'Hôpital, 75013 Paris, France
| | - K Claeys
- Groupe hospitalier Pitié-Salpêtrière, association institut de myologie, unité de morphologie neuromusculaire, 75013 Paris, France
| | - M Beuvin
- Sorbonne universités, UPMC Paris 06, center of research in myology, Inserm UMRS974, CNRS FRE3617, 75013 Paris, France
| | - B Goudeau
- Sorbonne universités, UPMC Paris 06, center of research in myology, Inserm UMRS974, CNRS FRE3617, 75013 Paris, France
| | - J-P Jais
- GH Necker Enfants-Malades, université Paris Descartes, faculté de médecine, biostatistique et informatique médicale, EA 4067, 75015 Paris, France
| | - I Nelson
- Sorbonne universités, UPMC Paris 06, center of research in myology, Inserm UMRS974, CNRS FRE3617, 75013 Paris, France
| | - P Richard
- AP-HP, groupe hospitalier Pitié-Salpêtrière, service de biochimie métabolique, U.F. cardiogénétique et myogénétique, 75013 Paris, France
| | - R Ben Yaou
- Sorbonne universités, UPMC Paris 06, center of research in myology, Inserm UMRS974, CNRS FRE3617, 75013 Paris, France; AP-HP, groupe hospitalier Pitié-Salpêtrière, institut de myologie, centre de référence de pathologie neuromusculaire Paris-Est, 47-83, boulevard de l'Hôpital, 75013 Paris, France
| | - N B Romero
- Sorbonne universités, UPMC Paris 06, center of research in myology, Inserm UMRS974, CNRS FRE3617, 75013 Paris, France; Groupe hospitalier Pitié-Salpêtrière, association institut de myologie, unité de morphologie neuromusculaire, 75013 Paris, France
| | - K Wahbi
- Sorbonne universités, UPMC Paris 06, center of research in myology, Inserm UMRS974, CNRS FRE3617, 75013 Paris, France; AP-HP, groupe hospitalier Pitié-Salpêtrière, institut de myologie, centre de référence de pathologie neuromusculaire Paris-Est, 47-83, boulevard de l'Hôpital, 75013 Paris, France; AP-HP, groupe hospitalier Cochin-Broca-Hôtel Dieu, service de cardiologie, 75013 Paris, France
| | - S Mathis
- CHU de la Milétrie, service de neurologie, 86021 Poitiers, France
| | - T Voit
- Sorbonne universités, UPMC Paris 06, center of research in myology, Inserm UMRS974, CNRS FRE3617, 75013 Paris, France; AP-HP, groupe hospitalier Pitié-Salpêtrière, institut de myologie, centre de référence de pathologie neuromusculaire Paris-Est, 47-83, boulevard de l'Hôpital, 75013 Paris, France
| | - D Furst
- University of Bonn, institute for cell biology, department of molecular cell biology, Bonn, Germany
| | - P van der Ven
- University of Bonn, institute for cell biology, department of molecular cell biology, Bonn, Germany
| | - R Gil
- CHU de la Milétrie, service de neurologie, 86021 Poitiers, France
| | - P Vicart
- Sorbonne Paris Cité, université Paris Diderot, CNRS, unité de biologie fonctionnelle et adaptative, UMR 8251, 75013 Paris, France
| | - M Fardeau
- Groupe hospitalier Pitié-Salpêtrière, association institut de myologie, unité de morphologie neuromusculaire, 75013 Paris, France
| | - G Bonne
- Sorbonne universités, UPMC Paris 06, center of research in myology, Inserm UMRS974, CNRS FRE3617, 75013 Paris, France
| | - A Behin
- AP-HP, groupe hospitalier Pitié-Salpêtrière, institut de myologie, centre de référence de pathologie neuromusculaire Paris-Est, 47-83, boulevard de l'Hôpital, 75013 Paris, France.
| |
Collapse
|
102
|
Begay RL, Tharp CA, Martin A, Graw SL, Sinagra G, Miani D, Sweet ME, Slavov DB, Stafford N, Zeller MJ, Alnefaie R, Rowland TJ, Brun F, Jones KL, Gowan K, Mestroni L, Garrity DM, Taylor MRG. FLNC Gene Splice Mutations Cause Dilated Cardiomyopathy. JACC Basic Transl Sci 2016; 1:344-359. [PMID: 28008423 PMCID: PMC5166708 DOI: 10.1016/j.jacbts.2016.05.004] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
A genetic etiology has been identified in 30% to 40% of dilated cardiomyopathy (DCM) patients, yet only 50% of these cases are associated with a known causative gene variant. Thus, in order to understand the pathophysiology of DCM, it is necessary to identify and characterize additional genes. In this study, whole exome sequencing in combination with segregation analysis was used to identify mutations in a novel gene, filamin C (FLNC), resulting in a cardiac-restricted DCM pathology. Here we provide functional data via zebrafish studies and protein analysis to support a model implicating FLNC haploinsufficiency as a mechanism of DCM. Deoxyribonucleic acid obtained from 2 large DCM families was studied using whole-exome sequencing and cosegregation analysis resulting in the identification of a novel disease gene, FLNC. The 2 families, from the same Italian region, harbored the same FLNC splice-site mutation (FLNC c.7251+1G>A). A third U.S. family was then identified with a novel FLNC splice-site mutation (FLNC c.5669-1delG) that leads to haploinsufficiency as shown by the FLNC Western blot analysis of the heart muscle. The FLNC ortholog flncb morpholino was injected into zebrafish embryos, and when flncb was knocked down caused a cardiac dysfunction phenotype. On electron microscopy, the flncb morpholino knockdown zebrafish heart showed defects within the Z-discs and sarcomere disorganization.
Collapse
Affiliation(s)
- Rene L Begay
- Cardiovascular Institute and Adult Medical Genetics Program, University of Colorado Denver, Aurora, CO
| | - Charles A Tharp
- Cardiovascular Institute and Adult Medical Genetics Program, University of Colorado Denver, Aurora, CO
| | - August Martin
- Center for Cardiovascular Research and Department of Biology, Colorado State University, Fort Collins, CO
| | - Sharon L Graw
- Cardiovascular Institute and Adult Medical Genetics Program, University of Colorado Denver, Aurora, CO
| | - Gianfranco Sinagra
- Cardiovascular Department, Ospedali Riuniti and University of Trieste, Trieste, Italy
| | - Daniela Miani
- Department of Cardiothoracic Science, University Hospital S. Maria della Misericordia, Udine, Italy
| | - Mary E Sweet
- Cardiovascular Institute and Adult Medical Genetics Program, University of Colorado Denver, Aurora, CO
| | - Dobromir B Slavov
- Cardiovascular Institute and Adult Medical Genetics Program, University of Colorado Denver, Aurora, CO
| | - Neil Stafford
- Center for Cardiovascular Research and Department of Biology, Colorado State University, Fort Collins, CO; Cardiovascular and Biofluid Mechanics Laboratory, Colorado State University, Fort Collins, CO
| | - Molly J Zeller
- Center for Cardiovascular Research and Department of Biology, Colorado State University, Fort Collins, CO
| | - Rasha Alnefaie
- Center for Cardiovascular Research and Department of Biology, Colorado State University, Fort Collins, CO
| | - Teisha J Rowland
- Cardiovascular Institute and Adult Medical Genetics Program, University of Colorado Denver, Aurora, CO
| | - Francesca Brun
- Cardiovascular Department, Ospedali Riuniti and University of Trieste, Trieste, Italy
| | - Kenneth L Jones
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver, Aurora, CO
| | - Katherine Gowan
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver, Aurora, CO
| | - Luisa Mestroni
- Cardiovascular Institute and Adult Medical Genetics Program, University of Colorado Denver, Aurora, CO
| | - Deborah M Garrity
- Center for Cardiovascular Research and Department of Biology, Colorado State University, Fort Collins, CO
| | - Matthew R G Taylor
- Cardiovascular Institute and Adult Medical Genetics Program, University of Colorado Denver, Aurora, CO
| |
Collapse
|
103
|
Three novel missense mutations in the filamin B gene are associated with isolated congenital talipes equinovarus. Hum Genet 2016; 135:1181-9. [DOI: 10.1007/s00439-016-1701-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Accepted: 06/21/2016] [Indexed: 12/30/2022]
|
104
|
Abstract
Rho family GTPases such as Cdc42 are key regulators of essential cellular processes through their effects on cytoskeletal dynamics, signaling and gene expression. Rho GTPases modulate these functions by engaging a wide variety of downstream effectors. Among these effectors is the largely understudied Cdc42EP/BORG family of Cdc42 effectors. BORG proteins have been linked to actin and septin regulation, but their role in development and disease is only starting to emerge. Recently, Cdc42EP3/BORG2 was shown to coordinate actin and septin cytoskeleton rearrangements in cancer-associated fibroblasts (CAFs). Interestingly, Cdc42EP3 expression potentiated cellular responses to mechanical stimulation leading to signaling and transcriptional adaptations required for the emergence of a fully activated CAF phenotype. These findings uncover a novel role for the BORG/septin network in cancer. Here, we demonstrate that Cdc42EP3 function in CAFs relies on tight regulation by Cdc42.
Collapse
Affiliation(s)
- Aaron J Farrugia
- a Tumour Microenvironment Team, Division of Cancer Biology , Institute of Cancer Research , London , UK
| | - Fernando Calvo
- a Tumour Microenvironment Team, Division of Cancer Biology , Institute of Cancer Research , London , UK
| |
Collapse
|
105
|
Leber Y, Ruparelia AA, Kirfel G, van der Ven PFM, Hoffmann B, Merkel R, Bryson-Richardson RJ, Fürst DO. Filamin C is a highly dynamic protein associated with fast repair of myofibrillar microdamage. Hum Mol Genet 2016; 25:2776-2788. [PMID: 27206985 DOI: 10.1093/hmg/ddw135] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Revised: 04/26/2016] [Accepted: 04/27/2016] [Indexed: 11/12/2022] Open
Abstract
Filamin c (FLNc) is a large dimeric actin-binding protein located at premyofibrils, myofibrillar Z-discs and myofibrillar attachment sites of striated muscle cells, where it is involved in mechanical stabilization, mechanosensation and intracellular signaling. Mutations in the gene encoding FLNc give rise to skeletal muscle diseases and cardiomyopathies. Here, we demonstrate by fluorescence recovery after photobleaching that a large fraction of FLNc is highly mobile in cultured neonatal mouse cardiomyocytes and in cardiac and skeletal muscles of live transgenic zebrafish embryos. Analysis of cardiomyocytes from Xirp1 and Xirp2 deficient animals indicates that both Xin actin-binding repeat-containing proteins stabilize FLNc selectively in premyofibrils. Using a novel assay to analyze myofibrillar microdamage and subsequent repair in cultured contracting cardiomyocytes by live cell imaging, we demonstrate that repair of damaged myofibrils is achieved within only 4 h, even in the absence of de novo protein synthesis. FLNc is immediately recruited to these sarcomeric lesions together with its binding partner aciculin and precedes detectable assembly of filamentous actin and recruitment of other myofibrillar proteins. These data disclose an unprecedented degree of flexibility of the almost crystalline contractile machinery and imply FLNc as a dynamic signaling hub, rather than a primarily structural protein. Our myofibrillar damage/repair model illustrates how (cardio)myocytes are kept functional in their mechanically and metabolically strained environment. Our results help to better understand the pathomechanisms and pathophysiology of early stages of FLNc-related myofibrillar myopathy and skeletal and cardiac diseases preceding pathological protein aggregation.
Collapse
Affiliation(s)
- Yvonne Leber
- Department of Molecular Cell Biology, Institute for Cell Biology, University of Bonn, D53121 Bonn, Germany
| | - Avnika A Ruparelia
- School of Biological Sciences, Monash University, Melbourne, Victoria 3800, Australia
| | - Gregor Kirfel
- Department of Molecular Cell Biology, Institute for Cell Biology, University of Bonn, D53121 Bonn, Germany
| | - Peter F M van der Ven
- Department of Molecular Cell Biology, Institute for Cell Biology, University of Bonn, D53121 Bonn, Germany
| | - Bernd Hoffmann
- Department of Biomechanics (ICS-7), Institute of Complex Systems, Forschungszentrum Jülich, D52428 Jülich, Germany and
| | - Rudolf Merkel
- Department of Biomechanics (ICS-7), Institute of Complex Systems, Forschungszentrum Jülich, D52428 Jülich, Germany and.,Department of Biomechanics, Institute for Physical and Theoretical Chemistry, University of Bonn, D53115 Bonn, Germany
| | | | - Dieter O Fürst
- Department of Molecular Cell Biology, Institute for Cell Biology, University of Bonn, D53121 Bonn, Germany
| |
Collapse
|
106
|
Zheng M, Zhang X, Sun N, Min C, Zhang X, Kim KM. RalA employs GRK2 and β-arrestins for the filamin A-mediated regulation of trafficking and signaling of dopamine D2 and D3 receptor. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2016; 1863:2072-83. [PMID: 27188791 DOI: 10.1016/j.bbamcr.2016.05.010] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Revised: 05/10/2016] [Accepted: 05/13/2016] [Indexed: 12/13/2022]
Abstract
Filamin A (FLNA) is known to act as platform for the signaling and intracellular trafficking of various GPCRs including dopamine D2 and D3 receptors (D2R, D3R). To understand molecular mechanisms involved in the FLNA-mediated regulation of D2R and D3R, comparative studies were conducted on the signaling and intracellular trafficking of the D2R and D3R in FLNA-knockdown cells, with a specific focus on the roles of the proteins that interact with FLNA and the D2R and D3R. Lowering the level of cellular FLNA caused an elevation in RalA activity and resulted in selective interference with the normal intracellular trafficking and signaling of the D2R and D3R, through GRK2 and β-arrestins, respectively. Knockdown of FLNA or coexpression of active RalA interfered with the recycling of the internalized D2R and resulted in the development of receptor tolerance. Active RalA was found to interact with GRK2 to sequester it from D2R. Knockdown of FLNA or coexpression of active RalA prevented D3R from coupling with G protein. The selective involvement of GRK2- and β-arrestins in the RalA-mediated cellular processes of the D2R and D3R was achieved via their different modes of interactions with the receptor and their distinct functional roles in receptor regulation. Our results show that FLNA is a multi-functional protein that acts as a platform on which D2R and D3R can interact with various proteins, through which selective regulation of these receptors occurs in combination with GRK2 and β-arrestins.
Collapse
Affiliation(s)
- Mei Zheng
- Department of Pharmacology, College of Pharmacy, Drug Development Research Institute, Chonnam National University, Gwang-Ju 500-757, Republic of Korea
| | - Xiaohan Zhang
- Department of Pharmacology, College of Pharmacy, Drug Development Research Institute, Chonnam National University, Gwang-Ju 500-757, Republic of Korea
| | - NingNing Sun
- Department of Pharmacology, College of Pharmacy, Drug Development Research Institute, Chonnam National University, Gwang-Ju 500-757, Republic of Korea
| | - Chengchun Min
- Department of Pharmacology, College of Pharmacy, Drug Development Research Institute, Chonnam National University, Gwang-Ju 500-757, Republic of Korea
| | - Xiaowei Zhang
- Department of Pharmacology, College of Pharmacy, Drug Development Research Institute, Chonnam National University, Gwang-Ju 500-757, Republic of Korea
| | - Kyeong-Man Kim
- Department of Pharmacology, College of Pharmacy, Drug Development Research Institute, Chonnam National University, Gwang-Ju 500-757, Republic of Korea.
| |
Collapse
|
107
|
Time course of lead induced proteomic changes in gill of the Antarctic limpet Nacella Concinna (Gastropoda: Patellidae). J Proteomics 2016; 151:145-161. [PMID: 27126604 DOI: 10.1016/j.jprot.2016.04.036] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Revised: 03/06/2016] [Accepted: 04/22/2016] [Indexed: 12/12/2022]
Abstract
The effect of increasing levels of metals from anthropogenic sources on Antarctic invertebrates is poorly understood. Here we exposed limpets (Nacella concinna) to 0, 0.12 and 0.25 μg L− 1 lead for 12, 24, 48 and 168 h. We subsequently quantified the changes in protein abundance from gill, using 2D gel electrophoresis and mass spectrometry. We identified several antioxidant proteins, including the metal binding Mn-superoxide dismutase and ferritin, increasing abundances early on. Chaperones involved in the redox-dependent maturation of proteins in the endoplasmic reticulum (ER) showed higher abundance with lead at 48 h. Lead also increased the abundance of Zn-binding carbonic anhydrase at 12 h, suggesting a challenge to acid-base balance. Metabolic proteins increased abundance at 168 h, suggesting a greater ATP demand during prolonged exposure. Changes in abundance of the small G-protein cdc42, a signaling protein modifying cytoskeleton, increased early and subsequently reversed during prolonged exposure, possibly leading to the modification of thick filament structure and function. We hypothesize that the replacement of metals initially affected antioxidant proteins and increased the production of reactive oxygen species. This disrupted the redox-sensitive maturation of proteins in the ER and caused increased ATP demand later on, accompanied by changes in cytoskeleton. SIGNIFICANCE Proteomic analysis of gill tissue in Antarctic limpets exposed to different concentrations of lead (Pb) over a 168 h time period showed that proteomic changes vary with time. These changes included an increase in the demand of scavenging reactive oxygen species, acid-base balance and a challenge to protein homeostasis in the endoplasmic reticulum early on and subsequently an increase in energy metabolism, cellular signaling, and cytoskeletal modifications. Based on this time course, we hypothesize that the main mode of action of lead is a replacement of metal-cofactors of key enzymes involved in the scavenging of reactive oxygen species and the regulation of acid-base balance.
Collapse
|
108
|
Ruparelia AA, Oorschot V, Ramm G, Bryson-Richardson RJ. FLNC myofibrillar myopathy results from impaired autophagy and protein insufficiency. Hum Mol Genet 2016; 25:2131-2142. [PMID: 26969713 DOI: 10.1093/hmg/ddw080] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Accepted: 03/07/2016] [Indexed: 01/23/2023] Open
Abstract
Myofibrillar myopathy is a progressive muscle disease characterized by the disintegration of muscle fibers and formation of protein aggregates. Causative mutations have been identified in nine genes encoding Z-disk proteins, including the actin binding protein filamin C (FLNC). To investigate the mechanism of disease in FLNCW2710X myopathy we overexpressed fluorescently tagged FLNC or FLNCW2710X in zebrafish. Expression of FLNCW2710X causes formation of protein aggregates but surprisingly, our studies reveal that the mutant protein localizes correctly to the Z-disk and is capable of rescuing the fiber disintegration phenotype that results from FLNC knockdown. This demonstrates that the functions necessary for muscle integrity are not impaired, and suggests that it is the formation of protein aggregates and subsequent sequestration of FLNC away from the Z-disk that results in myofibrillar disintegration. Similar to those found in patients, the aggregates in FLNCW2710X expressing fish contain the co-chaperone BAG3. FLNC is a target of the BAG3-mediated chaperone assisted selective autophagy (CASA) pathway and therefore we investigated its role, and the role of autophagy in general, in clearing protein aggregates. We reveal that despite BAG3 recruitment to the aggregates they are not degraded via CASA. Additionally, recruitment of BAG3 is sufficient to block alternative autophagy pathways which would otherwise clear the aggregates. This blockage can be relieved by reducing BAG3 levels or by stimulating autophagy. This study therefore identifies both BAG3 reduction and autophagy promotion as potential therapies for FLNCW2710X myofibrillar myopathy, and identifies protein insufficiency due to sequestration, compounded by impaired autophagy, as the cause.
Collapse
Affiliation(s)
| | - Viola Oorschot
- The Clive and Vera Ramaciotti Centre for Structural Cryo-Electron Microscopy and and
| | - Georg Ramm
- The Clive and Vera Ramaciotti Centre for Structural Cryo-Electron Microscopy and and Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC 3800, Australia
| | | |
Collapse
|
109
|
Segura I, Lange C, Knevels E, Moskalyuk A, Pulizzi R, Eelen G, Chaze T, Tudor C, Boulegue C, Holt M, Daelemans D, Matondo M, Ghesquière B, Giugliano M, Ruiz de Almodovar C, Dewerchin M, Carmeliet P. The Oxygen Sensor PHD2 Controls Dendritic Spines and Synapses via Modification of Filamin A. Cell Rep 2016; 14:2653-67. [PMID: 26972007 PMCID: PMC4805856 DOI: 10.1016/j.celrep.2016.02.047] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Revised: 12/21/2015] [Accepted: 02/05/2016] [Indexed: 01/09/2023] Open
Abstract
Neuronal function is highly sensitive to changes in oxygen levels, but how hypoxia affects dendritic spine formation and synaptogenesis is unknown. Here we report that hypoxia, chemical inhibition of the oxygen-sensing prolyl hydroxylase domain proteins (PHDs), and silencing of Phd2 induce immature filopodium-like dendritic protrusions, promote spine regression, reduce synaptic density, and decrease the frequency of spontaneous action potentials independently of HIF signaling. We identified the actin cross-linker filamin A (FLNA) as a target of PHD2 mediating these effects. In normoxia, PHD2 hydroxylates the proline residues P2309 and P2316 in FLNA, leading to von Hippel-Lindau (VHL)-mediated ubiquitination and proteasomal degradation. In hypoxia, PHD2 inactivation rapidly upregulates FLNA protein levels because of blockage of its proteasomal degradation. FLNA upregulation induces more immature spines, whereas Flna silencing rescues the immature spine phenotype induced by PHD2 inhibition. The oxygen sensor PHD2 is present in dendritic spines PHD2 inhibition by hypoxia reduces spine maturation, synaptic density, and activity Through hydroxylation, PHD2 targets filamin A for proteasomal degradation Filamin A stabilization promotes dendritic spine remodeling
Collapse
Affiliation(s)
- Inmaculada Segura
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, KU Leuven, 3000 Leuven, Belgium; Laboratory of Angiogenesis and Vascular Metabolism, Vesalius Research Center, VIB, 3000 Leuven, Belgium
| | - Christian Lange
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, KU Leuven, 3000 Leuven, Belgium; Laboratory of Angiogenesis and Vascular Metabolism, Vesalius Research Center, VIB, 3000 Leuven, Belgium
| | - Ellen Knevels
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, KU Leuven, 3000 Leuven, Belgium; Laboratory of Angiogenesis and Vascular Metabolism, Vesalius Research Center, VIB, 3000 Leuven, Belgium
| | - Anastasiya Moskalyuk
- Laboratory of Theoretical Neurobiology and Neuroengineering, University of Antwerp, 2610 Wilrijk, Belgium
| | - Rocco Pulizzi
- Laboratory of Theoretical Neurobiology and Neuroengineering, University of Antwerp, 2610 Wilrijk, Belgium
| | - Guy Eelen
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, KU Leuven, 3000 Leuven, Belgium; Laboratory of Angiogenesis and Vascular Metabolism, Vesalius Research Center, VIB, 3000 Leuven, Belgium
| | - Thibault Chaze
- Proteomics Platform, Institute Pasteur, 75015 Paris, France
| | | | - Cyril Boulegue
- Proteomics Platform, Institute Pasteur, 75015 Paris, France
| | - Matthew Holt
- Laboratory of Glia Biology, VIB, 3000 Leuven, Belgium
| | - Dirk Daelemans
- Laboratory of Virology and Chemotherapy, Rega Institute, KU Leuven, 3000 Leuven, Belgium
| | | | - Bart Ghesquière
- Metabolomics Core Facility, Vesalius Research Center, VIB, 3000 Leuven, Belgium
| | - Michele Giugliano
- Laboratory of Theoretical Neurobiology and Neuroengineering, University of Antwerp, 2610 Wilrijk, Belgium; Neuro-Electronics Research Flanders, 3001 Leuven, Belgium; Brain Mind Institute, Swiss Federal Institute of Technology of Lausanne, 1015 Lausanne, Switzerland
| | - Carmen Ruiz de Almodovar
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, KU Leuven, 3000 Leuven, Belgium; Laboratory of Angiogenesis and Vascular Metabolism, Vesalius Research Center, VIB, 3000 Leuven, Belgium
| | - Mieke Dewerchin
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, KU Leuven, 3000 Leuven, Belgium; Laboratory of Angiogenesis and Vascular Metabolism, Vesalius Research Center, VIB, 3000 Leuven, Belgium
| | - Peter Carmeliet
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, KU Leuven, 3000 Leuven, Belgium; Laboratory of Angiogenesis and Vascular Metabolism, Vesalius Research Center, VIB, 3000 Leuven, Belgium.
| |
Collapse
|
110
|
Juo LY, Liao WC, Shih YL, Yang BY, Liu AB, Yan YT. HSPB7 interacts with dimerized FLNC and its absence results in progressive myopathy in skeletal muscles. J Cell Sci 2016; 129:1661-70. [PMID: 26929074 PMCID: PMC4852768 DOI: 10.1242/jcs.179887] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Accepted: 02/17/2016] [Indexed: 12/30/2022] Open
Abstract
HSPB7 belongs to the small heat-shock protein (sHSP) family, and its expression is restricted to cardiac and skeletal muscles from embryonic stages to adulthood. Here, we found that skeletal-muscle-specific ablation of the HspB7 does not affect myogenesis during embryonic stages to postnatal day 1 (P1), but causes subsequent postnatal death owing to a respiration defect, with progressive myopathy phenotypes in the diaphragm. Deficiency of HSPB7 in the diaphragm muscle resulted in muscle fibrosis, sarcomere disarray and sarcolemma integrity loss. We identified dimerized filamin C (FLNC) as an interacting partner of HSPB7. Immunofluorescence studies demonstrated that the aggregation and mislocalization of FLNC occurred in the muscle of HspB7 mutant adult mice. Furthermore, the components of dystrophin glycoprotein complex, γ- and δ-sarcoglycan, but not dystrophin, were abnormally upregulated and mislocalized in HSPB7 mutant muscle. Collectively, our findings suggest that HSPB7 is essential for maintaining muscle integrity, which is achieved through its interaction with FLNC, in order to prevent the occurrence and progression of myopathy. Highlighted Article: HSPB7 plays a crucial role in the maintenance of the muscle integrity, possibly through stabilizing the function of FLNC.
Collapse
Affiliation(s)
- Liang-Yi Juo
- Institute of Biochemistry and Molecular Biology, National Yang-Ming University, Taipei 112, Taiwan Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan
| | - Wern-Chir Liao
- Institute of Biochemistry and Molecular Biology, National Yang-Ming University, Taipei 112, Taiwan Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan
| | - Yen-Ling Shih
- Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan
| | - Bih-Ying Yang
- Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan
| | - An-Bang Liu
- Department of Neurology, Buddhist Tzu Chi General Hospital and Buddhist Tzu Chi University, Hualien 970, Taiwan
| | - Yu-Ting Yan
- Institute of Biochemistry and Molecular Biology, National Yang-Ming University, Taipei 112, Taiwan Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan
| |
Collapse
|
111
|
New insights into the protein aggregation pathology in myotilinopathy by combined proteomic and immunolocalization analyses. Acta Neuropathol Commun 2016; 4:8. [PMID: 26842778 PMCID: PMC4739336 DOI: 10.1186/s40478-016-0280-0] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2016] [Accepted: 01/23/2016] [Indexed: 01/09/2023] Open
Abstract
Introduction Myofibrillar myopathies are characterized by progressive muscle weakness and impressive abnormal protein aggregation in muscle fibers. In about 10 % of patients, the disease is caused by mutations in the MYOT gene encoding myotilin. The aim of our study was to decipher the composition of protein deposits in myotilinopathy to get new information about aggregate pathology. Results Skeletal muscle samples from 15 myotilinopathy patients were included in the study. Aggregate and control samples were collected from muscle sections by laser microdissection and subsequently analyzed by a highly sensitive proteomic approach that enables a relative protein quantification. In total 1002 different proteins were detected. Seventy-six proteins showed a significant over-representation in aggregate samples including 66 newly identified aggregate proteins. Z-disc-associated proteins were the most abundant aggregate components, followed by sarcolemmal and extracellular matrix proteins, proteins involved in protein quality control and degradation, and proteins with a function in actin dynamics or cytoskeletal transport. Forty over-represented proteins were evaluated by immunolocalization studies. These analyses validated our mass spectrometric data and revealed different regions of protein accumulation in abnormal muscle fibers. Comparison of data from our proteomic analysis in myotilinopathy with findings in other myofibrillar myopathy subtypes indicates a characteristic basic pattern of aggregate composition and resulted in identification of a highly sensitive and specific diagnostic marker for myotilinopathy. Conclusions Our findings i) indicate that main protein components of aggregates belong to a network of interacting proteins, ii) provide new insights into the complex regulation of protein degradation in myotilinopathy that may be relevant for new treatment strategies, iii) imply a combination of a toxic gain-of-function leading to myotilin-positive protein aggregates and a loss-of-function caused by a shift in subcellular distribution with a deficiency of myotilin at Z-discs that impairs the integrity of myofibrils, and iv) demonstrate that proteomic analysis can be helpful in differential diagnosis of protein aggregate myopathies. Electronic supplementary material The online version of this article (doi:10.1186/s40478-016-0280-0) contains supplementary material, which is available to authorized users.
Collapse
|
112
|
Lah M, Niranjan T, Srikanth S, Holloway L, Schwartz CE, Wang T, Weaver DD. A distinct X-linked syndrome involving joint contractures, keloids, large optic cup-to-disc ratio, and renal stones results from a filamin A (FLNA) mutation. Am J Med Genet A 2016; 170A:881-90. [PMID: 26804200 DOI: 10.1002/ajmg.a.37567] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Accepted: 12/29/2015] [Indexed: 11/09/2022]
Abstract
We further evaluated a previously reported family with an apparently undescribed X-linked syndrome involving joint contractures, keloids, an increased optic cup-to-disc ratio, and renal stones to elucidate the genetic cause. To do this, we obtained medical histories and performed physical examination on 14 individuals in the family, five of whom are affected males and three are obligate carrier females. Linkage analysis was performed on all but one individual and chromosome X-exome sequencing was done on two affected males. The analysis localized the putative gene to Xq27-qter and chromosome X-exome sequencing revealed a mutation in exon 28 (c.4726G>A) of the filamin A (FLNA) gene, predicting that a conserved glycine had been replaced by arginine at amino acid 1576 (p.G1576R). Segregation analysis demonstrated that all known carrier females tested were heterozygous (G/A), all affected males were hemizygous for the mutation (A allele) and all normal males were hemizygous for the normal G allele. The data and the bioinformatic analysis indicate that the G1576R mutation in the FLNA gene is very likely pathogenic in this family. The syndrome affecting the family shares phenotypic overlap with other syndromes caused by FLNA mutations, but appears to be a distinct phenotype, likely representing a unique genetic syndrome.
Collapse
Affiliation(s)
- Melissa Lah
- Indiana University School of Medicine, Indianapolis, Indiana
| | - Tejasvi Niranjan
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University, Baltimore, Maryland
| | | | | | | | - Tao Wang
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University, Baltimore, Maryland
| | - David D Weaver
- Indiana University School of Medicine, Indianapolis, Indiana
| |
Collapse
|
113
|
Dotson D, Woodruff EA, Villalta F, Dong X. Filamin A Is Involved in HIV-1 Vpu-mediated Evasion of Host Restriction by Modulating Tetherin Expression. J Biol Chem 2016; 291:4236-46. [PMID: 26742839 DOI: 10.1074/jbc.m115.708123] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Indexed: 11/06/2022] Open
Abstract
Tetherin, also known as bone marrow stromal antigen 2 (BST-2), inhibits the release of a wide range of enveloped viruses, including human immunodeficiency virus, type 1 (HIV-1) by directly tethering nascent virions to the surface of infected cells. The HIV-1 accessary protein Vpu counteracts tetherin restriction via sequestration, down-regulation, and/or displacement mechanisms to remove tetherin from sites of virus budding. However, the exact mechanism of Vpu-mediated antagonism of tetherin restriction remains to be fully understood. Here we report a novel role for the actin cross-linking regulator filamin A (FLNa) in Vpu anti-tetherin activities. We demonstrate that FLNa associates with tetherin and that FLNa modulates tetherin turnover. FLNa deficiency was found to enhance cell surface and steady-state levels of tetherin expression. In contrast, we observed that overexpression of FLNa reduced tetherin expression levels both on the plasma membrane and in intracellular compartments. Although FLNb shows high amino acid sequence similarity with FLNa, we reveal that only FLNa, but not FLNb, plays an essential role in tetherin turnover. We further showed that FLNa deficiency inhibited Vpu-mediated enhancement of virus release through interfering with the activity of Vpu to down-regulate cellular tetherin. Taken together, our studies suggest that Vpu hijacks the FLNa function in the modulation of tetherin to neutralize the antiviral factor tetherin. These findings may provide novel strategies for the treatment of HIV-1 infection.
Collapse
Affiliation(s)
- Dominique Dotson
- From the Department of Microbiology and Immunology and Center for AIDS Health Disparities Research, School of Medicine, Meharry Medical College, Nashville, Tennessee 37208
| | - Elvin A Woodruff
- From the Department of Microbiology and Immunology and Center for AIDS Health Disparities Research, School of Medicine, Meharry Medical College, Nashville, Tennessee 37208
| | - Fernando Villalta
- From the Department of Microbiology and Immunology and Center for AIDS Health Disparities Research, School of Medicine, Meharry Medical College, Nashville, Tennessee 37208
| | - Xinhong Dong
- From the Department of Microbiology and Immunology and Center for AIDS Health Disparities Research, School of Medicine, Meharry Medical College, Nashville, Tennessee 37208
| |
Collapse
|
114
|
Madawala RJ, Poon CE, Dowland SN, Murphy CR. Actin crosslinking protein filamin A during early pregnancy in the rat uterus. Reprod Fertil Dev 2016; 28:960-968. [DOI: 10.1071/rd14240] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2014] [Accepted: 11/13/2014] [Indexed: 02/01/2023] Open
Abstract
During early pregnancy the endometrium undergoes a major transformation in order for it to become receptive to blastocyst implantation. The actin cytoskeleton and plasma membrane of luminal uterine epithelial cells (UECs) and the underlying stromal cells undergo dramatic remodelling to facilitate these changes. Filamin A (FLNA), a protein that crosslinks actin filaments and also mediates the anchorage of membrane proteins to the actin cytoskeleton, was investigated in the rat uterus at fertilisation (Day 1) and implantation (Day 6) to determine the role of FLNA in actin cytoskeletal remodelling of UECs and decidua during early pregnancy. Localisation of FLNA in UECs at the time of fertilisation was cytoplasmic, whilst at implantation it was distributed apically; its localisation is under the influence of progesterone. FLNA was also concentrated to the first two to three stromal cell layers at the time of fertilisation and shifted to the primary decidualisation zone at the time of implantation. This shift in localisation was found to be dependent on the decidualisation reaction. Protein abundance of the FLNA 280-kDa monomer and calpain-cleaved fragment (240 kDa) did not change during early pregnancy in UECs. Since major actin cytoskeletal remodelling occurs during early pregnancy in UECs and in decidual cells, the changing localisation of FLNA suggests that it may be an important regulator of cytoskeletal remodelling of these cells to allow uterine receptivity and decidualisation necessary for implantation in the rat.
Collapse
|
115
|
Janssens J, Philtjens S, Kleinberger G, Van Mossevelde S, van der Zee J, Cacace R, Engelborghs S, Sieben A, Banzhaf-Strathmann J, Dillen L, Merlin C, Cuijt I, Robberecht C, Schmid B, Santens P, Ivanoiu A, Vandenbulcke M, Vandenberghe R, Cras P, De Deyn PP, Martin JJ, Maudsley S, Haass C, Cruts M, Van Broeckhoven C. Investigating the role of filamin C in Belgian patients with frontotemporal dementia linked to GRN deficiency in FTLD-TDP brains. Acta Neuropathol Commun 2015; 3:68. [PMID: 26555887 PMCID: PMC4641381 DOI: 10.1186/s40478-015-0246-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Accepted: 10/21/2015] [Indexed: 12/13/2022] Open
Abstract
TAR DNA-binding protein 43 (TDP-43) inclusions are pathological hallmarks of patients with frontotemporal lobar degeneration (FTLD) and amyotrophic lateral sclerosis (ALS). Loss of TDP-43 in zebrafish engenders a severe muscle and vascular phenotype with a concomitant elevation of filamin C (FLNC) levels, an observation confirmed in the frontal cortex of FTLD-TDP patients. Here, we aimed to further assess the contribution of FLNC to frontotemporal dementia (FTD) etiology. We conducted a mutational screening of FLNC in a cohort of 529 unrelated Belgian FTD and FTD-ALS patients, and a control cohort of 920 unrelated and age-matched individuals. Additionally we performed an in-depth characterization of FLNC expression levels in FTD patients and a murine FTD model. In total 68 missense variants were identified of which 19 (MAF < 1 %) were patient-only. Gene burden analysis demonstrated a significant association between the presence of rare variants in FLNC and disease (P = 0.0349, RR = 1.46 [95 % CI 1.03–2.07]). Furthermore, elevated FLNC expression levels, observed previously in FTLD-TDP patients, were mainly attributable to FTD patients with the progranulin (GRN) p.0(IVS1 + 5G > C) loss-of-function mutation. Increased FLNC levels were, to a lesser extent, also identified in a FLNC p.V831I variant carrier and in FTD patients with the p.R159H mutation in valosin-containing protein (VCP). The GRN-associated increase of FLNC was confirmed in the frontal cortex of aged Grn knockout mice starting at 16–18 months of age. Combined quantitative proteomic and bioinformatic analyses of the frontal cortex of FTD patients possessing elevated FLNC levels, identified multiple altered protein factors involved in accelerated aging, neurodegeneration and synaptogenesis. Our findings further support the involvement of aberrant FLNC expression levels in FTD pathogenesis. Identification of increased FLNC levels in aged Grn mice and impaired pathways related to aging and neurodegeneration, implies a potential role for FLNC in mediating or accelerating the aging process.
Collapse
|
116
|
Tirupula KC, Ithychanda SS, Mohan ML, Naga Prasad SV, Qin J, Karnik SS. G protein-coupled receptors directly bind filamin A with high affinity and promote filamin phosphorylation. Biochemistry 2015; 54:6673-83. [PMID: 26460884 PMCID: PMC4642222 DOI: 10.1021/acs.biochem.5b00975] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Although interaction of a few G protein-coupled receptors (GPCRs) with Filamin A, a key actin cross-linking and biomechanical signal transducer protein, has been observed, a comprehensive structure-function analysis of this interaction is lacking. Through a systematic sequence-based analysis, we found that a conserved filamin binding motif is present in the cytoplasmic domains of >20% of the 824 GPCRs encoded in the human genome. Direct high-affinity interaction of filamin binding motif peptides of select GPCRs with the Ig domain of Filamin A was confirmed by nuclear magnetic resonance spectroscopy and isothermal titration calorimetric experiments. Engagement of the filamin binding motif with the Filamin A Ig domain induced the phosphorylation of filamin by protein kinase A in vitro. In transfected cells, agonist activation as well as constitutive activation of representative GPCRs dramatically elicited recruitment and phosphorylation of cellular Filamin A, a phenomenon long known to be crucial for regulating the structure and dynamics of the cytoskeleton. Our data suggest a molecular mechanism for direct GPCR-cytoskeleton coupling via filamin. Until now, GPCR signaling to the cytoskeleton was predominantly thought to be indirect, through canonical G protein-mediated signaling cascades involving GTPases, adenylyl cyclases, phospholipases, ion channels, and protein kinases. We propose that the GPCR-induced filamin phosphorylation pathway is a conserved, novel biochemical signaling paradigm.
Collapse
Affiliation(s)
- Kalyan C Tirupula
- Department of Molecular Cardiology, Lerner Research Institute, Cleveland Clinic , Cleveland, Ohio 44195, United States
| | - Sujay S Ithychanda
- Department of Molecular Cardiology, Lerner Research Institute, Cleveland Clinic , Cleveland, Ohio 44195, United States
| | - Maradumane L Mohan
- Department of Molecular Cardiology, Lerner Research Institute, Cleveland Clinic , Cleveland, Ohio 44195, United States
| | - Sathyamangla V Naga Prasad
- Department of Molecular Cardiology, Lerner Research Institute, Cleveland Clinic , Cleveland, Ohio 44195, United States
| | - Jun Qin
- Department of Molecular Cardiology, Lerner Research Institute, Cleveland Clinic , Cleveland, Ohio 44195, United States
| | - Sadashiva S Karnik
- Department of Molecular Cardiology, Lerner Research Institute, Cleveland Clinic , Cleveland, Ohio 44195, United States
| |
Collapse
|
117
|
Clavier S, Illien F, Sagan S, Bolbach G, Sachon E. Proteomic comparison of the EWS-FLI1 expressing cells EF with NIH-3T3 and actin remodeling effect of (R/W) 9 cell-penetrating peptide. EUPA OPEN PROTEOMICS 2015; 10:1-8. [PMID: 29900093 PMCID: PMC5988571 DOI: 10.1016/j.euprot.2015.10.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Revised: 08/25/2015] [Accepted: 10/25/2015] [Indexed: 12/13/2022]
Abstract
Comparison of tumoral EF versus non-tumoral 3T3 fibroblasts (SILAC). Characterization of EWS-FLI1 fusion protein impact on protein expression levels. Down-regulation of actin binding proteins responsible for passive dissemination. Investigation of (R/W)9 cell-penetrating peptide actin remodeling activity. First proteomics study using a cell-penetrating peptide (R/W)9.
EWS-FLI1 expression in NIH-3T3 fibroblasts has a profound impact on the phenotype, resulting in the cytoskeleton and adhesive capacity disorganization (EF cells). Besides this, (R/W)9, a cell-penetrating peptide (CPP), has an intrinsic actin remodeling activity in EF cells. To evaluate the impact of the oncogenic protein EWS-FLI1 on proteins expression levels, a quantitative comparison of tumoral EF and non-tumoral 3T3 proteomes was performed. Then to see if we could link the EWS-FLI1 oncogenic transformation to the phenotype reversion induced by (R/W)9, (R/W)9 influence on EF cells proteome was assessed. To our knowledge no such CPPomic study has been performed before. Biological significance Up to now very few global quantitative proteomic studies have been published to help understand the oncogenic transformation induced by EWS-FLI1 fusion protein and leading to Ewing sarcoma development and dissemination. The comparison we did in this study between a model tumoral cell line EF and its non-tumoral counterpart (3T3) allowed us to highlight several features either common to most tumor types or specific to Ewing sarcoma. Particularly, lack of actin cytoskeleton organization could very likely be explained by the down-regulation of many important actin binding proteins. These results are in accordance with the hypothesis of a passive/stochastic mode of dissemination conferring Ewing sarcoma tumoral cell a high metastatic potential.
Collapse
Affiliation(s)
- Séverine Clavier
- Sorbonne Université, UPMC-Univ Paris 6, Ecole Normale Supérieure, PSL Research University, Département de Chimie, CNRS, UMR7203 Laboratoire des BioMolécules, 4 Place Jussieu, Paris Cedex 05, 75252 Paris, France.,Sorbonne Université, UPMCUniv Paris 6, Plateforme de Spectrométrie de Masse et Protéomique-IBPS, cc41, 7-9 Quai Saint Bernard, Paris Cedex 05, 75252 Paris, France
| | - Françoise Illien
- Sorbonne Université, UPMCUniv Paris 6, Plateforme de Spectrométrie de Masse et Protéomique-IBPS, cc41, 7-9 Quai Saint Bernard, Paris Cedex 05, 75252 Paris, France
| | - Sandrine Sagan
- Sorbonne Université, UPMCUniv Paris 6, Plateforme de Spectrométrie de Masse et Protéomique-IBPS, cc41, 7-9 Quai Saint Bernard, Paris Cedex 05, 75252 Paris, France
| | - Gérard Bolbach
- Sorbonne Université, UPMC-Univ Paris 6, Ecole Normale Supérieure, PSL Research University, Département de Chimie, CNRS, UMR7203 Laboratoire des BioMolécules, 4 Place Jussieu, Paris Cedex 05, 75252 Paris, France.,Sorbonne Université, UPMCUniv Paris 6, Plateforme de Spectrométrie de Masse et Protéomique-IBPS, cc41, 7-9 Quai Saint Bernard, Paris Cedex 05, 75252 Paris, France
| | - Emmanuelle Sachon
- Sorbonne Université, UPMC-Univ Paris 6, Ecole Normale Supérieure, PSL Research University, Département de Chimie, CNRS, UMR7203 Laboratoire des BioMolécules, 4 Place Jussieu, Paris Cedex 05, 75252 Paris, France.,Sorbonne Université, UPMCUniv Paris 6, Plateforme de Spectrométrie de Masse et Protéomique-IBPS, cc41, 7-9 Quai Saint Bernard, Paris Cedex 05, 75252 Paris, France
| |
Collapse
|
118
|
Zhao Y, Shapiro SS, Eto M. F-actin clustering and cell dysmotility induced by the pathological W148R missense mutation of filamin B at the actin-binding domain. Am J Physiol Cell Physiol 2015; 310:C89-98. [PMID: 26491051 DOI: 10.1152/ajpcell.00274.2015] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Accepted: 10/19/2015] [Indexed: 11/22/2022]
Abstract
Filamin B (FLNB) is a dimeric actin-binding protein that orchestrates the reorganization of the actin cytoskeleton. Congenital mutations of FLNB at the actin-binding domain (ABD) are known to cause abnormalities of skeletal development, such as atelosteogenesis types I and III and Larsen's syndrome, although the underlying mechanisms are poorly understood. Here, using fluorescence microscopy, we characterized the reorganization of the actin cytoskeleton in cells expressing each of six pathological FLNB mutants that have been linked to skeletal abnormalities. The subfractionation assay showed a greater accumulation of the FLNB ABD mutants W148R and E227K than the wild-type protein to the cytoskeleton. Ectopic expression of FLNB-W148R and, to a lesser extent, FLNB-E227K induced prominent F-actin accumulations and the consequent rearrangement of focal adhesions, myosin II, and septin filaments and results in a delayed directional migration of the cells. The W148R protein-induced cytoskeletal rearrangement was partially attenuated by the inhibition of myosin II, p21-activated protein kinase, or Rho-associated protein kinase. The expression of a single-head ABD fragment with the mutations partially mimicked the rearrangement induced by the dimer. The F-actin clustering through the interaction with the mutant FLNB ABD may limit the cytoskeletal reorganization, preventing normal skeletal development.
Collapse
Affiliation(s)
- Yongtong Zhao
- Department of Molecular Physiology and Biophysics, Sidney Kimmel Medical College at Thomas Jefferson University, and Sidney Kimmel Cancer Center, Philadelphia, Pennsylvania
| | - Sandor S Shapiro
- Department of Molecular Physiology and Biophysics, Sidney Kimmel Medical College at Thomas Jefferson University, and Sidney Kimmel Cancer Center, Philadelphia, Pennsylvania
| | - Masumi Eto
- Department of Molecular Physiology and Biophysics, Sidney Kimmel Medical College at Thomas Jefferson University, and Sidney Kimmel Cancer Center, Philadelphia, Pennsylvania
| |
Collapse
|
119
|
Chevessier F, Schuld J, Orfanos Z, Plank AC, Wolf L, Maerkens A, Unger A, Schlötzer-Schrehardt U, Kley RA, Von Hörsten S, Marcus K, Linke WA, Vorgerd M, van der Ven PFM, Fürst DO, Schröder R. Myofibrillar instability exacerbated by acute exercise in filaminopathy. Hum Mol Genet 2015; 24:7207-20. [PMID: 26472074 DOI: 10.1093/hmg/ddv421] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Accepted: 10/02/2015] [Indexed: 12/12/2022] Open
Abstract
Filamin C (FLNC) mutations in humans cause myofibrillar myopathy (MFM) and cardiomyopathy, characterized by protein aggregation and myofibrillar degeneration. We generated the first patient-mimicking knock-in mouse harbouring the most common disease-causing filamin C mutation (p.W2710X). These heterozygous mice developed muscle weakness and myofibrillar instability, with formation of filamin C- and Xin-positive lesions streaming between Z-discs. These lesions, which are distinct from the classical MFM protein aggregates by their morphology and filamentous appearance, were greatly increased in number upon acute physical exercise in the mice. This pathology suggests that mutant filamin influences the mechanical stability of myofibrillar Z-discs, explaining the muscle weakness in mice and humans. Re-evaluation of biopsies from MFM-filaminopathy patients with different FLNC mutations revealed a similar, previously unreported lesion pathology, in addition to the classical protein aggregates, and suggested that structures previously interpreted as aggregates may be in part sarcomeric lesions. We postulate that these lesions define preclinical disease stages, preceding the formation of protein aggregates.
Collapse
Affiliation(s)
| | - Julia Schuld
- Department of Molecular Cell Biology, Institute for Cell Biology, University of Bonn, Bonn, Germany
| | - Zacharias Orfanos
- Department of Molecular Cell Biology, Institute for Cell Biology, University of Bonn, Bonn, Germany
| | - Anne-C Plank
- Department for Experimental Therapy, Preclinical Experimental Animal Center and
| | | | - Alexandra Maerkens
- Department of Neurology, Neuromuscular Center Ruhrgebiet, University Hospital Bergmannsheil, Department of Functional Proteomics, Medizinisches Proteom-Center and
| | - Andreas Unger
- Department of Cardiovascular Physiology, Ruhr-University Bochum, Bochum, Germany
| | | | - Rudolf A Kley
- Department of Neurology, Neuromuscular Center Ruhrgebiet, University Hospital Bergmannsheil
| | - Stephan Von Hörsten
- Department for Experimental Therapy, Preclinical Experimental Animal Center and
| | - Katrin Marcus
- Department of Functional Proteomics, Medizinisches Proteom-Center and
| | - Wolfgang A Linke
- Department of Cardiovascular Physiology, Ruhr-University Bochum, Bochum, Germany
| | - Matthias Vorgerd
- Department of Neurology, Neuromuscular Center Ruhrgebiet, University Hospital Bergmannsheil
| | - Peter F M van der Ven
- Department of Molecular Cell Biology, Institute for Cell Biology, University of Bonn, Bonn, Germany
| | - Dieter O Fürst
- Department of Molecular Cell Biology, Institute for Cell Biology, University of Bonn, Bonn, Germany,
| | | |
Collapse
|
120
|
Béhin A, Salort-Campana E, Wahbi K, Richard P, Carlier RY, Carlier P, Laforêt P, Stojkovic T, Maisonobe T, Verschueren A, Franques J, Attarian S, Maues de Paula A, Figarella-Branger D, Bécane HM, Nelson I, Duboc D, Bonne G, Vicart P, Udd B, Romero N, Pouget J, Eymard B. Myofibrillar myopathies: State of the art, present and future challenges. Rev Neurol (Paris) 2015; 171:715-29. [DOI: 10.1016/j.neurol.2015.06.002] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2014] [Revised: 05/11/2015] [Accepted: 06/02/2015] [Indexed: 12/18/2022]
|
121
|
Seppälä J, Tossavainen H, Rodic N, Permi P, Pentikäinen U, Ylänne J. Flexible Structure of Peptide-Bound Filamin A Mechanosensor Domain Pair 20-21. PLoS One 2015; 10:e0136969. [PMID: 26322797 PMCID: PMC4554727 DOI: 10.1371/journal.pone.0136969] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Accepted: 08/12/2015] [Indexed: 11/18/2022] Open
Abstract
Filamins (FLNs) are large, multidomain actin cross-linking proteins with diverse functions. Besides regulating the actin cytoskeleton, they serve as important links between the extracellular matrix and the cytoskeleton by binding cell surface receptors, functioning as scaffolds for signaling proteins, and binding several other cytoskeletal proteins that regulate cell adhesion dynamics. Structurally, FLNs are formed of an amino terminal actin-binding domain followed by 24 immunoglobulin-like domains (IgFLNs). Recent studies have demonstrated that myosin-mediated contractile forces can reveal hidden protein binding sites in the domain pairs IgFLNa18–19 and 20–21, enabling FLNs to transduce mechanical signals in cells. The atomic structures of these mechanosensor domain pairs in the resting state are known, as well as the structures of individual IgFLN21 with ligand peptides. However, little experimental data is available on how interacting protein binding deforms the domain pair structures. Here, using small-angle x-ray scattering-based modelling, x-ray crystallography, and NMR, we show that the adaptor protein migfilin-derived peptide-bound structure of IgFLNa20–21 is flexible and adopts distinctive conformations depending on the presence or absence of the interacting peptide. The conformational changes reported here may be common for all peptides and may play a role in the mechanosensor function of the site.
Collapse
Affiliation(s)
- Jonne Seppälä
- Department of Biological and Environmental Science and Nanoscience Center, University of Jyväskylä, Jyväskylä, Finland
- * E-mail:
| | - Helena Tossavainen
- Program in Structural Biology and Biophysics, Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Nebojsa Rodic
- Program in Structural Biology and Biophysics, Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Perttu Permi
- Program in Structural Biology and Biophysics, Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Ulla Pentikäinen
- Department of Biological and Environmental Science and Nanoscience Center, University of Jyväskylä, Jyväskylä, Finland
| | - Jari Ylänne
- Department of Biological and Environmental Science and Nanoscience Center, University of Jyväskylä, Jyväskylä, Finland
| |
Collapse
|
122
|
Na BR, Kim HR, Piragyte I, Oh HM, Kwon MS, Akber U, Lee HS, Park DS, Song WK, Park ZY, Im SH, Rho MC, Hyun YM, Kim M, Jun CD. TAGLN2 regulates T cell activation by stabilizing the actin cytoskeleton at the immunological synapse. ACTA ACUST UNITED AC 2015; 209:143-62. [PMID: 25869671 PMCID: PMC4395477 DOI: 10.1083/jcb.201407130] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
TAGLN2 stabilizes cortical F-actin and thereby maintains F-actin contents at the immunological synapse, which allows T cell activation following T cell receptor stimulation. The formation of an immunological synapse (IS) requires tight regulation of actin dynamics by many actin polymerizing/depolymerizing proteins. However, the significance of actin stabilization at the IS remains largely unknown. In this paper, we identify a novel function of TAGLN2—an actin-binding protein predominantly expressed in T cells—in stabilizing cortical F-actin, thereby maintaining F-actin contents at the IS and acquiring LFA-1 (leukocyte function-associated antigen-1) activation after T cell receptor stimulation. TAGLN2 blocks actin depolymerization and competes with cofilin both in vitro and in vivo. Knockout of TAGLN2 (TAGLN2−/−) reduced F-actin content and destabilized F-actin ring formation, resulting in decreased cell adhesion and spreading. TAGLN2−/− T cells displayed weakened cytokine production and cytotoxic effector function. These findings reveal a novel function of TAGLN2 in enhancing T cell responses by controlling actin stability at the IS.
Collapse
Affiliation(s)
- Bo-Ra Na
- School of Life Sciences, Immune Synapse Research Center and Cell Dynamics Research Center, Gwangju Institute of Science and Technology (GIST), Gwangju 500-712, South Korea
| | - Hye-Ran Kim
- School of Life Sciences, Immune Synapse Research Center and Cell Dynamics Research Center, Gwangju Institute of Science and Technology (GIST), Gwangju 500-712, South Korea
| | - Indre Piragyte
- School of Life Sciences, Immune Synapse Research Center and Cell Dynamics Research Center, Gwangju Institute of Science and Technology (GIST), Gwangju 500-712, South Korea
| | - Hyun-Mee Oh
- Bioindustrial Process Research Center, Korea Research Institute Bioscience and Biotechnology (KRIBB), Jeongeup 580-185, South Korea
| | - Min-Sung Kwon
- School of Life Sciences, Immune Synapse Research Center and Cell Dynamics Research Center, Gwangju Institute of Science and Technology (GIST), Gwangju 500-712, South Korea
| | - Uroos Akber
- School of Life Sciences, Immune Synapse Research Center and Cell Dynamics Research Center, Gwangju Institute of Science and Technology (GIST), Gwangju 500-712, South Korea
| | - Hyun-Su Lee
- School of Life Sciences, Immune Synapse Research Center and Cell Dynamics Research Center, Gwangju Institute of Science and Technology (GIST), Gwangju 500-712, South Korea
| | - Do-Sim Park
- Department of Laboratory Medicine, School of Medicine, Wonkwang University, Iksan 570-749, South Korea
| | - Woo Keun Song
- School of Life Sciences, Immune Synapse Research Center and Cell Dynamics Research Center, Gwangju Institute of Science and Technology (GIST), Gwangju 500-712, South Korea
| | - Zee-Yong Park
- School of Life Sciences, Immune Synapse Research Center and Cell Dynamics Research Center, Gwangju Institute of Science and Technology (GIST), Gwangju 500-712, South Korea
| | - Sin-Hyeog Im
- School of Life Sciences, Immune Synapse Research Center and Cell Dynamics Research Center, Gwangju Institute of Science and Technology (GIST), Gwangju 500-712, South Korea
| | - Mun-Chual Rho
- Department of Laboratory Medicine, School of Medicine, Wonkwang University, Iksan 570-749, South Korea
| | - Young-Min Hyun
- Department of Microbiology and Immunology, David H. Smith Center for Vaccine Biology and Immunology, University of Rochester, Rochester, NY 14642
| | - Minsoo Kim
- Department of Microbiology and Immunology, David H. Smith Center for Vaccine Biology and Immunology, University of Rochester, Rochester, NY 14642
| | - Chang-Duk Jun
- School of Life Sciences, Immune Synapse Research Center and Cell Dynamics Research Center, Gwangju Institute of Science and Technology (GIST), Gwangju 500-712, South Korea
| |
Collapse
|
123
|
Savoy RM, Chen L, Siddiqui S, Melgoza FU, Durbin-Johnson B, Drake C, Jathal MK, Bose S, Steele TM, Mooso BA, D'Abronzo LS, Fry WH, Carraway KL, Mudryj M, Ghosh PM. Transcription of Nrdp1 by the androgen receptor is regulated by nuclear filamin A in prostate cancer. Endocr Relat Cancer 2015; 22:369-86. [PMID: 25759396 PMCID: PMC4433410 DOI: 10.1530/erc-15-0021] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/10/2015] [Indexed: 02/06/2023]
Abstract
Prostate cancer (PCa) progression is regulated by the androgen receptor (AR); however, patients undergoing androgen-deprivation therapy (ADT) for disseminated PCa eventually develop castration-resistant PCa (CRPC). Results of previous studies indicated that AR, a transcription factor, occupies distinct genomic loci in CRPC compared with hormone-naïve PCa; however, the cause of this distinction was unknown. The E3 ubiquitin ligase Nrdp1 is a model AR target modulated by androgens in hormone-naïve PCa but not in CRPC. Using Nrdp1, we investigated how AR switches transcription programs during CRPC progression. The proximal Nrdp1 promoter contains an androgen response element (ARE); we demonstrated AR binding to this ARE in androgen-sensitive PCa. Analysis of hormone-naive human prostatectomy specimens revealed correlation between Nrdp1 and AR expression, supporting AR regulation of NRDP1 levels in androgen-sensitive tissue. However, despite sustained AR levels, AR binding to the Nrdp1 promoter and Nrdp1 expression were suppressed in CRPC. Elucidation of the suppression mechanism demonstrated correlation of NRDP1 levels with nuclear localization of the scaffolding protein filamin A (FLNA) which, as we previously showed, is itself repressed following ADT in many CRPC tumors. Restoration of nuclear FLNA in CRPC stimulated AR binding to Nrdp1 ARE, increased its transcription, and augmented NRDP1 protein expression and responsiveness to ADT, indicating that nuclear FLNA controls AR-mediated androgen-sensitive Nrdp1 transcription. Expression of other AR-regulated genes lost in CRPC was also re-established by nuclear FLNA. Thus, our results indicate that nuclear FLNA promotes androgen-dependent AR-regulated transcription in PCa, while loss of nuclear FLNA in CRPC alters the AR-regulated transcription program.
Collapse
Affiliation(s)
- Rosalinda M Savoy
- VA Northern California Health Care SystemMather, California, USADepartment of UrologySchool of Medicine, University of California Davis, 4860 Y Street, Suite 3500, Sacramento, California 95817, USADivision of BiostatisticsDepartment of Public Health Sciences, University of California Davis, Davis, California, USADepartment of StatisticsUniversity of California Davis, Davis, California, USADepartment of Biochemistry and Molecular MedicineUniversity of California Davis, Sacramento, California, USADepartment of Medical Microbiology and ImmunologyUniversity of California Davis, Davis, California, USA VA Northern California Health Care SystemMather, California, USADepartment of UrologySchool of Medicine, University of California Davis, 4860 Y Street, Suite 3500, Sacramento, California 95817, USADivision of BiostatisticsDepartment of Public Health Sciences, University of California Davis, Davis, California, USADepartment of StatisticsUniversity of California Davis, Davis, California, USADepartment of Biochemistry and Molecular MedicineUniversity of California Davis, Sacramento, California, USADepartment of Medical Microbiology and ImmunologyUniversity of California Davis, Davis, California, USA
| | - Liqun Chen
- VA Northern California Health Care SystemMather, California, USADepartment of UrologySchool of Medicine, University of California Davis, 4860 Y Street, Suite 3500, Sacramento, California 95817, USADivision of BiostatisticsDepartment of Public Health Sciences, University of California Davis, Davis, California, USADepartment of StatisticsUniversity of California Davis, Davis, California, USADepartment of Biochemistry and Molecular MedicineUniversity of California Davis, Sacramento, California, USADepartment of Medical Microbiology and ImmunologyUniversity of California Davis, Davis, California, USA
| | - Salma Siddiqui
- VA Northern California Health Care SystemMather, California, USADepartment of UrologySchool of Medicine, University of California Davis, 4860 Y Street, Suite 3500, Sacramento, California 95817, USADivision of BiostatisticsDepartment of Public Health Sciences, University of California Davis, Davis, California, USADepartment of StatisticsUniversity of California Davis, Davis, California, USADepartment of Biochemistry and Molecular MedicineUniversity of California Davis, Sacramento, California, USADepartment of Medical Microbiology and ImmunologyUniversity of California Davis, Davis, California, USA
| | - Frank U Melgoza
- VA Northern California Health Care SystemMather, California, USADepartment of UrologySchool of Medicine, University of California Davis, 4860 Y Street, Suite 3500, Sacramento, California 95817, USADivision of BiostatisticsDepartment of Public Health Sciences, University of California Davis, Davis, California, USADepartment of StatisticsUniversity of California Davis, Davis, California, USADepartment of Biochemistry and Molecular MedicineUniversity of California Davis, Sacramento, California, USADepartment of Medical Microbiology and ImmunologyUniversity of California Davis, Davis, California, USA
| | - Blythe Durbin-Johnson
- VA Northern California Health Care SystemMather, California, USADepartment of UrologySchool of Medicine, University of California Davis, 4860 Y Street, Suite 3500, Sacramento, California 95817, USADivision of BiostatisticsDepartment of Public Health Sciences, University of California Davis, Davis, California, USADepartment of StatisticsUniversity of California Davis, Davis, California, USADepartment of Biochemistry and Molecular MedicineUniversity of California Davis, Sacramento, California, USADepartment of Medical Microbiology and ImmunologyUniversity of California Davis, Davis, California, USA
| | - Christiana Drake
- VA Northern California Health Care SystemMather, California, USADepartment of UrologySchool of Medicine, University of California Davis, 4860 Y Street, Suite 3500, Sacramento, California 95817, USADivision of BiostatisticsDepartment of Public Health Sciences, University of California Davis, Davis, California, USADepartment of StatisticsUniversity of California Davis, Davis, California, USADepartment of Biochemistry and Molecular MedicineUniversity of California Davis, Sacramento, California, USADepartment of Medical Microbiology and ImmunologyUniversity of California Davis, Davis, California, USA
| | - Maitreyee K Jathal
- VA Northern California Health Care SystemMather, California, USADepartment of UrologySchool of Medicine, University of California Davis, 4860 Y Street, Suite 3500, Sacramento, California 95817, USADivision of BiostatisticsDepartment of Public Health Sciences, University of California Davis, Davis, California, USADepartment of StatisticsUniversity of California Davis, Davis, California, USADepartment of Biochemistry and Molecular MedicineUniversity of California Davis, Sacramento, California, USADepartment of Medical Microbiology and ImmunologyUniversity of California Davis, Davis, California, USA VA Northern California Health Care SystemMather, California, USADepartment of UrologySchool of Medicine, University of California Davis, 4860 Y Street, Suite 3500, Sacramento, California 95817, USADivision of BiostatisticsDepartment of Public Health Sciences, University of California Davis, Davis, California, USADepartment of StatisticsUniversity of California Davis, Davis, California, USADepartment of Biochemistry and Molecular MedicineUniversity of California Davis, Sacramento, California, USADepartment of Medical Microbiology and ImmunologyUniversity of California Davis, Davis, California, USA
| | - Swagata Bose
- VA Northern California Health Care SystemMather, California, USADepartment of UrologySchool of Medicine, University of California Davis, 4860 Y Street, Suite 3500, Sacramento, California 95817, USADivision of BiostatisticsDepartment of Public Health Sciences, University of California Davis, Davis, California, USADepartment of StatisticsUniversity of California Davis, Davis, California, USADepartment of Biochemistry and Molecular MedicineUniversity of California Davis, Sacramento, California, USADepartment of Medical Microbiology and ImmunologyUniversity of California Davis, Davis, California, USA VA Northern California Health Care SystemMather, California, USADepartment of UrologySchool of Medicine, University of California Davis, 4860 Y Street, Suite 3500, Sacramento, California 95817, USADivision of BiostatisticsDepartment of Public Health Sciences, University of California Davis, Davis, California, USADepartment of StatisticsUniversity of California Davis, Davis, California, USADepartment of Biochemistry and Molecular MedicineUniversity of California Davis, Sacramento, California, USADepartment of Medical Microbiology and ImmunologyUniversity of California Davis, Davis, California, USA
| | - Thomas M Steele
- VA Northern California Health Care SystemMather, California, USADepartment of UrologySchool of Medicine, University of California Davis, 4860 Y Street, Suite 3500, Sacramento, California 95817, USADivision of BiostatisticsDepartment of Public Health Sciences, University of California Davis, Davis, California, USADepartment of StatisticsUniversity of California Davis, Davis, California, USADepartment of Biochemistry and Molecular MedicineUniversity of California Davis, Sacramento, California, USADepartment of Medical Microbiology and ImmunologyUniversity of California Davis, Davis, California, USA
| | - Benjamin A Mooso
- VA Northern California Health Care SystemMather, California, USADepartment of UrologySchool of Medicine, University of California Davis, 4860 Y Street, Suite 3500, Sacramento, California 95817, USADivision of BiostatisticsDepartment of Public Health Sciences, University of California Davis, Davis, California, USADepartment of StatisticsUniversity of California Davis, Davis, California, USADepartment of Biochemistry and Molecular MedicineUniversity of California Davis, Sacramento, California, USADepartment of Medical Microbiology and ImmunologyUniversity of California Davis, Davis, California, USA
| | - Leandro S D'Abronzo
- VA Northern California Health Care SystemMather, California, USADepartment of UrologySchool of Medicine, University of California Davis, 4860 Y Street, Suite 3500, Sacramento, California 95817, USADivision of BiostatisticsDepartment of Public Health Sciences, University of California Davis, Davis, California, USADepartment of StatisticsUniversity of California Davis, Davis, California, USADepartment of Biochemistry and Molecular MedicineUniversity of California Davis, Sacramento, California, USADepartment of Medical Microbiology and ImmunologyUniversity of California Davis, Davis, California, USA VA Northern California Health Care SystemMather, California, USADepartment of UrologySchool of Medicine, University of California Davis, 4860 Y Street, Suite 3500, Sacramento, California 95817, USADivision of BiostatisticsDepartment of Public Health Sciences, University of California Davis, Davis, California, USADepartment of StatisticsUniversity of California Davis, Davis, California, USADepartment of Biochemistry and Molecular MedicineUniversity of California Davis, Sacramento, California, USADepartment of Medical Microbiology and ImmunologyUniversity of California Davis, Davis, California, USA
| | - William H Fry
- VA Northern California Health Care SystemMather, California, USADepartment of UrologySchool of Medicine, University of California Davis, 4860 Y Street, Suite 3500, Sacramento, California 95817, USADivision of BiostatisticsDepartment of Public Health Sciences, University of California Davis, Davis, California, USADepartment of StatisticsUniversity of California Davis, Davis, California, USADepartment of Biochemistry and Molecular MedicineUniversity of California Davis, Sacramento, California, USADepartment of Medical Microbiology and ImmunologyUniversity of California Davis, Davis, California, USA
| | - Kermit L Carraway
- VA Northern California Health Care SystemMather, California, USADepartment of UrologySchool of Medicine, University of California Davis, 4860 Y Street, Suite 3500, Sacramento, California 95817, USADivision of BiostatisticsDepartment of Public Health Sciences, University of California Davis, Davis, California, USADepartment of StatisticsUniversity of California Davis, Davis, California, USADepartment of Biochemistry and Molecular MedicineUniversity of California Davis, Sacramento, California, USADepartment of Medical Microbiology and ImmunologyUniversity of California Davis, Davis, California, USA
| | - Maria Mudryj
- VA Northern California Health Care SystemMather, California, USADepartment of UrologySchool of Medicine, University of California Davis, 4860 Y Street, Suite 3500, Sacramento, California 95817, USADivision of BiostatisticsDepartment of Public Health Sciences, University of California Davis, Davis, California, USADepartment of StatisticsUniversity of California Davis, Davis, California, USADepartment of Biochemistry and Molecular MedicineUniversity of California Davis, Sacramento, California, USADepartment of Medical Microbiology and ImmunologyUniversity of California Davis, Davis, California, USA VA Northern California Health Care SystemMather, California, USADepartment of UrologySchool of Medicine, University of California Davis, 4860 Y Street, Suite 3500, Sacramento, California 95817, USADivision of BiostatisticsDepartment of Public Health Sciences, University of California Davis, Davis, California, USADepartment of StatisticsUniversity of California Davis, Davis, California, USADepartment of Biochemistry and Molecular MedicineUniversity of California Davis, Sacramento, California, USADepartment of Medical Microbiology and ImmunologyUniversity of California Davis, Davis, California, USA
| | - Paramita M Ghosh
- VA Northern California Health Care SystemMather, California, USADepartment of UrologySchool of Medicine, University of California Davis, 4860 Y Street, Suite 3500, Sacramento, California 95817, USADivision of BiostatisticsDepartment of Public Health Sciences, University of California Davis, Davis, California, USADepartment of StatisticsUniversity of California Davis, Davis, California, USADepartment of Biochemistry and Molecular MedicineUniversity of California Davis, Sacramento, California, USADepartment of Medical Microbiology and ImmunologyUniversity of California Davis, Davis, California, USA VA Northern California Health Care SystemMather, California, USADepartment of UrologySchool of Medicine, University of California Davis, 4860 Y Street, Suite 3500, Sacramento, California 95817, USADivision of BiostatisticsDepartment of Public Health Sciences, University of California Davis, Davis, California, USADepartment of StatisticsUniversity of California Davis, Davis, California, USADepartment of Biochemistry and Molecular MedicineUniversity of California Davis, Sacramento, California, USADepartment of Medical Microbiology and ImmunologyUniversity of California Davis, Davis, California, USA VA Northern California Health Care SystemMather, California, USADepartment of UrologySchool of Medicine, University of California Davis, 4860 Y Street, Suite 3500, Sacramento, California 95817, USADivision of BiostatisticsDepartment of Public Health Sciences, University of California Davis, Davis, California, USADepartment of StatisticsUniversity of California Davis, Davis, California, USADepartment of Biochemistry and Molecular MedicineUniversity of California Davis, Sacramento, California, USADepartment of Medical Microbiology and ImmunologyUniversity of California Davis, Davis, California, USA
| |
Collapse
|
124
|
Abstract
PURPOSE OF REVIEW The present review examines the role of actin binding proteins (ABPs) on blood-testis barrier (BTB), an androgen-dependent ultrastructure in the testis, in particular their involvement on BTB remodeling during spermatogenesis. RECENT FINDINGS The BTB divides the seminiferous epithelium into the basal and the adluminal compartments. The BTB is constituted by coexisting actin-based tight junction, basal ectoplasmic specialization, and gap junction, and also intermediate filament-based desmosome between Sertoli cells near the basement membrane. Junctions at the BTB undergo continuous remodeling to facilitate the transport of preleptotene spermatocytes residing in the basal compartment across the immunological barrier during spermatogenesis. Thus, meiosis I/II and postmeiotic spermatid development take place in the adluminal compartment behind the BTB. BTB remodeling also regulates exchanges of biomolecules between the two compartments. As tight junction, basal ectoplasmic specialization, and gap junction use F-actin for attachment, actin microfilaments rapidly convert between their bundled and unbundled/branched configuration to confer BTB plasticity. The events of actin reorganization are regulated by two major classes of ABPs that convert actin microfilaments between their bundled and branched/unbundled configuration. SUMMARY We provide a model on how ABPs regulate BTB remodeling, shedding new light on unexplained male infertility, such as environmental toxicant-induced reproductive dysfunction since the testis, in particular the BTB, is sensitive to environmental toxicants, such as cadmium, bisphenol A, phthalates, and PFOS (perfluorooctanesulfonic acid or perfluorooctane sulfonate).
Collapse
Affiliation(s)
- Nan Li
- The Mary M. Wohlford Laboratory for Male Contraceptive Research, Center for Biomedical Research, Population Council, New York, New York, USA
| | | | | |
Collapse
|
125
|
Yang C, Zhang X, Guo Y, Meng F, Sachs F, Guo J. Mechanical dynamics in live cells and fluorescence-based force/tension sensors. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2015; 1853:1889-904. [PMID: 25958335 DOI: 10.1016/j.bbamcr.2015.05.001] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2014] [Revised: 04/07/2015] [Accepted: 05/01/2015] [Indexed: 01/13/2023]
Abstract
Three signaling systems play the fundamental roles in modulating cell activities: chemical, electrical, and mechanical. While the former two are well studied, the mechanical signaling system is still elusive because of the lack of methods to measure structural forces in real time at cellular and subcellular levels. Indeed, almost all biological processes are responsive to modulation by mechanical forces that trigger dispersive downstream electrical and biochemical pathways. Communication among the three systems is essential to make cells and tissues receptive to environmental changes. Cells have evolved many sophisticated mechanisms for the generation, perception and transduction of mechanical forces, including motor proteins and mechanosensors. In this review, we introduce some background information about mechanical dynamics in live cells, including the ubiquitous mechanical activity, various types of mechanical stimuli exerted on cells and the different mechanosensors. We also summarize recent results obtained using genetically encoded FRET (fluorescence resonance energy transfer)-based force/tension sensors; a new technique used to measure mechanical forces in structural proteins. The sensors have been incorporated into many specific structural proteins and have measured the force gradients in real time within live cells, tissues, and animals.
Collapse
Affiliation(s)
- Chao Yang
- Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing 210029, PR China
| | - Xiaohan Zhang
- Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing 210029, PR China
| | - Yichen Guo
- The University of Alabama, Tuscaloosa, AL, 35401, USA
| | - Fanjie Meng
- Physiology and Biophysics Department, Center for Single Molecule Studies, University at Buffalo, The State University of New York at Buffalo, Buffalo, NY, 14214, USA
| | - Frederick Sachs
- Physiology and Biophysics Department, Center for Single Molecule Studies, University at Buffalo, The State University of New York at Buffalo, Buffalo, NY, 14214, USA
| | - Jun Guo
- Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing 210029, PR China.
| |
Collapse
|
126
|
Wang Q, Zheng W, Wang Z, Yang J, Hussein S, Tang J, Chen XZ. Filamin-a increases the stability and plasma membrane expression of polycystin-2. PLoS One 2015; 10:e0123018. [PMID: 25861040 PMCID: PMC4393133 DOI: 10.1371/journal.pone.0123018] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2014] [Accepted: 02/26/2015] [Indexed: 12/13/2022] Open
Abstract
Polycystin-2 (PC2), encoded by the PKD2 gene, is mutated in ~15% of autosomal dominant polycystic kidney disease. Filamins are actin-binding proteins implicated in scaffolding and membrane stabilization. Here we studied the effects of filamin on PC2 stability using filamin-deficient human melanoma M2, filamin-A (FLNA)-replete A7, HEK293 and IMCD cells together with FLNA siRNA/shRNA knockdown (KD). We found that the presence of FLNA is associated with higher total and plasma membrane PC2 protein expression. Western blotting analysis in combination with FLNA KD showed that FLNA in A7 cells represses PC2 degradation, prolonging the half-life from 2.3 to 4.4 hours. By co-immunoprecipitation and Far Western blotting we found that the FLNA C-terminus (FLNAC) reduces the FLNA-PC2 binding and PC2 expression, presumably through competing with FLNA for binding PC2. We further found that FLNA mediates PC2 binding with actin through forming complex PC2-FLNA-actin. FLNAC acted as a blocking peptide and disrupted the link of PC2 with actin through disrupting the PC2-FLNA-actin complex. Finally, we demonstrated that the physical interaction of PC2-FLNA is Ca-dependent. Taken together, our current study indicates that FLNA anchors PC2 to the actin cytoskeleton through complex PC2-FLNA-actin to reduce degradation and increase stability, and possibly regulate PC2 function in a Ca-dependent manner.
Collapse
Affiliation(s)
- Qian Wang
- Membrane Protein Disease Research Group, Department of Physiology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Wang Zheng
- Membrane Protein Disease Research Group, Department of Physiology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Zuocheng Wang
- Membrane Protein Disease Research Group, Department of Physiology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - JungWoo Yang
- Membrane Protein Disease Research Group, Department of Physiology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Shaimaa Hussein
- Membrane Protein Disease Research Group, Department of Physiology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Jingfeng Tang
- Membrane Protein Disease Research Group, Department of Physiology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
- Membrane Protein Disease and Cancer Research Center, Hubei University of Technology, Wuhan, China
- * E-mail: (XZC); (JFT)
| | - Xing-Zhen Chen
- Membrane Protein Disease Research Group, Department of Physiology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
- Membrane Protein Disease and Cancer Research Center, Hubei University of Technology, Wuhan, China
- * E-mail: (XZC); (JFT)
| |
Collapse
|
127
|
Ithychanda SS, Fang X, Mohan ML, Zhu L, Tirupula KC, Naga Prasad SV, Wang YX, Karnik SS, Qin J. A mechanism of global shape-dependent recognition and phosphorylation of filamin by protein kinase A. J Biol Chem 2015; 290:8527-38. [PMID: 25666618 PMCID: PMC4375502 DOI: 10.1074/jbc.m114.633446] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Revised: 01/27/2015] [Indexed: 12/23/2022] Open
Abstract
Protein phosphorylation mediates essentially all aspects of cellular life. In humans, this is achieved by ∼500 kinases, each recognizing a specific consensus motif (CM) in the substrates. The majority of CMs are surface-exposed and are thought to be accessible to kinases for phosphorylation. Here we investigated the archetypical protein kinase A (PKA)-mediated phosphorylation of filamin, a major cytoskeletal protein that can adopt an autoinhibited conformation. Surprisingly, autoinhibited filamin is refractory to phosphorylation by PKA on a known Ser(2152) site despite its CM being exposed and the corresponding isolated peptide being readily phosphorylated. Structural analysis revealed that although the CM fits into the PKA active site its surrounding regions sterically clash with the kinase. However, upon ligand binding, filamin undergoes a conformational adjustment, allowing rapid phosphorylation on Ser(2152). These data uncover a novel ligand-induced conformational switch to trigger filamin phosphorylation. They further suggest a substrate shape-dependent filtering mechanism that channels specific exposed CM/kinase recognition in diverse signaling responses.
Collapse
Affiliation(s)
- Sujay Subbayya Ithychanda
- From the Department of Molecular Cardiology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195
| | - Xianyang Fang
- Protein-Nucleic Acid Interaction Section, Structural Biophysics Laboratory, NCI, National Institutes of Health, Frederick, Maryland 21702, and
| | - Maradumane L Mohan
- From the Department of Molecular Cardiology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195
| | - Liang Zhu
- From the Department of Molecular Cardiology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195, Department of Biochemistry, Case Western Reserve University, Cleveland, Ohio 44106
| | - Kalyan C Tirupula
- From the Department of Molecular Cardiology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195
| | - Sathyamangla V Naga Prasad
- From the Department of Molecular Cardiology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195
| | - Yun-Xing Wang
- Protein-Nucleic Acid Interaction Section, Structural Biophysics Laboratory, NCI, National Institutes of Health, Frederick, Maryland 21702, and
| | - Sadashiva S Karnik
- From the Department of Molecular Cardiology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195
| | - Jun Qin
- From the Department of Molecular Cardiology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195, Department of Biochemistry, Case Western Reserve University, Cleveland, Ohio 44106
| |
Collapse
|
128
|
Zhang J, Li C, Tang X, Lu Q, Sa R, Zhang H. Proteome changes in the small intestinal mucosa of broilers (Gallus gallus) induced by high concentrations of atmospheric ammonia. Proteome Sci 2015; 13:9. [PMID: 25741220 PMCID: PMC4347970 DOI: 10.1186/s12953-015-0067-4] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2014] [Accepted: 02/11/2015] [Indexed: 01/15/2023] Open
Abstract
Background Ammonia is a well-known toxicant both existing in atmospheric and aquatic system. So far, most studies of ammonia toxicity focused on mammals or aquatic animals. With the development of poultry industry, ammonia as a main source of contaminant in the air is causing more and more problems on broiler production, especially lower growth rate. The molecular mechanisms that underlie the negative effects of ammonia on the growth and intestine of broilers are yet unclear. We investigated the growth, gut morphology, and mucosal proteome of Arbor Acres broilers (Gallus gallus) exposed to high concentrations of atmospheric ammonia by performing a proteomics approach integrated with traditional methods. Results Exposure to ammonia interfered with the development of immune organ and gut villi. Meanwhile, it greatly reduced daily weight gain and feed intake, and enhanced feed conversion ratio. A total of 43 intestinal mucosal proteins were found to be differentially abundant. Up-regulated proteins are related to oxidative phosphorylation and apoptosis. Down-regulated proteins are related to cell structure and growth, transcriptional and translational regulation, immune response, oxidative stress and nutrient metabolism. These results indicated that exposure to ammonia triggered oxidative stress, and interfered with nutrient absorption and immune function in the small intestinal mucosa of broilers. Conclusions These findings have important implications for understanding the toxic mechanisms of ammonia on intestine of broilers, which provides new information that can be used for intervention using nutritional strategies in the future. Electronic supplementary material The online version of this article (doi:10.1186/s12953-015-0067-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jize Zhang
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, 100193 People's Republic of China
| | - Cong Li
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, 100193 People's Republic of China
| | - Xiangfang Tang
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, 100193 People's Republic of China
| | - Qingping Lu
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, 100193 People's Republic of China
| | - Renna Sa
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, 100193 People's Republic of China
| | - Hongfu Zhang
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, 100193 People's Republic of China
| |
Collapse
|
129
|
Garland MA, Stillman JH, Tomanek L. The proteomic response of cheliped myofibril tissue in the eurythermal porcelain crab Petrolisthes cinctipes to heat shock following acclimation to daily temperature fluctuations. J Exp Biol 2015; 218:388-403. [DOI: 10.1242/jeb.112250] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
The porcelain crab Petrolisthes cinctipes lives under rocks and in mussel beds in the mid-intertidal zone where it experiences immersion during high tide and saturating humid conditions in air during low tide, which can increase habitat temperature by up to 20°C. To identify the biochemical changes affected by increasing temperature fluctuations and subsequent heat shock, we acclimated P. cinctipes for 30 days to one of three temperature regimes: (1) constant 10°C, (2) daily temperature fluctuations between 10 and 20°C (5 h up-ramp to 20°C, 1 h down-ramp to 10°C) and (3) 10–30°C (up-ramp to 30°C). After acclimation, animals were exposed to either 10°C or a 30°C heat shock to analyze the proteomic changes in claw muscle tissue. Following acclimation to 10–30°C (measured at 10°C), enolase and ATP synthase increased in abundance. Following heat shock, isoforms of arginine kinase and glycolytic enzymes such as aldolase, triose phosphate isomerase and glyceraldehyde 3-phosphate dehydrogenase increased across all acclimation regimes. Full-length isoforms of hemocyanin increased abundance following acclimation to 10–30°C, but hemocyanin fragments increased after heat shock following constant 10°C and fluctuating 10–20°C, possibly playing a role as antimicrobial peptides. Following constant 10°C and fluctuating 10–20°C, paramyosin and myosin heavy chain type-B increased in abundance, respectively, whereas myosin light and heavy chain decreased with heat shock. Actin-binding proteins, which stabilize actin filaments (filamin and tropomyosin), increased during heat shock following 10–30°C; however, actin severing and depolymerization proteins (gelsolin and cofilin) increased during heat shock following 10–20°C, possibly promoting muscle fiber restructuring. RAF kinase inhibitor protein and prostaglandin reductase increased during heat shock following constant 10°C and fluctuating 10–20°C, possibly inhibiting an immune response during heat shock. The results suggest that ATP supply, muscle fiber restructuring and immune responses are all affected by temperature fluctuations and subsequent acute heat shock in muscle tissue. Furthermore, although heat shock after acclimation to constant 10°C and fluctuating 10–30°C showed the greatest effects on the proteome, moderately fluctuating temperatures (10–20°C) broadened the temperature range over which claw muscle was able to respond to an acute heat shock with limited changes in the muscle proteome.
Collapse
Affiliation(s)
- Michael A. Garland
- California Polytechnic State University, Department of Biological Sciences, Center for Coastal Marine Studies, Environmental Proteomics Laboratory, 1 Grand Avenue, San Luis Obispo, CA 93407-0401, USA
| | - Jonathon H. Stillman
- Romberg Tiburon Center for Environmental Studies, San Francisco State University, 3152 Paradise Drive, Tiburon, CA 94920-1205, USA
| | - Lars Tomanek
- California Polytechnic State University, Department of Biological Sciences, Center for Coastal Marine Studies, Environmental Proteomics Laboratory, 1 Grand Avenue, San Luis Obispo, CA 93407-0401, USA
| |
Collapse
|
130
|
Peverelli E, Treppiedi D, Giardino E, Vitali E, Lania AG, Mantovani G. Dopamine and Somatostatin Analogues Resistance of Pituitary Tumors: Focus on Cytoskeleton Involvement. Front Endocrinol (Lausanne) 2015; 6:187. [PMID: 26733942 PMCID: PMC4686608 DOI: 10.3389/fendo.2015.00187] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Accepted: 12/07/2015] [Indexed: 12/15/2022] Open
Abstract
Pituitary tumors, that origin from excessive proliferation of a specific subtype of pituitary cell, are mostly benign tumors, but may cause significant morbidity in affected patients, including visual and neurologic manifestations from mass-effect, or endocrine syndromes caused by hormone hypersecretion. Dopamine (DA) receptor DRD2 and somatostatin (SS) receptors (SSTRs) represent the main targets of pharmacological treatment of pituitary tumors since they mediate inhibitory effects on both hormone secretion and cell proliferation, and their expression is retained by most of these tumors. Although long-acting DA and SS analogs are currently used in the treatment of prolactin (PRL)- and growth hormone (GH)-secreting pituitary tumors, respectively, clinical practice indicates a great variability in the frequency and entity of favorable responses. The molecular basis of the pharmacological resistance are still poorly understood, and several potential molecular mechanisms have been proposed, including defective expression or genetic alterations of DRD2 and SSTRs, or an impaired signal transduction. Recently, a role for cytoskeleton protein filamin A (FLNA) in DRD2 and SSTRs receptors expression and signaling in PRL- and GH-secreting tumors, respectively, has been demonstrated, first revealing a link between FLNA expression and responsiveness of pituitary tumors to pharmacological therapy. This review provides an overview of the known molecular events involved in SS and DA resistance, focusing on the role played by FLNA.
Collapse
Affiliation(s)
- Erika Peverelli
- Endocrinology and Diabetology Unit, Department of Clinical Sciences and Community Health, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, University of Milan, Milan, Italy
| | - Donatella Treppiedi
- Endocrinology and Diabetology Unit, Department of Clinical Sciences and Community Health, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, University of Milan, Milan, Italy
| | - Elena Giardino
- Endocrinology and Diabetology Unit, Department of Clinical Sciences and Community Health, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, University of Milan, Milan, Italy
| | - Eleonora Vitali
- Laboratory of Cellular and Molecular Endocrinology, IRCCS Clinical and Research Institute Humanitas, Milan, Italy
| | - Andrea G. Lania
- Endocrine Unit, IRCCS Humanitas Clinical Institute, University of Milan, Milan, Italy
| | - Giovanna Mantovani
- Endocrinology and Diabetology Unit, Department of Clinical Sciences and Community Health, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, University of Milan, Milan, Italy
- *Correspondence: Giovanna Mantovani,
| |
Collapse
|
131
|
Janoštiak R, Pataki AC, Brábek J, Rösel D. Mechanosensors in integrin signaling: The emerging role of p130Cas. Eur J Cell Biol 2014; 93:445-54. [DOI: 10.1016/j.ejcb.2014.07.002] [Citation(s) in RCA: 98] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Revised: 06/11/2014] [Accepted: 07/01/2014] [Indexed: 12/17/2022] Open
|
132
|
Sethi R, Ylänne J. Small-angle X-ray scattering reveals compact domain-domain interactions in the N-terminal region of filamin C. PLoS One 2014; 9:e107457. [PMID: 25243668 PMCID: PMC4170960 DOI: 10.1371/journal.pone.0107457] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2014] [Accepted: 08/18/2014] [Indexed: 12/30/2022] Open
Abstract
Filamins are multi-domain, actin cross-linking, and scaffolding proteins. In addition to the actin cross-linking function, filamins have a role in mechanosensor signaling. The mechanosensor function is mediated by domain-domain interaction in the C-terminal region of filamins. Recently, we have shown that there is a three-domain interaction module in the N-terminal region of filamins, where the neighboring domains stabilize the structure of the middle domain and thereby regulate its interaction with ligands. In this study, we have used small-angle X-ray scattering as a tool to screen for potential domain-domain interactions in the N-terminal region. We found evidence of four domain-domain interactions with varying flexibility. These results confirm our previous study showing that domains 3, 4, and 5 exist as a compact three domain module. In addition, we report interactions between domains 11-12 and 14-15, which are thus new candidate sites for mechanical regulation.
Collapse
Affiliation(s)
- Ritika Sethi
- Department of Biological and Environmental Science and Nanoscience Center, University of Jyväskylä, Jyväskylä, Finland
| | - Jari Ylänne
- Department of Biological and Environmental Science and Nanoscience Center, University of Jyväskylä, Jyväskylä, Finland
| |
Collapse
|
133
|
Contrasting transcriptome landscapes of rabbit pluripotent stem cells in vitro and in vivo. Anim Reprod Sci 2014; 149:67-79. [DOI: 10.1016/j.anireprosci.2014.05.014] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2014] [Accepted: 05/26/2014] [Indexed: 01/25/2023]
|
134
|
Maeda E, Tsutsumi T, Kitamura N, Kurokawa T, Ping Gong J, Yasuda K, Ohashi T. Significant increase in Young's modulus of ATDC5 cells during chondrogenic differentiation induced by PAMPS/PDMAAm double-network gel: comparison with induction by insulin. J Biomech 2014; 47:3408-14. [PMID: 25110167 DOI: 10.1016/j.jbiomech.2014.07.021] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2014] [Revised: 07/08/2014] [Accepted: 07/16/2014] [Indexed: 12/31/2022]
Abstract
A double-network (DN) gel, which was composed of poly(2-acrylamido-2-methylpropanesulfonic acid) and poly(N,N'-dimethyl acrylamide) (PAMPS/PDMAAm), has the potential to induce chondrogenesis both in vitro and in vivo. The present study investigated the biomechanical and biological responses of chondrogenic progenitor ATDC5 cells cultured on the DN gel. ATDC5 cells were cultured on a polystyrene surface without insulin (Culture 1) and with insulin (Culture 2), and on the DN gel without insulin (Culture 3). The cultured cells were evaluated using micropipette aspiration for cell Young's modulus and qPCR for gene expression of chondrogenic and actin organization markers on days 3, 7 and 14. On day 3, the cells in Culture 3 formed nodules, in which the cells exhibited an actin cortical layer inside them, and gene expression of type-II collagen, aggrecan, and SOX9 was significantly higher in Culture 3 than Cultures 1 and 2 (p<0.05). Young's modulus in Culture 3 was significantly higher than that in Culture 1 throughout the testing period (p<0.05) and that in Culture 2 on day 14 (p<0.01). There was continuous expression of actin organization markers in Culture 3. This study highlights that the cells on the DN gel increased the modulus and mRNA expression of chondrogenic markers at an earlier time point with a greater magnitude compared to those on the polystyrene surface with insulin. This study also demonstrates a possible strong interrelation among alteration of cell mechanical properties, changes in actin organization and the induction of chondrogenic differentiation.
Collapse
Affiliation(s)
- Eijiro Maeda
- Laboratory of Micro and Nanomechanics, Division of Human Mechanical Systems and Design, Graduate School of Engineering, Hokkaido University, N13 W8, Kita-ku, Sapporo, Hokkaido 060-8628, Japan.
| | - Takehiro Tsutsumi
- Laboratory of Micro and Nanomechanics, Division of Human Mechanical Systems and Design, Graduate School of Engineering, Hokkaido University, N13 W8, Kita-ku, Sapporo, Hokkaido 060-8628, Japan
| | - Nobuto Kitamura
- Department of Sports Medicine and Joint Surgery, Graduate School of Medicine, Hokkaido University, N15 W7, Kita-ku, Sapporo, Hokkaido 060-8638, Japan
| | - Takayuki Kurokawa
- Laboratory of Soft and Wet Matter, Department of Advanced Transdisciplinary Sciences, Faculty of Advanced Life Science, Hokkaido University, N10, W8, Kita-ku, Sapporo, Hokkaido 060-0810, Japan
| | - Jian Ping Gong
- Laboratory of Soft and Wet Matter, Department of Advanced Transdisciplinary Sciences, Faculty of Advanced Life Science, Hokkaido University, N10, W8, Kita-ku, Sapporo, Hokkaido 060-0810, Japan
| | - Kazunori Yasuda
- Department of Sports Medicine and Joint Surgery, Graduate School of Medicine, Hokkaido University, N15 W7, Kita-ku, Sapporo, Hokkaido 060-8638, Japan
| | - Toshiro Ohashi
- Laboratory of Micro and Nanomechanics, Division of Human Mechanical Systems and Design, Graduate School of Engineering, Hokkaido University, N13 W8, Kita-ku, Sapporo, Hokkaido 060-8628, Japan
| |
Collapse
|
135
|
Pavone LM, Norris RA. Distinct signaling pathways activated by "extracellular" and "intracellular" serotonin in heart valve development and disease. Cell Biochem Biophys 2014; 67:819-28. [PMID: 23605455 DOI: 10.1007/s12013-013-9606-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Cardiac valve diseases are often due to developmental anomalies that progressively lead to the abnormal distribution and organization of extracellular matrix proteins overtime. Whereas mechanisms underlying adult valvulopathies are unknown, previous work has shown a critical involvement of the monoamine serotonin in disease pathogenesis. In particular, the interaction of serotonin with its receptors can activate transforming growth factor-β1 (TGF-β1) signaling, which in turn promotes extracellular matrix gene expression. Elevated levels of circulating serotonin can lead to aberrant TGF-β1 signaling with significant effects on cardiac valve structure and function. Additional functions of serotonin have recently been reported in which internalization of serotonin, through the serotonin transporter SERT, can exert important cytoskeletal functions in lieu of simply being degraded. Recent findings demonstrate that intracellular serotonin regulates cardiac valve remodeling, and perturbation of this pathway can also lead to heart valve defects. Thus, both extracellular and intracellular mechanisms of serotonin action appear to be operative in heart valve development, functionality, and disease. This review summarizes some of the salient aspects of serotonin activity during cardiac valve development and disease pathogenesis with an understanding that further elaboration of intracellular and extracellular serotonin pathways may lead to beneficial treatments for heart valve disease.
Collapse
Affiliation(s)
- Luigi Michele Pavone
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Via S. Pansini 5, Naples, 80131, Italy,
| | | |
Collapse
|
136
|
Hiepen C, Benn A, Denkis A, Lukonin I, Weise C, Boergermann JH, Knaus P. BMP2-induced chemotaxis requires PI3K p55γ/p110α-dependent phosphatidylinositol (3,4,5)-triphosphate production and LL5β recruitment at the cytocortex. BMC Biol 2014; 12:43. [PMID: 24885555 PMCID: PMC4071339 DOI: 10.1186/1741-7007-12-43] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2014] [Accepted: 05/13/2014] [Indexed: 01/13/2023] Open
Abstract
Background BMP-induced chemotaxis of mesenchymal progenitors is fundamental for vertebrate development, disease and tissue repair. BMP2 induces Smad and non-Smad signalling. Whereas signal transduction via Smads lead to transcriptional responses, non-Smad signalling induces both, transcriptional and immediate/early non-transcriptional responses. However, the molecular mechanisms by which BMP2 facilitates planar cell polarity, cortical actin rearrangements, lamellipodia formation and chemotaxis of mesenchymal progenitors are poorly understood. Our aim was to uncover the molecular mechanism by which BMP2 facilitates chemotaxis via the BMP2-dependent activation of PI3K and spatiotemporal control of PIP3 production important for actin rearrangements at the mesenchymal cell cytocortex. Results We unveiled the molecular mechanism by which BMP2 induces non-Smad signalling by PI3K and the role of the second messenger PIP3 in BMP2-induced planar cell polarity, cortical actin reorganisation and lamellipodia formation. By using protein interaction studies, we identified the class Ia PI3K regulatory subunit p55γ to act as a specific and non-redundant binding partner for BMP receptor type II (BMPRII) in concert with the catalytic subunit p110α. We mapped the PI3K interaction to a region within the BMPRII kinase. Either BMP2 stimulation or increasing amounts of BMPRI facilitated p55γ association with BMPRII, but BMPRII kinase activity was not required for the interaction. We visualised BMP2-dependent PIP3 production via PI3K p55γ/p110α and were able to localise PIP3 to the leading edge of intact cells during the process of BMP2-induced planar cell polarity and actin dependent lamellipodia formation. Using mass spectrometry, we found the highly PIP3-sensitive PH-domain protein LL5β to act as a novel BMP2 effector in orchestrating cortical actin rearrangements. By use of live cell imaging we found that knock-down of p55γ or LL5β or pharmacological inhibition of PI3K impaired BMP2-induced migratory responses. Conclusions Our results provide evidence for an important contribution of the BMP2-PI3K (p55γ/p110α)- PIP3-LL5β signalling axis in mesenchymal progenitor cell chemotaxis. We demonstrate molecular insights into BMP2-induced PI3K signalling on the level of actin reorganisation at the leading edge cytocortex. These findings are important to better understand BMP2–induced cytoskeletal reorganisation and chemotaxis of mesenchymal progenitors in different physiological or pathophysiological contexts.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Petra Knaus
- Institute for Chemistry and Biochemistry, Freie Universität Βerlin, 14195 Berlin, Germany.
| |
Collapse
|
137
|
Proteomic-based comparison between populations of the Great Scallop, Pecten maximus. J Proteomics 2014; 105:164-73. [PMID: 24704858 DOI: 10.1016/j.jprot.2014.03.026] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2014] [Revised: 03/13/2014] [Accepted: 03/22/2014] [Indexed: 10/25/2022]
Abstract
UNLABELLED Comparing populations residing in contrasting environments is an efficient way to decipher how organisms modulate their physiology. Here we present the proteomic signatures of two populations in a non-model marine species, the great scallop Pecten maximus, living in the northern (Hordaland, Norway) and in the center (Brest, France) of this species' latitudinal distribution range. The results showed 38 protein spots significantly differentially accumulated in mantle tissues between the two populations. We could unambiguously identify 11 of the protein spots by Maldi TOF-TOF mass spectrometry. Eight proteins corresponded to different isoforms of actin, two were identified as filamin, another protein related to the cytoskeleton structure, and one was the protease elastase. Our results suggest that scallops from the two populations assayed may modulate their cytoskeleton structures through regulation of intracellular pools of actin and filamin isoforms to better adapt to their environment. BIOLOGICAL SIGNIFICANCE Marine mollusks are non-model organisms that have been poorly studied at the proteomic level, and this article is the first studying the great scallop (P. maximus) at this level. Furthermore, it addresses population proteomics, a new promising field, especially in environmental sciences. This article is part of a Special Issue entitled: Proteomics of non-model organisms.
Collapse
|
138
|
Leem YH, Lee KS, Kim JH, Seok HK, Chang JS, Lee DH. Magnesium ions facilitate integrin alpha 2- and alpha 3-mediated proliferation and enhance alkaline phosphatase expression and activity in hBMSCs. J Tissue Eng Regen Med 2014; 10:E527-E536. [PMID: 24616281 DOI: 10.1002/term.1861] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2013] [Revised: 09/12/2013] [Accepted: 11/10/2013] [Indexed: 01/03/2023]
Abstract
Magnesium metal and its alloys have been proposed as a novel class of bone implant biomaterials because of their biodegradability and mechanical properties. The purpose of this study was to determine whether magnesium ions, which are released abundantly from alloys, affect proliferation and differentiation of human bone marrow-derived stromal cells (hBMSCs). High levels of magnesium ions did not induce cytotoxicity in hBMSCs, but treatment with 2.5-10 mm magnesium ions for 48-72 h significantly increased hBMSC proliferation. The expression of integrins α2 and α3, but not β1, was upregulated compared with the control and shifted from α3 to α2 in hBMSCs treated with magnesium ions. Knockdown of integrins α2 and/or α3 significantly reduced magnesium-induced proliferation of hBMSCs. Magnesium exposure profoundly enhanced alkaline phosphatase (ALP) gene expression and activity even at a relatively low magnesium concentration (2.5 mm). Exposure to magnesium ions facilitated hBMSC proliferation via integrin α2 and α3 expression and partly promoted differentiation into osteoblasts via the alteration of ALP expression and activity. Accordingly, magnesium could be a useful biomaterial for orthopaedic applications such as bone implant biomaterials for repair and regeneration of bone defects in orthopaedic and dental fields. Copyright © 2014 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Yea-Hyun Leem
- Department of Pharmacy, College of Pharmacy, Dankook University, Cheonan-si, Chungnam, Korea
| | - Kang-Sik Lee
- Department of Orthopedics, Seoul Asan Hospital, Seoul, Korea
| | - Jung-Hwa Kim
- Department of Orthopedics, Seoul Asan Hospital, Seoul, Korea
| | - Hyun-Kwang Seok
- Biomaterial Research Center, Korea Institute of Science and Technology, Seongbuk-Gu, Seoul, Korea
| | - Jae-Suk Chang
- Department of Orthopedics, Seoul Asan Hospital, Seoul, Korea.
| | - Dong-Ho Lee
- Department of Orthopedics, Seoul Asan Hospital, Seoul, Korea.
| |
Collapse
|
139
|
Sethi R, Seppälä J, Tossavainen H, Ylilauri M, Ruskamo S, Pentikäinen OT, Pentikäinen U, Permi P, Ylänne J. A novel structural unit in the N-terminal region of filamins. J Biol Chem 2014; 289:8588-98. [PMID: 24469451 DOI: 10.1074/jbc.m113.537456] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Immunoglobulin-like (Ig) domains are a widely expanded superfamily that act as interaction motifs or as structural spacers in multidomain proteins. Vertebrate filamins (FLNs), which are multifunctional actin-binding proteins, consist of 24 Ig domains. We have recently discovered that in the C-terminal rod 2 region of FLN, Ig domains interact with each other forming functional domain pairs, where the interaction with signaling and transmembrane proteins is mechanically regulated by weak actomyosin contraction forces. Here, we investigated if there are similar inter-domain interactions around domain 4 in the N-terminal rod 1 region of FLN. Protein crystal structures revealed a new type of domain organization between domains 3, 4, and 5. In this module, domains 4 and 5 interact rather tightly, whereas domain 3 has a partially flexible interface with domain 4. NMR peptide titration experiments showed that within the three-domain module, domain 4 is capable for interaction with a peptide derived from platelet glycoprotein Ib. Crystal structures of FLN domains 4 and 5 in complex with the peptide revealed a typical β sheet augmentation interaction observed for many FLN ligands. Domain 5 was found to stabilize domain 4, and this could provide a mechanism for the regulation of domain 4 interactions.
Collapse
Affiliation(s)
- Ritika Sethi
- From the Department of Biological and Environmental Science and Nanoscience Center, University of Jyväskylä, P. O. Box 35, Survontie 9, 40014 Jyväskylä
| | | | | | | | | | | | | | | | | |
Collapse
|
140
|
Savoy RM, Ghosh PM. The dual role of filamin A in cancer: can't live with (too much of) it, can't live without it. Endocr Relat Cancer 2013; 20:R341-56. [PMID: 24108109 PMCID: PMC4376317 DOI: 10.1530/erc-13-0364] [Citation(s) in RCA: 120] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Filamin A (FlnA) has been associated with actin as cytoskeleton regulator. Recently its role in the cell has come under scrutiny for FlnA's involvement in cancer development. FlnA was originally revealed as a cancer-promoting protein, involved in invasion and metastasis. However, recent studies have also found that under certain conditions, it prevented tumor formation or progression, confusing the precise function of FlnA in cancer development. Here, we try to decipher the role of FlnA in cancer and the implications for its dual role. We propose that differences in subcellular localization of FlnA dictate its role in cancer development. In the cytoplasm, FlnA functions in various growth signaling pathways, such as vascular endothelial growth factor, in addition to being involved in cell migration and adhesion pathways, such as R-Ras and integrin signaling. Involvement in these pathways and various others has shown a correlation between high cytoplasmic FlnA levels and invasive cancers. However, an active cleaved form of FlnA can localize to the nucleus rather than the cytoplasm and its interaction with transcription factors has been linked to a decrease in invasiveness of cancers. Therefore, overexpression of FlnA has a tumor-promoting effect, only when it is localized to the cytoplasm, whereas if FlnA undergoes proteolysis and the resulting C-terminal fragment localizes to the nucleus, it acts to suppress tumor growth and inhibit metastasis. Development of drugs to target FlnA and cause cleavage and subsequent localization to the nucleus could be a new and potent field of research in treating cancer.
Collapse
Affiliation(s)
- Rosalinda M Savoy
- Department of Urology, University of California Davis School of Medicine, University of California, 4860 Y Street, Suite 3500, Sacramento, California 95817, USA VA Northern California Health Care System, Mather, California, USA
| | | |
Collapse
|
141
|
Characterization of two ENU-induced mutations affecting mouse skeletal morphology. G3-GENES GENOMES GENETICS 2013; 3:1753-8. [PMID: 23979929 PMCID: PMC3789799 DOI: 10.1534/g3.113.007310] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Using the N-ethyl-N-nitrosourea (ENU) mutagenesis screen, we have identified two skeletal morphology mutants, Skm1 and Skm2. Positional cloning and candidate gene sequencing localized the causative point mutations within the genes coding for natriuretic peptide receptor C (NPR-C) and filamin b (FLNB), respectively. Mice that carry a mutation in Npr3 exhibit a skeletal overgrowth phenotype, resulting in an elongated body and kyphosis. Skm2 mice, carrying a mutation in Flnb, present with scoliosis and lordosis. These mutant mice will serve as useful models for the study of vertebral malformations.
Collapse
|
142
|
Conti V, Carabalona A, Pallesi-Pocachard E, Parrini E, Leventer RJ, Buhler E, McGillivray G, Michel FJ, Striano P, Mei D, Watrin F, Lise S, Pagnamenta AT, Taylor JC, Kini U, Clayton-Smith J, Novara F, Zuffardi O, Dobyns WB, Scheffer IE, Robertson SP, Berkovic SF, Represa A, Keays DA, Cardoso C, Guerrini R. Periventricular heterotopia in 6q terminal deletion syndrome: role of the C6orf70 gene. ACTA ACUST UNITED AC 2013; 136:3378-94. [PMID: 24056535 DOI: 10.1093/brain/awt249] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Periventricular nodular heterotopia is caused by defective neuronal migration that results in heterotopic neuronal nodules lining the lateral ventricles. Mutations in filamin A (FLNA) or ADP-ribosylation factor guanine nucleotide-exchange factor 2 (ARFGEF2) cause periventricular nodular heterotopia, but most patients with this malformation do not have a known aetiology. Using comparative genomic hybridization, we identified 12 patients with developmental brain abnormalities, variably combining periventricular nodular heterotopia, corpus callosum dysgenesis, colpocephaly, cerebellar hypoplasia and polymicrogyria, harbouring a common 1.2 Mb minimal critical deletion in 6q27. These anatomic features were mainly associated with epilepsy, ataxia and cognitive impairment. Using whole exome sequencing in 14 patients with isolated periventricular nodular heterotopia but no copy number variants, we identified one patient with periventricular nodular heterotopia, developmental delay and epilepsy and a de novo missense mutation in the chromosome 6 open reading frame 70 (C6orf70) gene, mapping in the minimal critical deleted region. Using immunohistochemistry and western blots, we demonstrated that in human cell lines, C6orf70 shows primarily a cytoplasmic vesicular puncta-like distribution and that the mutation affects its stability and subcellular distribution. We also performed in utero silencing of C6orf70 and of Phf10 and Dll1, the two additional genes mapping in the 6q27 minimal critical deleted region that are expressed in human and rodent brain. Silencing of C6orf70 in the developing rat neocortex produced periventricular nodular heterotopia that was rescued by concomitant expression of wild-type human C6orf70 protein. Silencing of the contiguous Phf10 or Dll1 genes only produced slightly delayed migration but not periventricular nodular heterotopia. The complex brain phenotype observed in the 6q terminal deletion syndrome likely results from the combined haploinsufficiency of contiguous genes mapping to a small 1.2 Mb region. Our data suggest that, of the genes within this minimal critical region, C6orf70 plays a major role in the control of neuronal migration and its haploinsufficiency or mutation causes periventricular nodular heterotopia.
Collapse
Affiliation(s)
- Valerio Conti
- 1 Paediatric Neurology and Neurogenetics Unit and Laboratories, A. Meyer Children's Hospital - Department of Neuroscience, Pharmacology and Child Health, University of Florence, 50139, Florence, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
143
|
Zakaria R, Lamsoul I, Uttenweiler-Joseph S, Erard M, Monsarrat B, Burlet-Schiltz O, Moog-Lutz C, Lutz PG. Phosphorylation of serine 323 of ASB2α is pivotal for the targeting of filamin A to degradation. Cell Signal 2013; 25:2823-30. [PMID: 24044920 DOI: 10.1016/j.cellsig.2013.09.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2013] [Accepted: 09/06/2013] [Indexed: 10/26/2022]
Abstract
ASB proteins are the specificity subunits of cullin5-RING E3 ubiquitin ligases (CRL5) that play roles in ubiquitin-mediated protein degradation. However, how their activity is regulated remains poorly understood. Here, we unravel a novel mechanism of regulation of a CRL5 through phosphorylation of its specificity subunit ASB2α. Indeed, using mass spectrometry, we showed for the first time that ASB2α is phosphorylated and that phosphorylation of serine-323 (Ser-323) of ASB2α is crucial for the targeting of the actin-binding protein filamin A (FLNa) to degradation. Mutation of ASB2α Ser-323 to Ala had no effect on intrinsic E3 ubiquitin ligase activity of ASB2α but abolished the ability of ASB2α to induce degradation of FLNa. In contrast, the ASB2α Ser-323 to Asp phosphomimetic mutant induced acute degradation of FLNa. Moreover, inhibition of the extracellular signal-regulated kinases 1 and 2 (Erk1/2) activity reduced ASB2α-mediated FLNa degradation. We further showed that the subcellular localization of ASB2α to actin-rich structures is dependent on ASB2α Ser-323 phosphorylation and propose that the interaction with FLNa depends on the electrostatic potential redistribution induced by the Ser-323 phosphate group. Taken together, these data unravel an important mechanism by which ASB2α-mediated FLNa degradation can be regulated.
Collapse
Affiliation(s)
- Rim Zakaria
- CNRS; IPBS (Institut de Pharmacologie et de Biologie Structurale), 205 route de Narbonne BP 64182, F-31077 Toulouse, France; Université de Toulouse, UPS, IPBS, F-31077 Toulouse, France
| | | | | | | | | | | | | | | |
Collapse
|
144
|
Ylilauri M, Pentikäinen OT. MMGBSA as a tool to understand the binding affinities of filamin-peptide interactions. J Chem Inf Model 2013; 53:2626-33. [PMID: 23988151 DOI: 10.1021/ci4002475] [Citation(s) in RCA: 218] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Filamins (FLN) are large dimeric proteins that cross-link actin and work as important scaffolds in human cells. FLNs consist of an N-terminal actin-binding domain followed by 24 immunoglobulin-like domains (FLN1-24). FLN domains are divided into four subgroups based on their amino acid sequences. One of these subgroups, including domains 4, 9, 12, 17, 19, 21, and 23, shares a similar ligand-binding site between the β strands C and D. Several proteins, such as integrins β2 and β7, glycoprotein Ibα (GPIbα), and migfilin, have been shown to bind to this site. Here, we computationally estimated the binding free energies of filamin A (FLNa) subunits with bound peptides using the molecular mechanics-generalized Born surface area (MMGBSA) method. The obtained computational results correlated well with the experimental data, and they ranked efficiently both the binding of one ligand to all used FLNa-domains and the binding of all used ligands to FLNa21. Furthermore, the steered molecular dynamics (SMD) simulations pinpointed the binding hot spots for these complexes. These results demonstrate that molecular dynamics combined with free energy calculations are applicable to estimating the energetics of protein-protein interactions and can be used to direct the development of novel FLN function modulators.
Collapse
Affiliation(s)
- Mikko Ylilauri
- Computational Bioscience Laboratory, Department of Biological and Environmental Science & Nanoscience Center, University of Jyväskylä , P.O. Box 35, Jyväskylä FI-40014, Finland
| | | |
Collapse
|
145
|
Fröhlich T, Kösters M, Graf A, Wolf E, Kobolak J, Brochard V, Dinnyés A, Jouneau A, Arnold GJ. iTRAQ proteome analysis reflects a progressed differentiation state of epiblast derived versus inner cell mass derived murine embryonic stem cells. J Proteomics 2013; 90:38-51. [DOI: 10.1016/j.jprot.2013.03.015] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2012] [Revised: 02/26/2013] [Accepted: 03/19/2013] [Indexed: 12/26/2022]
|
146
|
Kwon MS, Park KR, Kim YD, Na BR, Kim HR, Choi HJ, Piragyte I, Jeon H, Chung KH, Song WK, Eom SH, Jun CD. Swiprosin-1 is a novel actin bundling protein that regulates cell spreading and migration. PLoS One 2013; 8:e71626. [PMID: 23977092 PMCID: PMC3744483 DOI: 10.1371/journal.pone.0071626] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2013] [Accepted: 06/28/2013] [Indexed: 11/18/2022] Open
Abstract
Protein functions are often revealed by their localization to specialized cellular sites. Recent reports demonstrated that swiprosin-1 is found together with actin and actin-binding proteins in the cytoskeleton fraction of human mast cells and NK-like cells. However, direct evidence of whether swiprosin-1 regulates actin dynamics is currently lacking. We found that swiprosin-1 localizes to microvilli-like membrane protrusions and lamellipodia and exhibits actin-binding activity. Overexpression of swiprosin-1 enhanced lamellipodia formation and cell spreading. In contrast, swiprosin-1 knockdown showed reduced cell spreading and migration. Swiprosin-1 induced actin bundling in the presence of Ca(2+), and deletion of the EF-hand motifs partially reduced bundling activity. Swiprosin-1 dimerized in the presence of Ca(2+) via its coiled-coil domain, and a lysine (Lys)-rich region in the coiled-coil domain was essential for regulation of actin bundling. Consistent with these observations, mutations of the EF-hand motifs and coiled-coil region significantly reduced cell spreading and lamellipodia formation. We provide new evidence of how swiprosin-1 influences cytoskeleton reorganization and cell spreading.
Collapse
Affiliation(s)
- Min-Sung Kwon
- School of Life Sciences, Immune Synapse Research Center and Cell Dynamics Research Center, Gwangju Institute of Science and Technology, Gwangju, Korea
| | - Kyoung Ryoung Park
- School of Life Sciences, Immune Synapse Research Center and Cell Dynamics Research Center, Gwangju Institute of Science and Technology, Gwangju, Korea
| | - Young-Dae Kim
- School of Life Sciences, Immune Synapse Research Center and Cell Dynamics Research Center, Gwangju Institute of Science and Technology, Gwangju, Korea
| | - Bo-Ra Na
- School of Life Sciences, Immune Synapse Research Center and Cell Dynamics Research Center, Gwangju Institute of Science and Technology, Gwangju, Korea
| | - Hye-Ran Kim
- School of Life Sciences, Immune Synapse Research Center and Cell Dynamics Research Center, Gwangju Institute of Science and Technology, Gwangju, Korea
| | - Hak-Jong Choi
- School of Life Sciences, Immune Synapse Research Center and Cell Dynamics Research Center, Gwangju Institute of Science and Technology, Gwangju, Korea
| | - Indre Piragyte
- School of Life Sciences, Immune Synapse Research Center and Cell Dynamics Research Center, Gwangju Institute of Science and Technology, Gwangju, Korea
| | - Hyesung Jeon
- Biomedical Research Center, Korea Institute of Science and Technology, Seongbuk-gu, Seoul, Korea
| | - Kyung Hwun Chung
- School of Life Sciences, Immune Synapse Research Center and Cell Dynamics Research Center, Gwangju Institute of Science and Technology, Gwangju, Korea
| | - Woo Keun Song
- School of Life Sciences, Immune Synapse Research Center and Cell Dynamics Research Center, Gwangju Institute of Science and Technology, Gwangju, Korea
| | - Soo Hyun Eom
- School of Life Sciences, Immune Synapse Research Center and Cell Dynamics Research Center, Gwangju Institute of Science and Technology, Gwangju, Korea
| | - Chang-Duk Jun
- School of Life Sciences, Immune Synapse Research Center and Cell Dynamics Research Center, Gwangju Institute of Science and Technology, Gwangju, Korea
| |
Collapse
|
147
|
Sakane A, Alamir Mahmoud Abdallah A, Nakano K, Honda K, Kitamura T, Imoto I, Matsushita N, Sasaki T. Junctional Rab13-binding protein (JRAB) regulates cell spreading via filamins. Genes Cells 2013; 18:810-22. [PMID: 23890175 DOI: 10.1111/gtc.12078] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2013] [Accepted: 05/21/2013] [Indexed: 02/02/2023]
Abstract
We previously showed that Rab13 and its effector protein, junctional Rab13-binding protein (JRAB)/molecules interacting with CasL-like 2 (MICAL-L2), regulate junctional development by modulating cell adhesion molecule transport and actin cytoskeletal reorganization in epithelial cells. Here, we investigated how JRAB regulates reorganization of the actin cytoskeleton in NIH3T3 fibroblasts, in an attempt to obtain novel insights into the mechanism of JRAB action. To this end, we expressed mutant proteins that adopt a constitutively open or closed state and then examined effect on cellular morphology of the resulting actin cytoskeletal reorganization. Expression of the JRABΔCT mutant (constitutively 'closed' state) induced stress fibers, whereas expression of the JRABΔCC mutant (constitutively 'open' state) caused cell spreading with membrane ruffles. Next, we identified the proteins involved in JRAB-induced rearrangement of actin cytoskeleton leading to morphological changes. In NIH3T3 cells expressing HA-JRABΔCC, filamin, an actin cross-linking protein, coimmunoprecipitated with HA-JRABΔCC. Expression of ASB2 induced degradation of all three filamin isoforms and inhibited the JRABΔCC-induced cell spreading. Consistent with our previous results, actinin-1/-4 were also immunoprecipitated with HA-JRABΔCC. However, actinin-1/-4 have no effect on the cell spreading regulated by JRABΔCC. These data suggest that JRAB contributes to the rearrangement of the actin cytoskeleton during cell spreading via filamins.
Collapse
Affiliation(s)
- Ayuko Sakane
- Department of Biochemistry, Institute of Health Biosciences, The University of Tokushima Graduate School, Tokushima, 770-8503, Japan
| | | | | | | | | | | | | | | |
Collapse
|
148
|
Sequeira V, Nijenkamp LLAM, Regan JA, van der Velden J. The physiological role of cardiac cytoskeleton and its alterations in heart failure. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2013; 1838:700-22. [PMID: 23860255 DOI: 10.1016/j.bbamem.2013.07.011] [Citation(s) in RCA: 130] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/06/2013] [Revised: 07/01/2013] [Accepted: 07/08/2013] [Indexed: 12/11/2022]
Abstract
Cardiac muscle cells are equipped with specialized biochemical machineries for the rapid generation of force and movement central to the work generated by the heart. During each heart beat cardiac muscle cells perceive and experience changes in length and load, which reflect one of the fundamental principles of physiology known as the Frank-Starling law of the heart. Cardiac muscle cells are unique mechanical stretch sensors that allow the heart to increase cardiac output, and adjust it to new physiological and pathological situations. In the present review we discuss the mechano-sensory role of the cytoskeletal proteins with respect to their tight interaction with the sarcolemma and extracellular matrix. The role of contractile thick and thin filament proteins, the elastic protein titin, and their anchorage at the Z-disc and M-band, with associated proteins are reviewed in physiologic and pathologic conditions leading to heart failure. This article is part of a Special Issue entitled: Reciprocal influences between cell cytoskeleton and membrane channels, receptors and transporters. Guest Editor: Jean Claude Hervé
Collapse
Affiliation(s)
- Vasco Sequeira
- Laboratory for Physiology, Institute for Cardiovascular Research, VU University Medical Center, van der Boechorststraat 7, 1081 BT Amsterdam, The Netherlands
| | - Louise L A M Nijenkamp
- Laboratory for Physiology, Institute for Cardiovascular Research, VU University Medical Center, van der Boechorststraat 7, 1081 BT Amsterdam, The Netherlands
| | - Jessica A Regan
- Laboratory for Physiology, Institute for Cardiovascular Research, VU University Medical Center, van der Boechorststraat 7, 1081 BT Amsterdam, The Netherlands; Department of Physiology, Molecular Cardiovascular Research Program, Sarver Heart Center, University of Arizona, Tucson, AZ 85724, USA
| | - Jolanda van der Velden
- Laboratory for Physiology, Institute for Cardiovascular Research, VU University Medical Center, van der Boechorststraat 7, 1081 BT Amsterdam, The Netherlands; ICIN-Netherlands Heart Institute, The Netherlands.
| |
Collapse
|
149
|
Filamin and phospholipase C-ε are required for calcium signaling in the Caenorhabditis elegans spermatheca. PLoS Genet 2013; 9:e1003510. [PMID: 23671426 PMCID: PMC3650001 DOI: 10.1371/journal.pgen.1003510] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2012] [Accepted: 03/30/2013] [Indexed: 12/05/2022] Open
Abstract
The Caenorhabditis elegans spermatheca is a myoepithelial tube that stores sperm and undergoes cycles of stretching and constriction as oocytes enter, are fertilized, and exit into the uterus. FLN-1/filamin, a stretch-sensitive structural and signaling scaffold, and PLC-1/phospholipase C-ε, an enzyme that generates the second messenger IP3, are required for embryos to exit normally after fertilization. Using GCaMP, a genetically encoded calcium indicator, we show that entry of an oocyte into the spermatheca initiates a distinctive series of IP3-dependent calcium oscillations that propagate across the tissue via gap junctions and lead to constriction of the spermatheca. PLC-1 is required for the calcium release mechanism triggered by oocyte entry, and FLN-1 is required for timely initiation of the calcium oscillations. INX-12, a gap junction subunit, coordinates propagation of the calcium transients across the spermatheca. Gain-of-function mutations in ITR-1/IP3R, an IP3-dependent calcium channel, and loss-of-function mutations in LFE-2, a negative regulator of IP3 signaling, increase calcium release and suppress the exit defect in filamin-deficient animals. We further demonstrate that a regulatory cassette consisting of MEL-11/myosin phosphatase and NMY-1/non-muscle myosin is required for coordinated contraction of the spermatheca. In summary, this study answers long-standing questions concerning calcium signaling dynamics in the C. elegans spermatheca and suggests FLN-1 is needed in response to oocyte entry to trigger calcium release and coordinated contraction of the spermathecal tissue. During organism development and normal physiological function cells sense, integrate, and respond to a variety of cues or signals including biochemical and mechanical stimuli. In this study we used Caenorhabditis elegans, a small transparent worm, to study filamin (FLN-1), a structural protein that may act as a molecular strain gauge. The C. elegans spermatheca is a contractile tube that is stretched during normal function, making it an ideal candidate for study of how cells respond to stretch. Oocytes are ovulated into the spermatheca, fertilized, and then pushed into the uterus by constriction of the spermatheca. The ability of the spermatheca to constrict depends on inositol 1,4,5-triphosphate (IP3), a signaling molecule produced by the enzyme phospholipase C (PLC-1) that triggers calcium release within cells. In animals with mutated FLN-1 or PLC-1 the spermathecal cells fail to constrict. Using genetic analysis and a calcium-sensitive fluorescent protein, we show that FLN-1 functions with PLC-1 to regulate IP3 production, calcium release, and contraction of the spermatheca. Filamin may function to sense stretch caused by entering oocytes and to trigger constriction. These findings establish a link between filamin and calcium signaling that may apply to similar signaling pathways in other systems.
Collapse
|
150
|
Yue J, Huhn S, Shen Z. Complex roles of filamin-A mediated cytoskeleton network in cancer progression. Cell Biosci 2013; 3:7. [PMID: 23388158 PMCID: PMC3573937 DOI: 10.1186/2045-3701-3-7] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2012] [Accepted: 01/10/2013] [Indexed: 01/08/2023] Open
Abstract
Filamin-A (FLNA), also called actin-binding protein 280 (ABP-280), was originally identified as a non-muscle actin binding protein, which organizes filamentous actin into orthogonal networks and stress fibers. Filamin-A also anchors various transmembrane proteins to the actin cytoskeleton and provides a scaffold for a wide range of cytoplasmic and nuclear signaling proteins. Intriguingly, several studies have revealed that filamin-A associates with multiple non-cytoskeletal proteins of diverse function and is involved in several unrelated pathways. Mutations and aberrant expression of filamin-A have been reported in human genetic diseases and several types of cancer. In this review, we discuss the implications of filamin-A in cancer progression, including metastasis and DNA damage response.
Collapse
Affiliation(s)
- Jingyin Yue
- Department of Radiation Oncology, The Cancer Institute of New Jersey, Robert Wood Johnson Medical School, New Brunswick, NJ, 08903, USA.
| | | | | |
Collapse
|