51
|
Zhou C, Li C, Zhou B, Sun H, Koullourou V, Holt I, Puckelwartz MJ, Warren DT, Hayward R, Lin Z, Zhang L, Morris GE, McNally EM, Shackleton S, Rao L, Shanahan CM, Zhang Q. Novel nesprin-1 mutations associated with dilated cardiomyopathy cause nuclear envelope disruption and defects in myogenesis. Hum Mol Genet 2017; 26:2258-2276. [PMID: 28398466 PMCID: PMC5458344 DOI: 10.1093/hmg/ddx116] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Accepted: 03/22/2017] [Indexed: 02/05/2023] Open
Abstract
Nesprins-1 and -2 are highly expressed in skeletal and cardiac muscle and together with SUN (Sad1p/UNC84)-domain containing proteins and lamin A/C form the LInker of Nucleoskeleton-and-Cytoskeleton (LINC) bridging complex at the nuclear envelope (NE). Mutations in nesprin-1/2 have previously been found in patients with autosomal dominant Emery–Dreifuss muscular dystrophy (EDMD) as well as dilated cardiomyopathy (DCM). In this study, three novel rare variants (R8272Q, S8381C and N8406K) in the C-terminus of the SYNE1 gene (nesprin-1) were identified in seven DCM patients by mutation screening. Expression of these mutants caused nuclear morphology defects and reduced lamin A/C and SUN2 staining at the NE. GST pull-down indicated that nesprin-1/lamin/SUN interactions were disrupted. Nesprin-1 mutations were also associated with augmented activation of the ERK pathway in vitro and in hearts in vivo. During C2C12 muscle cell differentiation, nesprin-1 levels are increased concomitantly with kinesin light chain (KLC-1/2) and immunoprecipitation and GST pull-down showed that these proteins interacted via a recently identified LEWD domain in the C-terminus of nesprin-1. Expression of nesprin-1 mutants in C2C12 cells caused defects in myoblast differentiation and fusion associated with dysregulation of myogenic transcription factors and disruption of the nesprin-1 and KLC-1/2 interaction at the outer nuclear membrane. Expression of nesprin-1α2 WT and mutants in zebrafish embryos caused heart developmental defects that varied in severity. These findings support a role for nesprin-1 in myogenesis and muscle disease, and uncover a novel mechanism whereby disruption of the LINC complex may contribute to the pathogenesis of DCM.
Collapse
Affiliation(s)
- Can Zhou
- King's College London British Heart Foundation Centre of Research Excellence, Cardiovascular Division, London SE5 9NU, UK.,Department of Cardiology, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Chen Li
- King's College London British Heart Foundation Centre of Research Excellence, Cardiovascular Division, London SE5 9NU, UK.,Department of Cardiology, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Bin Zhou
- Laboratory of Molecular Translational Medicine.,Key Laboratory of Obstetric & Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education
| | - Huaqin Sun
- Key Laboratory of Obstetric & Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education.,SCU-CUHK Joint Laboratory for Reproductive Medicine, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
| | - Victoria Koullourou
- King's College London British Heart Foundation Centre of Research Excellence, Cardiovascular Division, London SE5 9NU, UK.,Department of Molecular and Cell Biology, University of Leicester, Leicester LE1 9HN, UK
| | - Ian Holt
- Wolfson Centre for Inherited Neuromuscular Disease, RJAH Orthopaedic Hospital, Oswestry SY10?7AG, UK and Institute for Science and Technology in Medicine, Keele University, ST5?5BG, UK
| | - Megan J Puckelwartz
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Derek T Warren
- King's College London British Heart Foundation Centre of Research Excellence, Cardiovascular Division, London SE5?9NU, UK
| | - Robert Hayward
- King's College London British Heart Foundation Centre of Research Excellence, Cardiovascular Division, London SE5?9NU, UK
| | - Ziyuan Lin
- Key Laboratory of Obstetric & Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education.,SCU-CUHK Joint Laboratory for Reproductive Medicine, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
| | - Lin Zhang
- Laboratory of Molecular Translational Medicine.,Key Laboratory of Obstetric & Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education
| | - Glenn E Morris
- Wolfson Centre for Inherited Neuromuscular Disease, RJAH Orthopaedic Hospital, Oswestry SY10?7AG, UK and Institute for Science and Technology in Medicine, Keele University, ST5?5BG, UK
| | - Elizabeth M McNally
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Sue Shackleton
- Department of Molecular and Cell Biology, University of Leicester, Leicester LE1?9HN, UK
| | - Li Rao
- Department of Cardiology, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Catherine M Shanahan
- King's College London British Heart Foundation Centre of Research Excellence, Cardiovascular Division, London SE5?9NU, UK
| | - Qiuping Zhang
- King's College London British Heart Foundation Centre of Research Excellence, Cardiovascular Division, London SE5?9NU, UK
| |
Collapse
|
52
|
Nesprin-1α-Dependent Microtubule Nucleation from the Nuclear Envelope via Akap450 Is Necessary for Nuclear Positioning in Muscle Cells. Curr Biol 2017; 27:2999-3009.e9. [PMID: 28966089 PMCID: PMC5640514 DOI: 10.1016/j.cub.2017.08.031] [Citation(s) in RCA: 103] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Revised: 08/08/2017] [Accepted: 08/15/2017] [Indexed: 11/20/2022]
Abstract
The nucleus is the main microtubule-organizing center (MTOC) in muscle cells due to the accumulation of centrosomal proteins and microtubule (MT) nucleation activity at the nuclear envelope (NE) [1, 2, 3, 4]. The relocalization of centrosomal proteins, including Pericentrin, Pcm1, and γ-tubulin, depends on Nesprin-1, an outer nuclear membrane (ONM) protein that connects the nucleus to the cytoskeleton via its N-terminal region [5, 6, 7]. Nesprins are also involved in the recruitment of kinesin to the NE and play a role in nuclear positioning in skeletal muscle cells [8, 9, 10, 11, 12]. However, a function for MT nucleation from the NE in nuclear positioning has not been established. Using the proximity-dependent biotin identification (BioID) method [13, 14], we found several centrosomal proteins, including Akap450, Pcm1, and Pericentrin, whose association with Nesprin-1α is increased in differentiated myotubes. We show that Nesprin-1α recruits Akap450 to the NE independently of kinesin and that Akap450, but not other centrosomal proteins, is required for MT nucleation from the NE. Furthermore, we demonstrate that this mechanism is disrupted in congenital muscular dystrophy patient myotubes carrying a nonsense mutation within the SYNE1 gene (23560 G>T) encoding Nesprin-1 [15, 16]. Finally, using computer simulation and cell culture systems, we provide evidence for a role of MT nucleation from the NE on nuclear spreading in myotubes. Our data thus reveal a novel function for Nesprin-1α/Nesprin-1 in nuclear positioning through recruitment of Akap450-mediated MT nucleation activity to the NE. BioID of Nesprin-1α identifies centrosomal proteins at myotube nuclear envelope Nesprin-1α-containing LINC complexes recruit Akap450 to myotube nuclear envelope Akap450 is required for microtubule nucleation at the nuclear envelope Microtubule nucleation at the nuclear envelope is involved in nuclear positioning
Collapse
|
53
|
Stroud MJ, Feng W, Zhang J, Veevers J, Fang X, Gerace L, Chen J. Nesprin 1α2 is essential for mouse postnatal viability and nuclear positioning in skeletal muscle. J Cell Biol 2017; 216:1915-1924. [PMID: 28533284 PMCID: PMC5496623 DOI: 10.1083/jcb.201612128] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2017] [Revised: 03/08/2017] [Accepted: 04/18/2017] [Indexed: 12/13/2022] Open
Abstract
The position of the nucleus in a cell is controlled by interactions between the linker of nucleoskeleton and cytoskeleton (LINC) complex and the cytoskeleton. Defects in nuclear positioning and abnormal aggregation of nuclei occur in many muscle diseases and correlate with muscle dysfunction. Nesprin 1, which includes multiple isoforms, is an integral component of the LINC complex, critical for nuclear positioning and anchorage in skeletal muscle, and is thought to provide an essential link between nuclei and actin. However, previous studies have yet to identify which isoform is responsible. To elucidate this, we generated a series of nesprin 1 mutant mice. We showed that the actin-binding domains of nesprin 1 were dispensable, whereas nesprin 1α2, which lacks actin-binding domains, was crucial for postnatal viability, nuclear positioning, and skeletal muscle function. Furthermore, we revealed that kinesin 1 was displaced in fibers of nesprin 1α2-knockout mice, suggesting that this interaction may play an important role in positioning of myonuclei and functional skeletal muscle.
Collapse
Affiliation(s)
- Matthew J Stroud
- Department of Medicine, University of California, San Diego, La Jolla, CA
| | - Wei Feng
- Department of Medicine, University of California, San Diego, La Jolla, CA
| | - Jianlin Zhang
- Department of Medicine, University of California, San Diego, La Jolla, CA
| | - Jennifer Veevers
- Department of Medicine, University of California, San Diego, La Jolla, CA
| | - Xi Fang
- Department of Medicine, University of California, San Diego, La Jolla, CA
| | - Larry Gerace
- Department of Cell and Molecular Biology, The Scripps Research Institute, La Jolla, CA
| | - Ju Chen
- Department of Medicine, University of California, San Diego, La Jolla, CA
| |
Collapse
|
54
|
Potter C, Zhu W, Razafsky D, Ruzycki P, Kolesnikov AV, Doggett T, Kefalov VJ, Betleja E, Mahjoub MR, Hodzic D. Multiple Isoforms of Nesprin1 Are Integral Components of Ciliary Rootlets. Curr Biol 2017. [PMID: 28625779 DOI: 10.1016/j.cub.2017.05.066] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
SYNE1 (synaptic nuclear envelope 1) encodes multiple isoforms of Nesprin1 (nuclear envelope spectrin 1) that associate with the nuclear envelope (NE) through a C-terminal KASH (Klarsicht/Anc1/Syne homology) domain (Figure 1A) [1-4]. This domain interacts directly with the SUN (Sad1/Unc84) domain of Sun proteins [5-7], a family of transmembrane proteins of the inner nuclear membrane (INM) [8, 9], to form the so-called LINC complexes (linkers of the nucleoskeleton and cytoskeleton) that span the entire NE and mediate nuclear positioning [10-12]. In a stark departure from this classical depiction of Nesprin1 in the context of the NE, we report here that rootletin recruits Nesprin1α at the ciliary rootlets of photoreceptors and identify asymmetric NE aggregates of Nesprin1α and Sun2 that dock filaments of rootletin at the nuclear surface. In NIH 3T3 cells, we show that recombinant rootletin filaments also dock to the NE through the specific recruitment of an ∼600-kDa endogenous isoform of Nesprin1 (Nes1600kDa) and of Sun2. In agreement with the association of Nesprin1α with photoreceptor ciliary rootlets and the functional interaction between rootletin and Nesprin1 in fibroblasts, we demonstrate that multiple isoforms of Nesprin1 are integral components of ciliary rootlets of multiciliated ependymal and tracheal cells. Together, these data provide a novel functional paradigm for Nesprin1 at ciliary rootlets and suggest that the wide spectrum of human pathologies linked to truncating mutations of SYNE1 [13-15] may originate in part from ciliary defects.
Collapse
Affiliation(s)
- Chloe Potter
- Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, 660 S. Euclid Avenue, St. Louis, MO 63110, USA
| | - Wanqiu Zhu
- Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, 660 S. Euclid Avenue, St. Louis, MO 63110, USA
| | - David Razafsky
- Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, 660 S. Euclid Avenue, St. Louis, MO 63110, USA
| | - Philip Ruzycki
- Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, 660 S. Euclid Avenue, St. Louis, MO 63110, USA
| | - Alexander V Kolesnikov
- Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, 660 S. Euclid Avenue, St. Louis, MO 63110, USA
| | - Teresa Doggett
- Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, 660 S. Euclid Avenue, St. Louis, MO 63110, USA
| | - Vladimir J Kefalov
- Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, 660 S. Euclid Avenue, St. Louis, MO 63110, USA
| | - Ewelina Betleja
- Division of Nephrology, Department of Medicine, Washington University School of Medicine, 660 S. Euclid Avenue, St. Louis, MO 63110, USA
| | - Moe R Mahjoub
- Division of Nephrology, Department of Medicine, Washington University School of Medicine, 660 S. Euclid Avenue, St. Louis, MO 63110, USA
| | - Didier Hodzic
- Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, 660 S. Euclid Avenue, St. Louis, MO 63110, USA.
| |
Collapse
|
55
|
Gassó P, Sánchez-Gistau V, Mas S, Sugranyes G, Rodríguez N, Boloc D, de la Serna E, Romero S, Moreno D, Moreno C, Díaz-Caneja CM, Lafuente A, Castro-Fornieles J. Association of CACNA1C and SYNE1 in offspring of patients with psychiatric disorders. Psychiatry Res 2016; 245:427-435. [PMID: 27620326 DOI: 10.1016/j.psychres.2016.08.058] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Revised: 07/29/2016] [Accepted: 08/21/2016] [Indexed: 12/25/2022]
Abstract
Schizophrenia (SZ) and bipolar disorder (BD) are severe mental diseases associated with cognitive impairment, mood disturbance, and psychosis. Both disorders are highly heritable and share a common genetic background. The present study assesses, for the first time, differences in genotype frequencies of polymorphisms located in genes involved in neurodevelopment and synaptic plasticity between genetic high-risk individuals (offspring of patients with SZ or BD; N=100: 31 and 69, respectively) and control subjects (offspring of community controls; N=96). Individuals from both groups had similar ages, around 12 years. A higher percentage of men were included in the genetic high-risk group (58%) compared with the control group (40.6%). A total of 244 validated SNPs located in 35 candidate gene regions were analyzed in 196 participants. Multivariate methods based on logistic regression analysis were performed to assess differences in genotype frequencies. Bonferroni correction was applied for the multiple comparisons performed. Two polymorphisms, CACNA1C rs10848683 and SYNE1 rs214950, showed significant differences. The frequency of heterozygotes for CACNA1C rs10848683 in genetic high-risk individuals was double that in controls (OR=3.15; P=0.00016). For SYNE1 rs214950, higher frequencies of heterozygotes (OR=1.97) and homozygotes for the minor allele (OR=17.89; P=0.00020) were found in the genetic high-risk group than in the control group. In conclusion, polymorphisms in CACNA1C and SYNE1 could confer a greater risk of developing SZ and BD in individuals who are already at high risk because of their family history. This could help identify subjects with a very high genetic risk, in whom early detection and early intervention could lead to better prognosis.
Collapse
Affiliation(s)
- Patricia Gassó
- Department of Pathological Anatomy, Pharmacology and Microbiology, University of Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.
| | - Vanessa Sánchez-Gistau
- Department of Child and Adolescent Psychiatry and Psychology, Institute of Neurosciences, Hospital Clinic of Barcelona, Spain; Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Spain
| | - Sergi Mas
- Department of Pathological Anatomy, Pharmacology and Microbiology, University of Barcelona, Spain; Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Gisela Sugranyes
- Department of Child and Adolescent Psychiatry and Psychology, Institute of Neurosciences, Hospital Clinic of Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Natalia Rodríguez
- Department of Pathological Anatomy, Pharmacology and Microbiology, University of Barcelona, Spain
| | - Daniel Boloc
- Department of Pathological Anatomy, Pharmacology and Microbiology, University of Barcelona, Spain
| | - Elena de la Serna
- Department of Child and Adolescent Psychiatry and Psychology, Institute of Neurosciences, Hospital Clinic of Barcelona, Spain; Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Spain
| | - Soledad Romero
- Department of Child and Adolescent Psychiatry and Psychology, Institute of Neurosciences, Hospital Clinic of Barcelona, Spain; Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Spain
| | - Dolores Moreno
- Child and Adolescent Psychiatry Department, Hospital General Universitario Gregorio Marañón, Madrid, Spain; Department of Psychiatry, Complutense University of Madrid, Spain; Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Spain; Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
| | - Carmen Moreno
- Child and Adolescent Psychiatry Department, Hospital General Universitario Gregorio Marañón, Madrid, Spain; Department of Psychiatry, Complutense University of Madrid, Spain; Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Spain; Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
| | - Covadonga M Díaz-Caneja
- Child and Adolescent Psychiatry Department, Hospital General Universitario Gregorio Marañón, Madrid, Spain; Department of Psychiatry, Complutense University of Madrid, Spain; Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Spain; Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
| | - Amalia Lafuente
- Department of Pathological Anatomy, Pharmacology and Microbiology, University of Barcelona, Spain; Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Josefina Castro-Fornieles
- Department of Child and Adolescent Psychiatry and Psychology, Institute of Neurosciences, Hospital Clinic of Barcelona, Spain; Department of Psychiatry and Clinical Psychobiology, University of Barcelona, Spain; Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| |
Collapse
|
56
|
Thakar K, May CK, Rogers A, Carroll CW. Opposing roles for distinct LINC complexes in regulation of the small GTPase RhoA. Mol Biol Cell 2016; 28:182-191. [PMID: 28035049 PMCID: PMC5221622 DOI: 10.1091/mbc.e16-06-0467] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Revised: 09/20/2016] [Accepted: 11/02/2016] [Indexed: 12/01/2022] Open
Abstract
Different forms of nuclear envelope–spanning LINC complexes have opposing roles in the transcription-independent control of the small GTPase RhoA. Competition between LINC complexes in the nuclear envelope may therefore dictate the outcome of signaling to cytoskeletal networks. Linker of Nucleoskeleton and Cytoskeleton (LINC) complexes span the nuclear envelope and transduce force from dynamic cytoskeletal networks to the nuclear lamina. Here we show that LINC complexes also signal from the nuclear envelope to critical regulators of the actin cytoskeleton. Specifically, we find that LINC complexes that contain the inner nuclear membrane protein Sun2 promote focal adhesion assembly by activating the small GTPase RhoA. A key effector in this process is the transcription factor/coactivator complex composed of SRF/Mkl1. A constitutively active form of SRF/Mkl1 was not sufficient to induce focal adhesion assembly in cells lacking Sun2, however, suggesting that LINC complexes support RhoA activity through a transcription-independent mechanism. Strikingly, we also find that the inner nuclear membrane protein Sun1 antagonizes Sun2 LINC complexes and inhibits RhoA activation and focal adhesion assembly. Thus different LINC complexes have opposing roles in the transcription-independent control of the actin cytoskeleton through the small GTPase RhoA.
Collapse
Affiliation(s)
- Ketan Thakar
- Department of Cell Biology, Yale School of Medicine, New Haven, CT 06520
| | - Christopher K May
- Department of Cell Biology, Yale School of Medicine, New Haven, CT 06520
| | - Anna Rogers
- Department of Cell Biology, Yale School of Medicine, New Haven, CT 06520
| | | |
Collapse
|
57
|
Rajgor D, Hanley JG, Shanahan CM. Identification of novel nesprin-1 binding partners and cytoplasmic matrin-3 in processing bodies. Mol Biol Cell 2016; 27:3894-3902. [PMID: 27733621 PMCID: PMC5170611 DOI: 10.1091/mbc.e16-06-0346] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Revised: 09/20/2016] [Accepted: 10/05/2016] [Indexed: 11/25/2022] Open
Abstract
Several new nesprin-1 binding partners were identified, of which many are well-characterized RNA-binding proteins involved in various forms of nuclear RNA-processing events. Matrin-3 was one such protein identified, and a new cytoplasmic localization for matrin-3 is shown. Nesprins are highly conserved spectrin repeat–containing scaffold proteins predominantly known to function at the nuclear envelope (NE). However, nesprin isoforms are emerging with localizations and scaffolding functions at sites away from the NE, suggesting their functions are more diverse than originally thought. In this study, we combined nesprin-1 coimmunoprecipitations with mass spectrometry to identify novel nesprin-1 binding partners for isoforms that localize to subcellular compartments beyond the NE. We show that one of these interactors, matrin-3 (matr3), localizes to mRNA processing bodies (PBs), where we have previously shown a nesprin-1 isoform to localize. Furthermore, we show that Matr3 is part of PB mRNP complexes, is a regulator of miRNA-mediated gene silencing, and possibly shuttles to stress granules in stressed cells. More importantly, we identify a new C-terminally truncated Matr3 isoform that is likely to be involved in these functions and PB localization. This study highlights several novel nesprin-1 binding partners and a new function and localization for Matr3 in cytoplasmic RNA granules.
Collapse
Affiliation(s)
- Dipen Rajgor
- Cardiovascular Division, King's College London, London SE5 9NU, United Kingdom .,Department of Biochemistry, University of Bristol, Bristol BS8 1TD, United Kingdom
| | - Jonathan G Hanley
- Department of Biochemistry, University of Bristol, Bristol BS8 1TD, United Kingdom
| | | |
Collapse
|
58
|
Wang S, Volk T. Composite biopolymer scaffolds shape muscle nucleus: Insights and perspectives from Drosophila. BIOARCHITECTURE 2016; 5:35-43. [PMID: 26605802 DOI: 10.1080/19490992.2015.1106061] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Contractile muscle fibers produce enormous intrinsic forces during contraction/relaxation waves. These forces are directly applied to their cytoplasmic organelles including mitochondria, sarcoplasmic reticulum, and multiple nuclei. Data from our analysis of Drosophila larval somatic muscle fibers suggest that an intricate network of organized microtubules (MT) intermingled with Spectrin-Repeat-Containing Proteins (SRCPs) are major structural elements that protect muscle organelles and maintain their structure and position during muscle contraction. Whereas the perinuclear MT network provides structural rigidity to the myonucleus, the SRCPs Nesprin and Spectraplakin form semiflexible filamentous biopolymer networks, providing nuclei with the elasticity required to resist the contractile cytoplasmic forces produced by the muscle. Spectrin repeats are domains found in numerous structural proteins, which are able to unfold under tension and are subject to mechanical stresses in the cell. This unique composite scaffold combines rigidity and resilience in order to neutralize the oscillating cellular forces occurring during muscle contraction/relaxation waves and thereby protect myonuclei. We suggest that the elastic properties of SRCPs are critical for nuclear protection and proper function in muscle fibers.
Collapse
Affiliation(s)
- Shuoshuo Wang
- a Department of Molecular Genetics ; Weizmann Institute of Science ; Rehovot , Israel
| | - Talila Volk
- a Department of Molecular Genetics ; Weizmann Institute of Science ; Rehovot , Israel
| |
Collapse
|
59
|
Zhang Q, Minaisah RM, Ferraro E, Li C, Porter LJ, Zhou C, Gao F, Zhang J, Rajgor D, Autore F, Shanahan CM, Warren DT. N-terminal nesprin-2 variants regulate β-catenin signalling. Exp Cell Res 2016; 345:168-79. [PMID: 27321956 PMCID: PMC4948682 DOI: 10.1016/j.yexcr.2016.06.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Revised: 06/13/2016] [Accepted: 06/14/2016] [Indexed: 01/14/2023]
Abstract
The spatial compartmentalisation of biochemical signalling pathways is essential for cell function. Nesprins are a multi-isomeric family of proteins that have emerged as signalling scaffolds, herein, we investigate the localisation and function of novel nesprin-2 N-terminal variants. We show that these nesprin-2 variants display cell specific distribution and reside in both the cytoplasm and nucleus. Immunofluorescence microscopy revealed that nesprin-2 N-terminal variants colocalised with β-catenin at cell-cell junctions in U2OS cells. Calcium switch assays demonstrated that nesprin-2 and β-catenin are lost from cell-cell junctions in low calcium conditions whereas emerin localisation at the NE remained unaltered, furthermore, an N-terminal fragment of nesprin-2 was sufficient for cell-cell junction localisation and interacted with β-catenin. Disruption of these N-terminal nesprin-2 variants, using siRNA depletion resulted in loss of β-catenin from cell-cell junctions, nuclear accumulation of active β-catenin and augmented β-catenin transcriptional activity. Importantly, we show that U2OS cells lack nesprin-2 giant, suggesting that the N-terminal nesprin-2 variants regulate β-catenin signalling independently of the NE. Together, these data identify N-terminal nesprin-2 variants as novel regulators of β-catenin signalling that tether β-catenin to cell-cell contacts to inhibit β-catenin transcriptional activity.
Collapse
Affiliation(s)
- Qiuping Zhang
- British Heart Foundation Centre of Research Excellence, Cardiovascular Division, King's College, SE5 9NU London, UK
| | - Rose-Marie Minaisah
- British Heart Foundation Centre of Research Excellence, Cardiovascular Division, King's College, SE5 9NU London, UK
| | - Elisa Ferraro
- British Heart Foundation Centre of Research Excellence, Cardiovascular Division, King's College, SE5 9NU London, UK
| | - Chen Li
- British Heart Foundation Centre of Research Excellence, Cardiovascular Division, King's College, SE5 9NU London, UK
| | - Lauren J Porter
- British Heart Foundation Centre of Research Excellence, Cardiovascular Division, King's College, SE5 9NU London, UK
| | - Can Zhou
- British Heart Foundation Centre of Research Excellence, Cardiovascular Division, King's College, SE5 9NU London, UK
| | - Fang Gao
- British Heart Foundation Centre of Research Excellence, Cardiovascular Division, King's College, SE5 9NU London, UK
| | - Junyi Zhang
- British Heart Foundation Centre of Research Excellence, Cardiovascular Division, King's College, SE5 9NU London, UK
| | - Dipen Rajgor
- British Heart Foundation Centre of Research Excellence, Cardiovascular Division, King's College, SE5 9NU London, UK
| | - Flavia Autore
- British Heart Foundation Centre of Research Excellence, Cardiovascular Division, King's College, SE5 9NU London, UK
| | - Catherine M Shanahan
- British Heart Foundation Centre of Research Excellence, Cardiovascular Division, King's College, SE5 9NU London, UK
| | - Derek T Warren
- British Heart Foundation Centre of Research Excellence, Cardiovascular Division, King's College, SE5 9NU London, UK.
| |
Collapse
|
60
|
Specific localization of nesprin-1-α2, the short isoform of nesprin-1 with a KASH domain, in developing, fetal and regenerating muscle, using a new monoclonal antibody. BMC Cell Biol 2016; 17:26. [PMID: 27350129 PMCID: PMC4924313 DOI: 10.1186/s12860-016-0105-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Accepted: 06/20/2016] [Indexed: 01/25/2023] Open
Abstract
Background Nesprin-1-giant (1008kD) is a protein of the outer nuclear membrane that links nuclei to the actin cytoskeleton via amino-terminal calponin homology domains. The short nesprin-1 isoform, nesprin-1-α2, is present only in skeletal and cardiac muscle and several pathogenic mutations occur within it, but the functions of this short isoform without calponin homology domains are unclear. The aim of this study was to determine mRNA levels and protein localization of nesprin-1-α2 at different stages of muscle development in order to shed light on its functions. Results mRNA levels of all known nesprin-1 isoforms with a KASH domain were determined by quantitative PCR. The mRNA for the 111 kD muscle-specific short isoform, nesprin-1-α2, was not detected in pre-differentiation human myoblasts but was present at significant levels in multinucleate myotubes. We developed a monoclonal antibody against the unique amino-terminal sequence of nesprin-1-α2, enabling specific immunolocalization for the first time. Nesprin-1-α2 protein was undetectable in pre-differentiation myoblasts but appeared at the nuclear rim in post-mitotic, multinucleate myotubes and reached its highest levels in fetal muscle. In muscle from a Duchenne muscular dystrophy biopsy, nesprin-1-α2 protein was detected mainly in regenerating fibres expressing neonatal myosin. Nesprin-1-giant was present at all developmental stages, but was also highest in fetal and regenerating fibres. In fetal muscle, both isoforms were present in the cytoplasm, as well as at the nuclear rim. A pathogenic early stop codon (E7854X) in nesprin-1 caused reduced mRNA levels and loss of protein levels of both nesprin-1-giant and (unexpectedly) nesprin-1-α2, but did not affect myogenesis in vitro. Conclusions Nesprin-1-α2 mRNA and protein expression is switched on during myogenesis, alongside other known markers of muscle differentiation. The results show that nesprin-1-α2 is dynamically controlled and may be involved in some process occurring during early myofibre formation, such as re-positioning of nuclei.
Collapse
|
61
|
Mechanotransduction and nuclear function. Curr Opin Cell Biol 2016; 40:98-105. [PMID: 27018929 DOI: 10.1016/j.ceb.2016.03.006] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Revised: 02/16/2016] [Accepted: 03/08/2016] [Indexed: 12/23/2022]
Abstract
Many signaling pathways converge on the nucleus to regulate crucial nuclear events such as transcription, DNA replication and cell cycle progression. Although the vast majority of research in this area has focused on signals generated in response to hormones or other soluble factors, the nucleus also responds to mechanical forces. During the past decade or so, much has been learned about how mechanical force can affect transcription, as well as the growth and differentiation of cells. Much has also been learned about how force is transmitted via the cytoskeleton to the nucleus and then across the nuclear envelope to the nuclear lamina and chromatin. In this brief review, we focus on some of the key proteins that transmit mechanical signals across the nuclear envelope.
Collapse
|
62
|
Abstract
Mechanoresponses in mesenchymal stem cells (MSCs) guide both differentiation and function. In this review, we focus on advances in0 our understanding of how the cytoplasmic cytoskeleton, nuclear envelope and nucleoskeleton, which are connected via LINC (Linker of Nucleoskeleton and Cytoskeleton) complexes, are emerging as an integrated dynamic signaling platform to regulate MSC mechanobiology. This dynamic interconnectivity affects mechanical signaling and transfer of signals into the nucleus. In this way, nuclear and LINC-mediated cytoskeletal connectivity play a critical role in maintaining mechanical signaling that affects MSC fate by serving as both mechanosensory and mechanoresponsive structures. We review disease and age related compromises of LINC complexes and nucleoskeleton that contribute to the etiology of musculoskeletal diseases. Finally we invite the idea that acquired dysfunctions of LINC might be a contributing factor to conditions such as aging, microgravity and osteoporosis and discuss potential mechanical strategies to modulate LINC connectivity to combat these conditions.
Collapse
|
63
|
Abstract
Nesprins are a family of multi-isomeric scaffolding proteins that were originally identified at the nuclear envelope (NE), where they bind to lamin A/C, emerin, and SUN-domain containing proteins, to form the LInker of Nucleoskeleton-and-Cytoskeleton (LINC) complex that connects the NE to the actin cytoskeleton. However, nesprin genes also give rise to a variety of tissue-specific variants of different sizes with potential roles beyond the NE. These variants are generated through alternative initiation, termination, and splicing, which makes nesprin biology very complex to study due to the difficulty in generating specific antibodies and/or short interfering RNAs (siRNA) to particular isoforms. In order to distinguish genuine nesprin variants and eliminate confusion with degradation products of larger nesprin isoforms, in this chapter we discuss methods including 5' and 3' Rapid Amplification of cDNA Ends (RACE) and RT-PCR in combination with EST database searching, for identifying and validating putative nesprin isoforms. This information is essential to allow a better understanding of nesprin functions in different cell types.
Collapse
|
64
|
Loebrich S, Rathje M, Hager E, Ataman B, Harmin DA, Greenberg ME, Nedivi E. Genomic mapping and cellular expression of human CPG2 transcripts in the SYNE1 gene. Mol Cell Neurosci 2015; 71:46-55. [PMID: 26704904 DOI: 10.1016/j.mcn.2015.12.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Revised: 12/11/2015] [Accepted: 12/14/2015] [Indexed: 01/13/2023] Open
Abstract
Bipolar disorder (BD) is a prevalent and severe mood disorder characterized by recurrent episodes of mania and depression. Both genetic and environmental factors have been implicated in BD etiology, but the biological underpinnings remain elusive. Recent genome-wide association studies (GWAS) for identifying genes conferring risk for schizophrenia, BD, and major depression, identified an association between single-nucleotide polymorphisms (SNPs) in the SYNE1 gene and increased risk of BD. SYNE1 has also been identified as a risk locus for multiple other neurological or neuromuscular genetic disorders. The BD associated SNPs map within the gene region homologous to part of rat Syne1 encompassing the brain specific transcripts encoding CPG2, a postsynaptic neuronal protein localized to excitatory synapses and an important regulator of glutamate receptor internalization. Here, we use RNA-seq, ChIP-seq and RACE to map the human SYNE1 transcriptome, focusing on the CPG2 locus. We validate several CPG2 transcripts, including ones not previously annotated in public databases, and identify and clone a full-length CPG2 cDNA expressed in human neocortex, hippocampus and striatum. Using lenti-viral gene knock down/replacement and surface receptor internalization assays, we demonstrate that human CPG2 protein localizes to dendritic spines in rat hippocampal neurons and is functionally equivalent to rat CPG2 in regulating glutamate receptor internalization. This study provides a valuable gene-mapping framework for relating multiple genetic disease loci in SYNE1 with their transcripts, and for evaluating the effects of missense SNPs identified by patient genome sequencing on neuronal function.
Collapse
Affiliation(s)
- Sven Loebrich
- The Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Mette Rathje
- The Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Emily Hager
- The Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Bulent Ataman
- Department of Neurobiology, Harvard Medical School, Boston, MA 02114, USA
| | - David A Harmin
- Department of Neurobiology, Harvard Medical School, Boston, MA 02114, USA
| | | | - Elly Nedivi
- The Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| |
Collapse
|
65
|
Kim DI, Birendra KC, Roux KJ. Making the LINC: SUN and KASH protein interactions. Biol Chem 2015; 396:295-310. [PMID: 25720065 DOI: 10.1515/hsz-2014-0267] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Accepted: 01/20/2015] [Indexed: 01/15/2023]
Abstract
Cell nuclei are physically integrated with the cytoskeleton through the linker of nucleoskeleton and cytoskeleton (LINC) complex, a structure that spans the nuclear envelope to link the nucleoskeleton and cytoskeleton. Outer nuclear membrane KASH domain proteins and inner nuclear membrane SUN domain proteins interact to form the core of the LINC complex. In this review, we provide a comprehensive analysis of the reported protein-protein interactions for KASH and SUN domain proteins. This critical structure, directly connecting the genome with the rest of the cell, contributes to a myriad of cellular functions and, when perturbed, is associated with human disease.
Collapse
|
66
|
WANG ZHAOHUI, ZHAO ZHIJUN, XU KANG, SUN GUOBING, SONG LIN, YIN HONGXIANG, CHEN XIAOQI. Hereditary protein S deficiency leads to ischemic stroke. Mol Med Rep 2015; 12:3279-3284. [PMID: 25997409 PMCID: PMC4526054 DOI: 10.3892/mmr.2015.3793] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2014] [Accepted: 04/10/2015] [Indexed: 11/05/2022] Open
Abstract
Hereditary protein S (PS) deficiency is an independent risk factor for venous thromboembolism. However, the correlation between PS and arterial thrombotic disease, such as cerebral thrombosis, is not clear. The present study focused on the molecular mechanisms underlying ischemic stroke caused by a PS gene mutation in one family. The activity of antithrombin, protein C and PS in the plasma of the proband was measured, and the genes encoding PS were amplified and sequenced. The cellular localization and expression of PS were analyzed in HEK‑293 cells. The proband was a 50‑year‑old male. Plasma PS activity of the proband was 38.9%, which was significantly decreased compared with normal levels. Sequencing analysis revealed a PROS1 c.1486_1490delGATTA mutation on exon 12. This frameshift mutation converts Asp496 in the precursor PS into the termination codon. In addition, the PROS1 mutation was correlated with low PS activity in the family. Functional tests revealed that the mutant protein aggregated in the cytoplasm and its secretion and expression decreased. In conclusion, protein S mutation appeared to be the primary cause of thrombosis in the family of the present study. However, the correlation between PS deficiency and ischemic stroke requires further investigation.
Collapse
Affiliation(s)
- ZHAO-HUI WANG
- Department of Neurology, Hanyang Hospital, Wuhan University of Science and Technology, Wuhan, Hubei 430050, P.R. China
| | - ZHI-JUN ZHAO
- Department of Ultrasonic Diagnosis and Interventional Therapy, The First Hospital Affiliated to Henan University, Kaifeng, Henan 475001, P.R. China
| | - KANG XU
- Department of Neurology, Xinhua Hospital of Hubei, Wuhan, Hubei 430015, P.R. China
| | - GUO-BING SUN
- Department of Neurology, Xinhua Hospital of Hubei, Wuhan, Hubei 430015, P.R. China
| | - LIN SONG
- Department of Neurology, Xinhua Hospital of Hubei, Wuhan, Hubei 430015, P.R. China
| | - HONG-XIANG YIN
- Department of Neurology, Xinhua Hospital of Hubei, Wuhan, Hubei 430015, P.R. China
| | - XIAO-QI CHEN
- Department of Neurology, Xinhua Hospital of Hubei, Wuhan, Hubei 430015, P.R. China
- Correspondence to: Mr. Xiao-Qi Chen, Department of Neurology, Xinhua Hospital of Hubei, 11 Lingjiaohu Road, Wuhan, Hubei 430015, P.R. China, E-mail:
| |
Collapse
|
67
|
Razafsky D, Hodzic D. Nuclear envelope: positioning nuclei and organizing synapses. Curr Opin Cell Biol 2015; 34:84-93. [PMID: 26079712 DOI: 10.1016/j.ceb.2015.06.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Revised: 05/28/2015] [Accepted: 06/01/2015] [Indexed: 10/23/2022]
Abstract
The nuclear envelope plays an essential role in nuclear positioning within cells and tissues. This review highlights advances in understanding the mechanisms of nuclear positioning during skeletal muscle and central nervous system development. New findings, particularly about A-type lamins and Nesprin1, may link nuclear envelope integrity to synaptic integrity. Thus synaptic defects, rather than nuclear mispositioning, may underlie human pathologies associated with mutations of nuclear envelope proteins.
Collapse
Affiliation(s)
- David Razafsky
- Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, 660 S. Euclid Avenue, St Louis, MO 63110, USA
| | - Didier Hodzic
- Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, 660 S. Euclid Avenue, St Louis, MO 63110, USA.
| |
Collapse
|
68
|
Worman HJ, Schirmer EC. Nuclear membrane diversity: underlying tissue-specific pathologies in disease? Curr Opin Cell Biol 2015; 34:101-12. [PMID: 26115475 PMCID: PMC4522394 DOI: 10.1016/j.ceb.2015.06.003] [Citation(s) in RCA: 82] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2014] [Revised: 06/05/2015] [Accepted: 06/10/2015] [Indexed: 11/16/2022]
Abstract
Human 'laminopathy' diseases result from mutations in genes encoding nuclear lamins or nuclear envelope (NE) transmembrane proteins (NETs). These diseases present a seeming paradox: the mutated proteins are widely expressed yet pathology is limited to specific tissues. New findings suggest tissue-specific pathologies arise because these widely expressed proteins act in various complexes that include tissue-specific components. Diverse mechanisms to achieve NE tissue-specificity include tissue-specific regulation of the expression, mRNA splicing, signaling, NE-localization and interactions of potentially hundreds of tissue-specific NETs. New findings suggest these NETs underlie tissue-specific NE roles in cytoskeletal mechanics, cell-cycle regulation, signaling, gene expression and genome organization. This view of the NE as 'specialized' in each cell type is important to understand the tissue-specific pathology of NE-linked diseases.
Collapse
Affiliation(s)
- Howard J Worman
- Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, USA; Department of Pathology and Cell Biology, College of Physicians and Surgeons, Columbia University, New York, USA
| | - Eric C Schirmer
- Wellcome Trust Centre for Cell Biology, University of Edinburgh, Edinburgh, UK.
| |
Collapse
|
69
|
Wang JY, Yu IS, Huang CC, Chen CY, Wang WP, Lin SW, Jeang KT, Chi YH. Sun1 deficiency leads to cerebellar ataxia in mice. Dis Model Mech 2015; 8:957-67. [PMID: 26035387 PMCID: PMC4527285 DOI: 10.1242/dmm.019240] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Accepted: 04/23/2015] [Indexed: 01/22/2023] Open
Abstract
Migration and organization of the nucleus are essential for the proliferation and differentiation of cells, including neurons. However, the relationship between the positioning of the nucleus and cellular morphogenesis remains poorly understood. Inherited recessive cerebellar ataxia has been attributed to mutations in SYNE1, a component of the linker of nucleoskeleton and cytoskeleton (LINC) complex. Regardless, Syne1-mutant mice present with normal cerebellar development. The Sad1-Unc-84 homology (SUN)-domain proteins are located at the inner nuclear membrane and recruit Syne proteins through the KASH domain to the outer nuclear membrane. Here, we report an unrecognized contribution of Sun1 and Sun2 to the postnatal development of murine cerebellum. Mice depleted of Sun1 showed a marked reduction in the cerebellar volume, and this phenotype is exacerbated with additional loss of a Sun2 allele. Consistent with these histological changes, Sun1(-/-) and Sun1(-/-)Sun2(+/-) mice exhibited defective motor coordination. Results of immunohistochemical analyses suggested that Sun1 is highly expressed in Purkinje cells and recruits Syne2 to the periphery of the nucleus. Approximately 33% of Purkinje cells in Sun1(-/-) mice and 66% of Purkinje cells in Sun1(-/-)Sun2(+/-) mice were absent from the surface of the internal granule layer (IGL), whereas the proliferation and migration of granule neurons were unaffected. Furthermore, the Sun1(-/-)Sun2(+/-) Purkinje cells exhibited retarded primary dendrite specification, reduced dendritic complexity and aberrant patterning of synapses. Our findings reveal a cell-type-specific role for Sun1 and Sun2 in nucleokinesis during cerebellar development, and we propose the use of Sun-deficient mice as a model for studying cerebellar ataxia that is associated with mutation of human SYNE genes or loss of Purkinje cells.
Collapse
Affiliation(s)
- Jing-Ya Wang
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Miaoli County 35053, Taiwan
| | - I-Shing Yu
- Department of Clinical Laboratory Sciences and Medical Biotechnology, National Taiwan University Hospital, Taipei 10048, Taiwan Center of Genomic Medicine, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei 10048, Taiwan
| | - Chien-Chi Huang
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Zhunan, Miaoli County 35053, Taiwan
| | - Chia-Yen Chen
- Laboratory of Molecular Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Wan-Ping Wang
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Miaoli County 35053, Taiwan
| | - Shu-Wha Lin
- Department of Clinical Laboratory Sciences and Medical Biotechnology, National Taiwan University Hospital, Taipei 10048, Taiwan Center of Genomic Medicine, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei 10048, Taiwan Department of Laboratory Medicine, National Taiwan University Hospital, Taipei 10048, Taiwan
| | - Kuan-Teh Jeang
- Laboratory of Molecular Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Ya-Hui Chi
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Miaoli County 35053, Taiwan Graduate Institute of Basic Medical Science, China Medical University, Taichung 40402, Taiwan
| |
Collapse
|
70
|
Razafsky D, Hodzic D. A variant of Nesprin1 giant devoid of KASH domain underlies the molecular etiology of autosomal recessive cerebellar ataxia type I. Neurobiol Dis 2015; 78:57-67. [PMID: 25843669 DOI: 10.1016/j.nbd.2015.03.027] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Revised: 03/24/2015] [Accepted: 03/26/2015] [Indexed: 12/21/2022] Open
Abstract
Nonsense mutations across the whole coding sequence of Syne1/Nesprin1 have been linked to autosomal recessive cerebellar ataxia Type I (ARCA1). However, nothing is known about the molecular etiology of this late-onset debilitating pathology. In this work, we report that Nesprin1 giant is specifically expressed in CNS tissues. We also identified a CNS-specific splicing event that leads to the abundant expression of a KASH-LESS variant of Nesprin1 giant (KLNes1g) in the cerebellum. KLNes1g displayed a noncanonical localization at glomeruli of cerebellar mossy fibers whereas Nesprin2 exclusively decorated the nuclear envelope of all cerebellar neurons. In immunogold electron microscopy, KLNes1g colocalized both with synaptic vesicles within mossy fibers and with dendritic membranes of cerebellar granule neurons. We further identified vesicle- and membrane-associated proteins in KLNes1g immunoprecipitates. Together, our results suggest that the loss of function of KLNes1g resulting from Nesprin1 nonsense mutations underlies the molecular etiology of ARCA1.
Collapse
Affiliation(s)
- David Razafsky
- Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, 660 S. Euclid, St Louis, MO 63110, USA
| | - Didier Hodzic
- Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, 660 S. Euclid, St Louis, MO 63110, USA.
| |
Collapse
|
71
|
Chang W, Worman HJ, Gundersen GG. Accessorizing and anchoring the LINC complex for multifunctionality. ACTA ACUST UNITED AC 2015; 208:11-22. [PMID: 25559183 PMCID: PMC4284225 DOI: 10.1083/jcb.201409047] [Citation(s) in RCA: 214] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The linker of nucleoskeleton and cytoskeleton (LINC) complex, composed of outer and inner nuclear membrane Klarsicht, ANC-1, and Syne homology (KASH) and Sad1 and UNC-84 (SUN) proteins, respectively, connects the nucleus to cytoskeletal filaments and performs diverse functions including nuclear positioning, mechanotransduction, and meiotic chromosome movements. Recent studies have shed light on the source of this diversity by identifying factors associated with the complex that endow specific functions as well as those that differentially anchor the complex within the nucleus. Additional diversity may be provided by accessory factors that reorganize the complex into higher-ordered arrays. As core components of the LINC complex are associated with several diseases, understanding the role of accessory and anchoring proteins could provide insights into pathogenic mechanisms.
Collapse
Affiliation(s)
- Wakam Chang
- Department of Pathology and Cell Biology and Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, NY 10032
| | - Howard J Worman
- Department of Pathology and Cell Biology and Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, NY 10032 Department of Pathology and Cell Biology and Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, NY 10032
| | - Gregg G Gundersen
- Department of Pathology and Cell Biology and Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, NY 10032
| |
Collapse
|
72
|
Warren DT, Tajsic T, Porter LJ, Minaisah RM, Cobb A, Jacob A, Rajgor D, Zhang QP, Shanahan CM. Nesprin-2-dependent ERK1/2 compartmentalisation regulates the DNA damage response in vascular smooth muscle cell ageing. Cell Death Differ 2015; 22:1540-50. [PMID: 25744025 PMCID: PMC4532777 DOI: 10.1038/cdd.2015.12] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2014] [Revised: 12/19/2014] [Accepted: 01/21/2015] [Indexed: 11/30/2022] Open
Abstract
Prelamin A accumulation and persistent DNA damage response (DDR) are hallmarks of vascular smooth muscle cell (VSMC) ageing and dysfunction. Although prelamin A is proposed to interfere with DNA repair, our understanding of the crosstalk between prelamin A and the repair process remains limited. The extracellular signal-regulated kinases 1 and 2 (ERK1/2) have emerged as key players in the DDR and are known to enhance ataxia telangiectasia-mutated protein (ATM) activity at DNA lesions, and in this study, we identified a novel relationship between prelamin A accumulation and ERK1/2 nuclear compartmentalisation during VSMC ageing. We show both prelamin A accumulation and increased DNA damage occur concomitantly, before VSMC replicative senescence, and induce the localisation of ERK1/2 to promyelocytic leukaemia protein nuclear bodies (PML NBs) at the sites of DNA damage via nesprin-2 and lamin A interactions. Importantly, VSMCs treated with DNA damaging agents also displayed prelamin A accumulation and ERK compartmentalisation at PML NBs, suggesting that prelamin A and nesprin-2 are novel components of the DDR. In support of this, disruption of ERK compartmentalisation at PML NBs, by either depletion of nesprin-2 or lamins A/C, resulted in the loss of ATM from DNA lesions. However, ATM signalling and DNA repair remained intact after lamins A/C depletion, whereas nesprin-2 disruption ablated downstream Chk2 activation and induced genomic instability. We conclude that lamins A/C and PML act as scaffolds to organise DNA-repair foci and compartmentalise nesprin-2/ERK signalling. However, nesprin-2/ERK signalling fidelity, but not their compartmentalisation at PML NBs, is essential for efficient DDR in VSMCs.
Collapse
Affiliation(s)
- D T Warren
- British Heart Foundation Centre of Research Excellence, Cardiovascular Division, King's College London, London SE5 9NU, UK
| | - T Tajsic
- Department of Medicine, Addenbrooke's Hospital, Cambridge CB2 2QQ, UK
| | - L J Porter
- British Heart Foundation Centre of Research Excellence, Cardiovascular Division, King's College London, London SE5 9NU, UK
| | - R M Minaisah
- British Heart Foundation Centre of Research Excellence, Cardiovascular Division, King's College London, London SE5 9NU, UK
| | - A Cobb
- British Heart Foundation Centre of Research Excellence, Cardiovascular Division, King's College London, London SE5 9NU, UK
| | - A Jacob
- British Heart Foundation Centre of Research Excellence, Cardiovascular Division, King's College London, London SE5 9NU, UK
| | - D Rajgor
- British Heart Foundation Centre of Research Excellence, Cardiovascular Division, King's College London, London SE5 9NU, UK
| | - Q P Zhang
- British Heart Foundation Centre of Research Excellence, Cardiovascular Division, King's College London, London SE5 9NU, UK
| | - C M Shanahan
- British Heart Foundation Centre of Research Excellence, Cardiovascular Division, King's College London, London SE5 9NU, UK
| |
Collapse
|
73
|
Rajgor D, Mellad JA, Soong D, Rattner JB, Fritzler MJ, Shanahan CM. Mammalian microtubule P-body dynamics are mediated by nesprin-1. ACTA ACUST UNITED AC 2014; 205:457-75. [PMID: 24862572 PMCID: PMC4033771 DOI: 10.1083/jcb.201306076] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Nesprins are a multi-isomeric family of spectrin-repeat (SR) proteins, predominantly known as nuclear envelope scaffolds. However, isoforms that function beyond the nuclear envelope remain poorly examined. Here, we characterize p50(Nesp1), a 50-kD isoform that localizes to processing bodies (PBs), where it acts as a microtubule-associated protein capable of linking mRNP complexes to microtubules. Overexpression of dominant-negative p50(Nesp1) caused Rck/p54, but not GW182, displacement from microtubules, resulting in reduced PB movement and cross talk with stress granules (SGs). These cells disassembled canonical SGs induced by sodium arsenite, but not those induced by hydrogen peroxide, leading to cell death and revealing PB-microtubule attachment is required for hydrogen peroxide-induced SG anti-apoptotic functions. Furthermore, p50(Nesp1) was required for miRNA-mediated silencing and interacted with core miRISC silencers Ago2 and Rck/p54 in an RNA-dependent manner and with GW182 in a microtubule-dependent manner. These data identify p50(Nesp1) as a multi-functional PB component and microtubule scaffold necessary for RNA granule dynamics and provides evidence for PB and SG micro-heterogeneity.
Collapse
Affiliation(s)
- Dipen Rajgor
- Cardiovascular Division, BHF Centre of Excellence, James Black Centre, King's College London, London SE5 9NU, England, UK
| | - Jason A Mellad
- Cardiovascular Division, BHF Centre of Excellence, James Black Centre, King's College London, London SE5 9NU, England, UK
| | - Daniel Soong
- Cardiovascular Division, BHF Centre of Excellence, James Black Centre, King's College London, London SE5 9NU, England, UK
| | - Jerome B Rattner
- Department of Biochemistry and Molecular Biology, University of Calgary, Calgary T2N 4N1, Alberta, Canada
| | - Marvin J Fritzler
- Department of Biochemistry and Molecular Biology, University of Calgary, Calgary T2N 4N1, Alberta, Canada
| | - Catherine M Shanahan
- Cardiovascular Division, BHF Centre of Excellence, James Black Centre, King's College London, London SE5 9NU, England, UK
| |
Collapse
|
74
|
Fedorchak GR, Kaminski A, Lammerding J. Cellular mechanosensing: getting to the nucleus of it all. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2014; 115:76-92. [PMID: 25008017 PMCID: PMC4252489 DOI: 10.1016/j.pbiomolbio.2014.06.009] [Citation(s) in RCA: 136] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2014] [Accepted: 06/28/2014] [Indexed: 12/12/2022]
Abstract
Cells respond to mechanical forces by activating specific genes and signaling pathways that allow the cells to adapt to their physical environment. Examples include muscle growth in response to exercise, bone remodeling based on their mechanical load, or endothelial cells aligning under fluid shear stress. While the involved downstream signaling pathways and mechanoresponsive genes are generally well characterized, many of the molecular mechanisms of the initiating 'mechanosensing' remain still elusive. In this review, we discuss recent findings and accumulating evidence suggesting that the cell nucleus plays a crucial role in cellular mechanotransduction, including processing incoming mechanoresponsive signals and even directly responding to mechanical forces. Consequently, mutations in the involved proteins or changes in nuclear envelope composition can directly impact mechanotransduction signaling and contribute to the development and progression of a variety of human diseases, including muscular dystrophy, cancer, and the focus of this review, dilated cardiomyopathy. Improved insights into the molecular mechanisms underlying nuclear mechanotransduction, brought in part by the emergence of new technologies to study intracellular mechanics at high spatial and temporal resolution, will not only result in a better understanding of cellular mechanosensing in normal cells but may also lead to the development of novel therapies in the many diseases linked to defects in nuclear envelope proteins.
Collapse
Affiliation(s)
- Gregory R Fedorchak
- Department of Biomedical Engineering, Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY 14853, USA
| | - Ashley Kaminski
- Department of Biomedical Engineering, Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY 14853, USA
| | - Jan Lammerding
- Department of Biomedical Engineering, Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY 14853, USA.
| |
Collapse
|
75
|
King SJ, Nowak K, Suryavanshi N, Holt I, Shanahan CM, Ridley AJ. Nesprin-1 and nesprin-2 regulate endothelial cell shape and migration. Cytoskeleton (Hoboken) 2014; 71:423-34. [PMID: 24931616 DOI: 10.1002/cm.21182] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Revised: 06/06/2014] [Accepted: 06/09/2014] [Indexed: 12/26/2022]
Abstract
Nesprins are large multi-domain proteins that link the nuclear envelope to the cytoskeleton and nucleoskeleton. Here we show that nesprin-1 and nesprin-2 play important roles in regulating cell shape and migration in endothelial cells. Nesprin-1 or nesprin-2 depletion by RNAi increased endothelial cell spread area and the length of cellular protrusions, as well as stimulating stress fibre assembly which correlated with an increase in F-actin levels. Nuclear area was also increased by nesprin depletion, and localization of the inner nuclear membrane protein emerin to the nuclear envelope was reduced. Depletion of nesprin-1 or nesprin-2 reduced migration of endothelial cells into a cell-free area, and decreased loop formation in an in vitro angiogenesis assay. Taken together, our results indicate that nesprin-1 and nesprin-2 both regulate nuclear and cytoplasmic architecture, which we propose leads to their effects on endothelial cell migration and angiogenic loop formation.
Collapse
Affiliation(s)
- Samantha J King
- Randall Division of Cell and Molecular Biophysics, King's College London, New Hunt's House, Guy's Campus, London, SE1 1UL, United Kingdom; Cardiovascular Division and British Heart Foundation Centre of Research Excellence, King's College London, James Black Centre, Denmark Hill Campus, London, SE5 9NU, United Kingdom
| | | | | | | | | | | |
Collapse
|
76
|
Chapman MA, Zhang J, Banerjee I, Guo LT, Zhang Z, Shelton GD, Ouyang K, Lieber RL, Chen J. Disruption of both nesprin 1 and desmin results in nuclear anchorage defects and fibrosis in skeletal muscle. Hum Mol Genet 2014; 23:5879-92. [PMID: 24943590 DOI: 10.1093/hmg/ddu310] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Proper localization and anchorage of nuclei within skeletal muscle is critical for cellular function. Alterations in nuclear anchoring proteins modify a number of cellular functions including mechanotransduction, nuclear localization, chromatin positioning/compaction and overall organ function. In skeletal muscle, nesprin 1 and desmin are thought to link the nucleus to the cytoskeletal network. Thus, we hypothesize that both of these factors play a key role in skeletal muscle function. To examine this question, we utilized global ablation murine models of nesprin 1, desmin or both nesprin 1 and desmin. Herein, we have created the nesprin-desmin double-knockout (DKO) mouse, eliminating a major fraction of nuclear-cytoskeletal connections and enabling understanding of the importance of nuclear anchorage in skeletal muscle. Globally, DKO mice are marked by decreased lifespan, body weight and muscle strength. With regard to skeletal muscle, DKO myonuclear anchorage was dramatically decreased compared with wild-type, nesprin 1(-/-) and desmin(-/-) mice. Additionally, nuclear-cytoskeletal strain transmission was decreased in DKO skeletal muscle. Finally, loss of nuclear anchorage in DKO mice coincided with a fibrotic response as indicated by increased collagen and extracellular matrix deposition and increased passive mechanical properties of muscle bundles. Overall, our data demonstrate that nesprin 1 and desmin serve redundant roles in nuclear anchorage and that the loss of nuclear anchorage in skeletal muscle results in a pathological response characterized by increased tissue fibrosis and mechanical stiffness.
Collapse
Affiliation(s)
| | | | | | - Ling T Guo
- Department of Pathology, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Zhiwei Zhang
- Department of Cardiothoracic Surgery, The Second Xiangya Hospital of Central South University, No. 139 Renmin Road, Changsha, Hunan 410011, P.R. China and
| | - G Diane Shelton
- Department of Pathology, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Kunfu Ouyang
- Department of Medicine, School of Chemical Biology and Biotechnology, Peking University, Shenzhen 518055, P.R. China
| | - Richard L Lieber
- Department of Bioengineering and Department of Orthopaedic Surgery, University of California San Diego, and Department of Veteran's Affairs, 9500 Gilman Drive, La Jolla, CA 92093-0863, USA
| | | |
Collapse
|
77
|
Abstract
Nuclear envelope (NE) proteins have fundamental roles in maintaining nuclear structure, cell signaling, chromatin organization, and gene regulation, and mutations in genes encoding NE components were identified as primary cause of a number of age associated diseases and cancer. Nesprin-1 belongs to a family of multi-isomeric NE proteins that are characterized by spectrin repeats. We analyzed NE components in various tumor cell lines and found that Nesprin-1 levels were strongly reduced associated with alterations in further NE components. By reducing the amounts of Nesprin-1 by RNAi mediated knockdown, we could reproduce those alterations in mouse and human cell lines. In a search for novel Nesprin-1 binding proteins, we identified MSH2 and MSH6, proteins of the DNA damage response pathway, as interactors and found alterations in the corresponding pathways in cells with lower Nesprin-1 levels. We also noticed increased number of γH2AX foci in the absence of exogenous DNA damage as was seen in tumor cells. The levels of phosphorylated kinases Chk1 and 2 were altered in a manner resembling tumor cells and the levels of Ku70 were low and the protein was not recruited to the DNA after hydroxyurea (HU) treatment. Our findings indicate a role for Nesprin-1 in the DNA damage response pathway and propose Nesprin-1 as novel player in tumorigenesis and genome instability.
Collapse
Affiliation(s)
- Ilknur Sur
- Institute of Biochemistry I; Medical Faculty; Center for Molecular Medicine Cologne (CMMC) and Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD); University of Cologne; Cologne, Germany
| | - Sascha Neumann
- Institute of Biochemistry I; Medical Faculty; Center for Molecular Medicine Cologne (CMMC) and Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD); University of Cologne; Cologne, Germany
| | - Angelika A Noegel
- Institute of Biochemistry I; Medical Faculty; Center for Molecular Medicine Cologne (CMMC) and Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD); University of Cologne; Cologne, Germany
| |
Collapse
|
78
|
Duong NT, Morris GE, Lam LT, Zhang Q, Sewry CA, Shanahan CM, Holt I. Nesprins: tissue-specific expression of epsilon and other short isoforms. PLoS One 2014; 9:e94380. [PMID: 24718612 PMCID: PMC3981789 DOI: 10.1371/journal.pone.0094380] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2014] [Accepted: 03/15/2014] [Indexed: 11/22/2022] Open
Abstract
Nesprin-1-giant and nesprin-2-giant regulate nuclear positioning by the interaction of their C-terminal KASH domains with nuclear membrane SUN proteins and their N-terminal calponin-homology domains with cytoskeletal actin. A number of short isoforms lacking the actin-binding domains are produced by internal promotion. We have evaluated the significance of these shorter isoforms using quantitative RT-PCR and western blotting with site-specific monoclonal antibodies. Within a complete map of nesprin isoforms, we describe two novel nesprin-2 epsilon isoforms for the first time. Epsilon isoforms are similar in size and structure to nesprin-1-alpha. Expression of nesprin isoforms was highly tissue-dependent. Nesprin-2-epsilon-1 was found in early embryonic cells, while nesprin-2-epsilon-2 was present in heart and other adult tissues, but not skeletal muscle. Some cell lines lack shorter isoforms and express only one of the two nesprin genes, suggesting that either of the giant nesprins is sufficient for basic cell functions. For the first time, localisation of endogenous nesprin away from the nuclear membrane was shown in cells where removal of the KASH domain by alternative splicing occurs. By distinguishing between degradation products and true isoforms on western blots, it was found that previously-described beta and gamma isoforms are expressed either at only low levels or with a limited tissue distribution. Two of the shortest alpha isoforms, nesprin-1-alpha-2 and nesprin-2-alpha-1, were found almost exclusively in cardiac and skeletal muscle and a highly conserved and alternatively-spliced exon, available in both nesprin genes, was always included in these tissues. These "muscle-specific" isoforms are thought to form a complex with emerin and lamin A/C at the inner nuclear membrane and mutations in all three proteins cause Emery-Dreifuss muscular dystrophy and/or inherited dilated cardiomyopathy, disorders in which only skeletal muscle and/or heart are affected.
Collapse
Affiliation(s)
- Nguyen Thuy Duong
- Wolfson Centre for Inherited Neuromuscular Disease, RJAH Orthopaedic Hospital, Oswestry, United Kingdom
- Institute of Genome Research (IGR), Vietnam Academy of Science and Technology (VAST), Hanoi, Vietnam
| | - Glenn E. Morris
- Wolfson Centre for Inherited Neuromuscular Disease, RJAH Orthopaedic Hospital, Oswestry, United Kingdom
- Institute for Science and Technology in Medicine, Keele University, Staffordshire, United Kingdom
| | - Le Thanh Lam
- Wolfson Centre for Inherited Neuromuscular Disease, RJAH Orthopaedic Hospital, Oswestry, United Kingdom
| | - Qiuping Zhang
- Cardiovascular Division, James Black Centre, King’s College, London, United Kingdom
| | - Caroline A. Sewry
- Wolfson Centre for Inherited Neuromuscular Disease, RJAH Orthopaedic Hospital, Oswestry, United Kingdom
- Dubowitz Neuromuscular Centre, Institute for Child Health and Great Ormond Street Hospital, London, United Kingdom
| | | | - Ian Holt
- Wolfson Centre for Inherited Neuromuscular Disease, RJAH Orthopaedic Hospital, Oswestry, United Kingdom
- Institute for Science and Technology in Medicine, Keele University, Staffordshire, United Kingdom
| |
Collapse
|
79
|
Luxton GWG, Starr DA. KASHing up with the nucleus: novel functional roles of KASH proteins at the cytoplasmic surface of the nucleus. Curr Opin Cell Biol 2014; 28:69-75. [PMID: 24704701 DOI: 10.1016/j.ceb.2014.03.002] [Citation(s) in RCA: 92] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2013] [Revised: 03/05/2014] [Accepted: 03/08/2014] [Indexed: 10/25/2022]
Abstract
Nuclear-cytoskeletal connections are central to fundamental cellular processes, including nuclear positioning and chromosome movements in meiosis. The cytoskeleton is coupled to the nucleoskeleton through conserved KASH-SUN bridges, or LINC complexes, that span the nuclear envelope. KASH proteins localize to the outer nuclear membrane where they connect the nucleus to the cytoskeleton. New findings have expanded the functional diversity of KASH proteins, showing that they interact with microtubule motors, actin, intermediate filaments, a nonconventional myosin, RanGAP, and each other. The role of KASH proteins in cellular mechanics is discussed. Genetic mutations in KASH proteins are associated with autism, hearing loss, cancer, muscular dystrophy and other diseases.
Collapse
Affiliation(s)
- G W Gant Luxton
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, MN 55455, United States.
| | - Daniel A Starr
- Department of Molecular and Cellular Biology, University of California, Davis, CA 95616, United States.
| |
Collapse
|
80
|
Stroud MJ, Banerjee I, Veevers J, Chen J. Linker of nucleoskeleton and cytoskeleton complex proteins in cardiac structure, function, and disease. Circ Res 2014; 114:538-48. [PMID: 24481844 DOI: 10.1161/circresaha.114.301236] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
The linker of nucleoskeleton and cytoskeleton (LINC) complex, composed of proteins within the inner and the outer nuclear membranes, connects the nuclear lamina to the cytoskeleton. The importance of this complex has been highlighted by the discovery of mutations in genes encoding LINC complex proteins, which cause skeletal or cardiac myopathies. Herein, this review summarizes structure, function, and interactions of major components of the LINC complex, highlights how mutations in these proteins may lead to cardiac disease, and outlines future challenges in the field.
Collapse
Affiliation(s)
- Matthew J Stroud
- From the Department of Cardiology, University of California San Diego School of Medicine, La Jolla, CA
| | | | | | | |
Collapse
|
81
|
Xu W, Cohen-Woods S, Chen Q, Noor A, Knight J, Hosang G, Parikh SV, De Luca V, Tozzi F, Muglia P, Forte J, McQuillin A, Hu P, Gurling HMD, Kennedy JL, McGuffin P, Farmer A, Strauss J, Vincent JB. Genome-wide association study of bipolar disorder in Canadian and UK populations corroborates disease loci including SYNE1 and CSMD1. BMC MEDICAL GENETICS 2014; 15:2. [PMID: 24387768 PMCID: PMC3901032 DOI: 10.1186/1471-2350-15-2] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/10/2013] [Accepted: 12/20/2013] [Indexed: 11/10/2022]
Abstract
BACKGROUND Recently, genome-wide association studies (GWAS) for cases versus controls using single nucleotide polymorphism microarray data have shown promising findings for complex neuropsychiatric disorders, including bipolar disorder (BD). METHODS Here we describe a comprehensive genome-wide study of bipolar disorder (BD), cross-referencing analysis from a family-based study of 229 small families with association analysis from over 950 cases and 950 ethnicity-matched controls from the UK and Canada. Further, loci identified in these analyses were supported by pathways identified through pathway analysis on the samples. RESULTS Although no genome-wide significant markers were identified, the combined GWAS findings have pointed to several genes of interest that support GWAS findings for BD from other groups or consortia, such as at SYNE1 on 6q25, PPP2R2C on 4p16.1, ZNF659 on 3p24.3, CNTNAP5 (2q14.3), and CDH13 (16q23.3). This apparent corroboration across multiple sites gives much confidence to the likelihood of genetic involvement in BD at these loci. In particular, our two-stage strategy found association in both our combined case/control analysis and the family-based analysis on 1q21.2 (closest gene: sphingosine-1-phosphate receptor 1 gene, S1PR1) and on 1q24.1 near the gene TMCO1, and at CSMD1 on 8p23.2, supporting several previous GWAS reports for BD and for schizophrenia. Pathway analysis suggests association of pathways involved in calcium signalling, neuropathic pain signalling, CREB signalling in neurons, glutamate receptor signalling and axonal guidance signalling. CONCLUSIONS The findings presented here show support for a number of genes previously implicated genes in the etiology of BD, including CSMD1 and SYNE1, as well as evidence for previously unreported genes such as the brain-expressed genes ADCY2, NCALD, WDR60, SCN7A and SPAG16.
Collapse
Affiliation(s)
- Wei Xu
- Dalla Lana School of Public Health, University of Toronto, Toronto, Canada
| | - Sarah Cohen-Woods
- MRC SGDP Centre, King’s College London, Institute of Psychiatry, De Crespigny Park, London SE5 8AF, UK
| | - Qian Chen
- Cancer Care Ontario, Toronto, Canada
| | - Abdul Noor
- Neurogenetics Section, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health (CAMH), R-32, 250 College Street, Toronto, ON M5T 1R8, Canada
| | - Jo Knight
- Neurogenetics Section, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health (CAMH), R-32, 250 College Street, Toronto, ON M5T 1R8, Canada
- Department of Psychiatry, University of Toronto, Toronto, Canada
| | - Georgina Hosang
- MRC SGDP Centre, King’s College London, Institute of Psychiatry, De Crespigny Park, London SE5 8AF, UK
| | - Sagar V Parikh
- Department of Psychiatry, University of Toronto, Toronto, Canada
- Centre for Addiction and Mental Health (CAMH), Toronto, Canada
| | | | - Federica Tozzi
- GSK Research & Development, Medical Genetics, Clinical Pharmacology and Discovery Medicine, Via Fleming 4, Verona, Italy
- GSK Research & Development, Medical Genetics, Clinical Pharmacology and Discovery Medicine, Greenford Road, Greenford, Middlesex UB6 OHE, UK
| | - Pierandrea Muglia
- GSK Research & Development, Medical Genetics, Clinical Pharmacology and Discovery Medicine, Via Fleming 4, Verona, Italy
- Exploratory Medicine & Early Development, NeuroSearch, Copenhagen, Denmark
- GSK Research & Development, Medical Genetics, Clinical Pharmacology and Discovery Medicine, Greenford Road, Greenford, Middlesex UB6 OHE, UK
| | - Julia Forte
- GSK Research & Development, Medical Genetics, Clinical Pharmacology and Discovery Medicine, Via Fleming 4, Verona, Italy
- GSK Research & Development, Medical Genetics, Clinical Pharmacology and Discovery Medicine, Greenford Road, Greenford, Middlesex UB6 OHE, UK
| | - Andrew McQuillin
- Molecular Psychiatry Laboratory, Mental Health Sciences Unit, Faculty of Brain Sciences, University College London, London, UK
| | - Pingzhao Hu
- The Centre for Applied Genomics, The Hospital for Sick Children Research Institute, Toronto, Canada
| | - Hugh MD Gurling
- Molecular Psychiatry Laboratory, Mental Health Sciences Unit, Faculty of Brain Sciences, University College London, London, UK
| | - James L Kennedy
- Neurogenetics Section, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health (CAMH), R-32, 250 College Street, Toronto, ON M5T 1R8, Canada
- Department of Psychiatry, University of Toronto, Toronto, Canada
| | - Peter McGuffin
- MRC SGDP Centre, King’s College London, Institute of Psychiatry, De Crespigny Park, London SE5 8AF, UK
| | - Anne Farmer
- MRC SGDP Centre, King’s College London, Institute of Psychiatry, De Crespigny Park, London SE5 8AF, UK
| | - John Strauss
- Department of Psychiatry, University of Toronto, Toronto, Canada
- Centre for Addiction and Mental Health (CAMH), Toronto, Canada
| | - John B Vincent
- Neurogenetics Section, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health (CAMH), R-32, 250 College Street, Toronto, ON M5T 1R8, Canada
- Department of Psychiatry, University of Toronto, Toronto, Canada
- The Institute of Medical Science, University of Toronto, Toronto, Canada
| |
Collapse
|
82
|
|
83
|
Razafsky D, Wirtz D, Hodzic D. Nuclear envelope in nuclear positioning and cell migration. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2014; 773:471-90. [PMID: 24563361 PMCID: PMC4310828 DOI: 10.1007/978-1-4899-8032-8_21] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Hauling and anchoring the nucleus within immobile or motile cells, tissues, and/or syncytia represents a major challenge. In the past 15 years, Linkers of the Nucleoskeleton to the Cytoskeleton (LINC complexes) have emerged as evolutionary-conserved molecular devices that span the nuclear envelope and provide interacting interfaces for cytoskeletal networks and molecular motors to the nuclear envelope. Here, we review the molecular composition of LINC complexes and focus on how their genetic alteration in vivo has provided a wealth of information related to the relevance of nuclear positioning during tissue development and homeostasis with a special emphasis on the central nervous system. As it may be relevant for metastasis in a range of cancers, the involvement of LINC complexes in migration of nonneuronal cells via its interaction with the perinuclear actin cap will also be developed.
Collapse
Affiliation(s)
- David Razafsky
- Washington University School of Medicine, Department of Ophthalmology and Visual Sciences, 660 South Euclid Ave, St Louis, MO, 63110, USA
| | - Denis Wirtz
- The Johns Hopkins University, Department of Chemical and Biomolecular engineering, 3400 North Charles St., Baltimore, MD, 21218, USA
| | - Didier Hodzic
- Washington University School of Medicine, Department of Ophthalmology and Visual Sciences, 660 South Euclid Ave, St Louis, MO, 63110, USA
| |
Collapse
|
84
|
Abstract
Despite decades of research, cancer metastasis remains an incompletely understood process that is as complex as it is devastating. In recent years, there has been an increasing push to investigate the biomechanical aspects of tumorigenesis, complementing the research on genetic and biochemical changes. In contrast to the high genetic variability encountered in cancer cells, almost all metastatic cells are subject to the same physical constraints as they leave the primary tumor, invade surrounding tissues, transit through the circulatory system, and finally infiltrate new tissues. Advances in live cell imaging and other biophysical techniques, including measurements of subcellular mechanics, have yielded stunning new insights into the physics of cancer cells. While much of this research has been focused on the mechanics of the cytoskeleton and the cellular microenvironment, it is now emerging that the mechanical properties of the cell nucleus and its connection to the cytoskeleton may play a major role in cancer metastasis, as deformation of the large and stiff nucleus presents a substantial obstacle during the passage through the dense interstitial space and narrow capillaries. Here, we present an overview of the molecular components that govern the mechanical properties of the nucleus, and we discuss how changes in nuclear structure and composition observed in many cancers can modulate nuclear mechanics and promote metastatic processes. Improved insights into this interplay between nuclear mechanics and metastatic progression may have powerful implications in cancer diagnostics and therapy and may reveal novel therapeutic targets for pharmacological inhibition of cancer cell invasion.
Collapse
Affiliation(s)
- Celine Denais
- Department of Biomedical Engineering, Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY, 14853, USA,
| | | |
Collapse
|
85
|
Neumann S, Noegel AA. Nesprins in Cell Stability and Migration. CANCER BIOLOGY AND THE NUCLEAR ENVELOPE 2014; 773:491-504. [DOI: 10.1007/978-1-4899-8032-8_22] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
86
|
Alam S, Lovett DB, Dickinson RB, Roux KJ, Lele TP. Nuclear forces and cell mechanosensing. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2014; 126:205-15. [PMID: 25081619 DOI: 10.1016/b978-0-12-394624-9.00008-7] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Cells respond to mechanical signals, but the subcellular mechanisms are not well understood. The nucleus has recently emerged as an important mechanosensory organelle in the cell, as it is intimately connected to the cytoskeleton. Mechanical forces applied to cells that act on membrane-embedded receptors are transmitted through the cytoskeleton to the nuclear surface. Interfering with linkers of the nucleus to the cytoskeleton causes defects in cell mechanosensing and cell function. In this chapter, we discuss recent work in this area, highlighting the role that the nuclear linkages with the cytoskeleton play in cellular mechanotransduction.
Collapse
Affiliation(s)
- Samer Alam
- Department of Chemical Engineering, University of Florida, Gainesville, Florida, USA
| | - David B Lovett
- Department of Chemical Engineering, University of Florida, Gainesville, Florida, USA
| | - Richard B Dickinson
- Department of Chemical Engineering, University of Florida, Gainesville, Florida, USA
| | - Kyle J Roux
- Sanford Children's Health Research Center, University of South Dakota, Sioux Falls, South Dakota, USA
| | - Tanmay P Lele
- Department of Chemical Engineering, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
87
|
Cartwright S, Karakesisoglou I. Nesprins in health and disease. Semin Cell Dev Biol 2013; 29:169-79. [PMID: 24374011 DOI: 10.1016/j.semcdb.2013.12.010] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2013] [Revised: 11/29/2013] [Accepted: 12/15/2013] [Indexed: 01/20/2023]
Abstract
LINC (Linker of Nucleoskeleton and Cytoskeleton) complex is an evolutionary conserved structure that spans the entire nuclear envelope (NE), and integrates the nuclear interior with the cytoskeleton, in order to support a diverse array of fundamental biological processes. Key components of the LINC complex are the nesprins (Nuclear Envelope SPectrin Repeat proteINS) that were initially described as large integral NE proteins. However, nesprin genes are complex and generate many variants, which occupy various sub-cellular compartments suggesting additional functions. Hence, the potential involvement of nesprins in disease has expanded immensely on what we already know. That is, nesprins are implicated in diseases such as cancer, myopathies, arthrogryposis, neurological disorders and hearing loss. Here we review nesprins by providing an in depth account of their structure, molecular interactions and cellular functions with relevance to their potential roles in disease. Specifically, we speculate about possible pathomechanisms underlying nesprin-associated diseases.
Collapse
Affiliation(s)
- Sarah Cartwright
- School of Biological and Biomedical Sciences, University of Durham, Durham DH1 3LE, UK
| | | |
Collapse
|
88
|
Hettmer S, Teot LA, van Hummelen P, MacConaill L, Bronson RT, Dall'Osso C, Mao J, McMahon AP, Gruber PJ, Grier HE, Rodriguez-Galindo C, Fletcher CD, Wagers AJ. Mutations in Hedgehog pathway genes in fetal rhabdomyomas. J Pathol 2013; 231:44-52. [PMID: 23780909 PMCID: PMC3875333 DOI: 10.1002/path.4229] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2013] [Revised: 06/03/2013] [Accepted: 06/07/2013] [Indexed: 02/06/2023]
Abstract
Ligand-independent, constitutive activation of Hedgehog signalling in mice expressing a mutant, activated SmoM2 allele results in the development of multifocal, highly differentiated tumours that express myogenic markers (including desmin, actin, MyoD and myogenin). The histopathology of these tumours, commonly classified as rhabdomyosarcomas, more closely resembles human fetal rhabdomyoma (FRM), a benign tumour that can be difficult to distinguish from highly differentiated rhabdomyosarcomas. We evaluated the spectrum of Hedgehog (HH) pathway gene mutations in a cohort of human FRM tumours by targeted Illumina sequencing and fluorescence in situ hybridization testing for PTCH1. Our studies identified functionally relevant aberrations at the PTCH1 locus in three of five FRM tumours surveyed, including a PTCH1 frameshift mutation in one tumour and homozygous deletions of PTCH1 in two tumours. These data suggest that activated Hedgehog signalling contributes to the biology of human FRM.
Collapse
Affiliation(s)
- Simone Hettmer
- Howard Hughes Medical Institute, Department of Stem Cell and Regenerative Biology, Harvard University, Harvard Stem Cell Institute, Cambridge, MA, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
89
|
Liggett JL, Choi CK, Donnell RL, Kihm KD, Kim JS, Min KW, Noegel AA, Baek SJ. Nonsteroidal anti-inflammatory drug sulindac sulfide suppresses structural protein Nesprin-2 expression in colorectal cancer cells. Biochim Biophys Acta Gen Subj 2013; 1840:322-31. [PMID: 24080406 DOI: 10.1016/j.bbagen.2013.09.032] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2013] [Revised: 08/22/2013] [Accepted: 09/20/2013] [Indexed: 12/21/2022]
Abstract
BACKGROUND Nonsteroidal anti-inflammatory drugs (NSAIDs) are well known for treating inflammatory disease and have been reported to have anti-tumorigenic effects. Their mechanisms are not fully understood, but both cyclooxygenase (COX) dependent and independent pathways are involved. Our goal was to shed further light on COX-independent activity. METHODS Human colorectal cancer cells were observed under differential interference contrast microscopy (DICM), fluorescent microscopy, and micro-impedance measurement. Microarray analysis was performed using HCT-116 cells treated with sulindac sulfide (SS). PCR and Western blots were performed to confirm the microarray data and immunohistochemistry was performed to screen for Nesprin-2 expression. Micro-impedance was repeating including Nesprin-2 knock-down by siRNA. RESULTS HCT-116 cells treated with SS showed dramatic morphological changes under DICM and fluorescent microscopy, as well as weakened cellular adhesion as measured by micro-impedance. Nesprin-2 was selected from two independent microarrays, based on its novelty in relation to cancer and its role in cell organization. SS diminished Nesprin-2 mRNA expression as assessed by reverse transcriptase and real time PCR. Various other NSAIDs were also tested and demonstrated that inhibition of Nesprin-2 mRNA was not unique to SS. Additionally, immunohistochemistry showed higher levels of Nesprin-2 in many tumors in comparison with normal tissues. Further micro-impedance experiments on cells with reduced Nesprin-2 expression showed a proportional loss of cellular adhesion. CONCLUSIONS Nesprin-2 is down-regulated by NSAIDs and highly expressed in many cancers. GENERAL SIGNIFICANCE Our data suggest that Nesprin-2 may be a potential novel oncogene in human cancer cells and NSAIDs could decrease its expression.
Collapse
Affiliation(s)
- Jason L Liggett
- Department of Biomedical and Diagnostic Sciences, University of Tennessee, Knoxville, TN 37996, USA
| | | | | | | | | | | | | | | |
Collapse
|
90
|
Razafsky DS, Ward CL, Kolb T, Hodzic D. Developmental regulation of linkers of the nucleoskeleton to the cytoskeleton during mouse postnatal retinogenesis. Nucleus 2013; 4:399-409. [PMID: 23974729 DOI: 10.4161/nucl.26244] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Sun proteins and Nesprins are two families of proteins whose direct interactions across the nuclear envelope provide for the core of Linkers of the Nucleoskeleton to the Cytoskeleton (LINC complexes) that physically connect the nucleus interior to cytoskeletal networks. Whereas LINC complexes play essential roles in nuclear migration anchorage and underlie normal CNS development, the developmental regulation of their composition remains largely unknown. In this study, we examined the spatiotemporal expression of lamins, Sun proteins and Nesprins during postnatal mouse retinal development. Whereas retinal precursor cells mostly express B-type lamins, Sun1, and high molecular weight isoforms of Nesprins, post-mitotic retinal cells are characterized by a drastic downregulation of the latter, the expression of A-type lamins, and the strong induction of a specific isoform of Nesprin1 late in retinal development. Importantly, our results emphasize different spatiotemporal expression for Nesprin1 and Nesprin2 and further suggest an important role for KASH-less isoforms of Nesprin1 in the CNS. In conclusion, the transition from retinal precursor cells undergoing interkinetic nuclear migration to post-mitotic retinal cells undergoing nuclear translocation and/or anchorage is accompanied by a profound remodeling of LINC complexes composition. This remodeling may reflect different requirements of nuclear dynamics at different stages of CNS development.
Collapse
Affiliation(s)
- David S Razafsky
- Department of Ophthalmology and Visual Sciences; Washington University School of Medicine; St Louis, MO USA; Division of Molecular Genetics; German Cancer Research Center (DKFZ); Heidelberg, Germany
| | | | | | | |
Collapse
|
91
|
Yang L, Munck M, Swaminathan K, Kapinos LE, Noegel AA, Neumann S. Mutations in LMNA modulate the lamin A--Nesprin-2 interaction and cause LINC complex alterations. PLoS One 2013; 8:e71850. [PMID: 23977161 PMCID: PMC3748058 DOI: 10.1371/journal.pone.0071850] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2013] [Accepted: 07/03/2013] [Indexed: 11/29/2022] Open
Abstract
Background In eukaryotes the genetic material is enclosed by a continuous membrane system, the nuclear envelope (NE). Along the NE specific proteins assemble to form meshworks and mutations in these proteins have been described in a group of human diseases called laminopathies. Laminopathies include lipodystrophies, muscle and cardiac diseases as well as metabolic or progeroid syndromes. Most laminopathies are caused by mutations in the LMNAgene encoding lamins A/C. Together with Nesprins (Nuclear Envelope Spectrin Repeat Proteins) they are core components of the LINC complex (Linker of Nucleoskeleton and Cytoskeleton). The LINC complex connects the nucleoskeleton and the cytoskeleton and plays a role in the transfer of mechanically induced signals along the NE into the nucleus, and its components have been attributed functions in maintaining nuclear and cellular organization as well as signal transduction. Results Here we narrowed down the interaction sites between lamin A and Nesprin-2 to aa 403–425 in lamin A and aa 6146–6347 in Nesprin-2. Laminopathic mutations in and around the involved region of lamin A (R401C, G411D, G413C, V415I, R419C, L421P, R427G, Q432X) modulate the interaction with Nesprin-2 and this may contribute to the disease phenotype. The most notable mutation is the lamin A mutation Q432X that alters LINC complex protein assemblies and causes chromosomal and transcription factor rearrangements. Conclusion Mutations in Nesprin-2 and lamin A are characterised by complex genotype phenotype relations. Our data show that each mutation in LMNAanalysed here has a distinct impact on the interaction among both proteins that substantially explains how distinct mutations in widely expressed genes lead to the formation of phenotypically different diseases.
Collapse
Affiliation(s)
- Liu Yang
- Institute for Biochemistry I, Medical Faculty, University of Cologne, and Center for Molecular Medicine Cologne (CMMC) and Cologne Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Medical Faculty, University of Cologne, Cologne, Germany
| | - Martina Munck
- Institute for Biochemistry I, Medical Faculty, University of Cologne, and Center for Molecular Medicine Cologne (CMMC) and Cologne Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Medical Faculty, University of Cologne, Cologne, Germany
| | - Karthic Swaminathan
- Institute for Biochemistry I, Medical Faculty, University of Cologne, and Center for Molecular Medicine Cologne (CMMC) and Cologne Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Medical Faculty, University of Cologne, Cologne, Germany
| | - Larisa E. Kapinos
- Biozentrum and the Nanoscience Institute, University of Basel, Basel, Switzerland
| | - Angelika A. Noegel
- Institute for Biochemistry I, Medical Faculty, University of Cologne, and Center for Molecular Medicine Cologne (CMMC) and Cologne Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Medical Faculty, University of Cologne, Cologne, Germany
- * E-mail: (AAN); (SN)
| | - Sascha Neumann
- Institute for Biochemistry I, Medical Faculty, University of Cologne, and Center for Molecular Medicine Cologne (CMMC) and Cologne Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Medical Faculty, University of Cologne, Cologne, Germany
- * E-mail: (AAN); (SN)
| |
Collapse
|
92
|
Abstract
Nuclear envelope
spectrin-repeat
proteins (Nesprins), are a novel family of
nuclear and cytoskeletal proteins with rapidly expanding roles as intracellular scaffolds
and linkers. Originally described as proteins that localise to the nuclear envelope (NE)
and establish nuclear-cytoskeletal connections, nesprins have now been found to comprise a
diverse spectrum of tissue specific isoforms that localise to multiple sub-cellular
compartments. Here, we describe how nesprins are necessary in maintaining cellular
architecture by acting as essential scaffolds and linkers at both the NE and other
sub-cellular domains. More importantly, we speculate how nesprin mutations may disrupt
tissue specific nesprin scaffolds and explain the tissue specific nature of many
nesprin-associated diseases, including laminopathies.
Collapse
|
93
|
Autore F, Pfuhl M, Quan X, Williams A, Roberts RG, Shanahan CM, Fraternali F. Large-scale modelling of the divergent spectrin repeats in nesprins: giant modular proteins. PLoS One 2013; 8:e63633. [PMID: 23671687 PMCID: PMC3646009 DOI: 10.1371/journal.pone.0063633] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2012] [Accepted: 04/09/2013] [Indexed: 11/29/2022] Open
Abstract
Nesprin-1 and nesprin-2 are nuclear envelope (NE) proteins characterized by a common structure of an SR (spectrin repeat) rod domain and a C-terminal transmembrane KASH [Klarsicht-ANC-Syne-homology] domain and display N-terminal actin-binding CH (calponin homology) domains. Mutations in these proteins have been described in Emery-Dreifuss muscular dystrophy and attributed to disruptions of interactions at the NE with nesprins binding partners, lamin A/C and emerin. Evolutionary analysis of the rod domains of the nesprins has shown that they are almost entirely composed of unbroken SR-like structures. We present a bioinformatical approach to accurate definition of the boundaries of each SR by comparison with canonical SR structures, allowing for a large-scale homology modelling of the 74 nesprin-1 and 56 nesprin-2 SRs. The exposed and evolutionary conserved residues identify important pbs for protein-protein interactions that can guide tailored binding experiments. Most importantly, the bioinformatics analyses and the 3D models have been central to the design of selected constructs for protein expression. 1D NMR and CD spectra have been performed of the expressed SRs, showing a folded, stable, high content α-helical structure, typical of SRs. Molecular Dynamics simulations have been performed to study the structural and elastic properties of consecutive SRs, revealing insights in the mechanical properties adopted by these modules in the cell.
Collapse
Affiliation(s)
- Flavia Autore
- Randall Division of Cell and Molecular Biophysics, School of Physical Sciences and Engineering, King's College London, London, United Kingdom
- Division of Cardiovascular Medicine, BHF Centre of Research Excellence, King's College London, London, United Kingdom
| | - Mark Pfuhl
- Randall Division of Cell and Molecular Biophysics, School of Physical Sciences and Engineering, King's College London, London, United Kingdom
| | - Xueping Quan
- Randall Division of Cell and Molecular Biophysics, School of Physical Sciences and Engineering, King's College London, London, United Kingdom
| | - Aisling Williams
- Randall Division of Cell and Molecular Biophysics, School of Physical Sciences and Engineering, King's College London, London, United Kingdom
- Division of Cardiovascular Medicine, BHF Centre of Research Excellence, King's College London, London, United Kingdom
| | - Roland G. Roberts
- Division of Medical and Molecular Genetics, Kings College London, Guy's Hospital, London, United Kingdom
| | - Catherine M. Shanahan
- Division of Cardiovascular Medicine, BHF Centre of Research Excellence, King's College London, London, United Kingdom
| | - Franca Fraternali
- Randall Division of Cell and Molecular Biophysics, School of Physical Sciences and Engineering, King's College London, London, United Kingdom
- The Thomas Young Centre for Theory and Simulation of Materials, London, United Kingdom
| |
Collapse
|
94
|
McCarthy CB, Santini MS, Pimenta PFP, Diambra LA. First comparative transcriptomic analysis of wild adult male and female Lutzomyia longipalpis, vector of visceral leishmaniasis. PLoS One 2013; 8:e58645. [PMID: 23554910 PMCID: PMC3595279 DOI: 10.1371/journal.pone.0058645] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2012] [Accepted: 02/05/2013] [Indexed: 01/08/2023] Open
Abstract
Leishmaniasis is a vector-borne disease with a complex epidemiology and ecology. Visceral leishmaniasis (VL) is its most severe clinical form as it results in death if not treated. In Latin America VL is caused by the protist parasite Leishmania infantum (syn. chagasi) and transmitted by Lutzomyia longipalpis. This phlebotomine sand fly is only found in the New World, from Mexico to Argentina. However, due to deforestation, migration and urbanisation, among others, VL in Latin America is undergoing an evident geographic expansion as well as dramatic changes in its transmission patterns. In this context, the first VL outbreak was recently reported in Argentina, which has already caused 7 deaths and 83 reported cases. Insect vector transcriptomic analyses enable the identification of molecules involved in the insect's biology and vector-parasite interaction. Previous studies on laboratory reared Lu. longipalpis have provided a descriptive repertoire of gene expression in the whole insect, midgut, salivary gland and male reproductive organs. Nevertheless, the study of wild specimens would contribute a unique insight into the development of novel bioinsecticides. Given the recent VL outbreak in Argentina and the compelling need to develop appropriate control strategies, this study focused on wild male and female Lu. longipalpis from an Argentine endemic (Posadas, Misiones) and a Brazilian non-endemic (Lapinha Cave, Minas Gerais) VL location. In this study, total RNA was extracted from the sand flies, submitted to sequence independent amplification and high-throughput pyrosequencing. This is the first time an unbiased and comprehensive transcriptomic approach has been used to analyse an infectious disease vector in its natural environment. Transcripts identified in the sand flies showed characteristic profiles which correlated with the environment of origin and with taxa previously identified in these same specimens. Among these, various genes represented putative targets for vector control via RNA interference (RNAi).
Collapse
Affiliation(s)
- Christina B McCarthy
- Centro Regional de Estudios Genómicos, Facultad de Ciencias Exactas, Universidad Nacional de La Plata, Florencio Varela, Buenos Aires, Argentina.
| | | | | | | |
Collapse
|
95
|
LINC complexes mediate the positioning of cone photoreceptor nuclei in mouse retina. PLoS One 2012; 7:e47180. [PMID: 23071752 PMCID: PMC3465324 DOI: 10.1371/journal.pone.0047180] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2012] [Accepted: 09/10/2012] [Indexed: 11/19/2022] Open
Abstract
It has long been observed that many neuronal types position their nuclei within restricted cytoplasmic boundaries. A striking example is the apical localization of cone photoreceptors nuclei at the outer edge of the outer nuclear layer of mammalian retinas. Yet, little is known about how such nuclear spatial confinement is achieved and further maintained. Linkers of the Nucleoskeleton to the Cytoskeleton (LINC complexes) consist of evolutionary-conserved macromolecular assemblies that span the nuclear envelope to connect the nucleus with the peripheral cytoskeleton. Here, we applied a new transgenic strategy to disrupt LINC complexes either in cones or rods. In adult cones, we observed a drastic nuclear mislocalization on the basal side of the ONL that affected cone terminals overall architecture. We further provide evidence that this phenotype may stem from the inability of cone precursor nuclei to migrate towards the apical side of the outer nuclear layer during early postnatal retinal development. By contrast, disruption of LINC complexes within rod photoreceptors, whose nuclei are scattered across the outer nuclear layer, had no effect on the positioning of their nuclei thereby emphasizing differential requirements for LINC complexes by different neuronal types. We further show that Sun1, a component of LINC complexes, but not A-type lamins, which interact with LINC complexes at the nuclear envelope, participate in cone nuclei positioning. This study provides key mechanistic aspects underlying the well-known spatial confinement of cone nuclei as well as a new mouse model to evaluate the pathological relevance of nuclear mispositioning.
Collapse
|