201
|
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: 50] [Impact Index Per Article: 4.5] [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
|
202
|
Wright AT, Magnaldo T, Sontag RL, Anderson LN, Sadler NC, Piehowski PD, Gache Y, Weber TJ. Deficient expression of aldehyde dehydrogenase 1A1 is consistent with increased sensitivity of Gorlin syndrome patients to radiation carcinogenesis. Mol Carcinog 2013; 54:473-84. [PMID: 24285572 DOI: 10.1002/mc.22115] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2013] [Revised: 10/28/2013] [Accepted: 11/07/2013] [Indexed: 12/30/2022]
Abstract
Human phenotypes that are highly susceptible to radiation carcinogenesis have been identified. Sensitive phenotypes often display robust regulation of molecular features that modify biological response, which can facilitate identification of the pathways/networks that contribute to pathophysiological outcomes. Here we interrogate primary dermal fibroblasts isolated from Gorlin syndrome patients (GDFs), who display a pronounced inducible tumorigenic response to radiation, in comparison to normal human dermal fibroblasts (NHDFs). Our approach exploits newly developed thiol reactive probes to define changes in protein thiol profiles in live cell studies, which minimizes artifacts associated with cell lysis. Redox probes revealed deficient expression of an apparent 55 kDa protein thiol in GDFs from independent Gorlin syndrome patients, compared with NHDFs. Proteomics tentatively identified this protein as aldehyde dehydrogenase 1A1 (ALDH1A1), a key enzyme regulating retinoic acid synthesis, and ALDH1A1 protein deficiency in GDFs was confirmed by Western blot. A number of additional protein thiol differences in GDFs were identified, including radiation responsive annexin family members and lamin A/C. Collectively, candidates identified in our study have plausible implications for radiation health effects and cancer susceptibility.
Collapse
Affiliation(s)
- Aaron T Wright
- Omic Biological Applications, Pacific Northwest National Laboratory, Richland, Washington
| | | | | | | | | | | | | | | |
Collapse
|
203
|
Das A, Grotsky DA, Neumann MA, Kreienkamp R, Gonzalez-Suarez I, Redwood AB, Kennedy BK, Stewart CL, Gonzalo S. Lamin A Δexon9 mutation leads to telomere and chromatin defects but not genomic instability. Nucleus 2013; 4:410-9. [PMID: 24153156 DOI: 10.4161/nucl.26873] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Over 300 mutations in the LMNA gene, encoding A-type lamins, are associated with 15 human degenerative disorders and premature aging syndromes. Although genomic instability seems to contribute to the pathophysiology of some laminopathies, there is limited information about what mutations cause genomic instability and by which molecular mechanisms. Mouse embryonic fibroblasts depleted of A-type lamins or expressing mutants lacking exons 8-11 (Lmna(Δ8-11/Δ8-11)) exhibit alterations in telomere biology and DNA repair caused by cathepsin L-mediated degradation of 53BP1 and reduced expression of BRCA1 and RAD51. Thus, a region encompassing exons 8-11 seems essential for genome integrity. Given that deletion of lamin A exon 9 in the mouse (Lmna(Δ9/Δ9)) results in a progeria phenotype, we tested if this domain is important for genome integrity. Lmna(Δ9/Δ9) MEFs exhibit telomere shortening and heterochromatin alterations but do not activate cathepsin L-mediated degradation of 53BP1 and maintain expression of BRCA1 and RAD51. Accordingly, Lmna(Δ9/Δ9) MEFs do not present genomic instability, and expression of mutant lamin A Δexon9 in lamin-depleted cells restores DNA repair factors levels and partially rescues nuclear abnormalities. These data reveal that the domain encoded by exon 9 is important to maintain telomere homeostasis and heterochromatin structure but does not play a role in DNA repair, thus pointing to other exons in the lamin A tail as responsible for the genomic instability phenotype in Lmna(Δ8-11/Δ8-11) mice. Our study also suggests that the levels of DNA repair factors 53BP1, BRCA1 and RAD51 could potentially serve as biomarkers to identify laminopathies that present with genomic instability.
Collapse
Affiliation(s)
- Arindam Das
- Edward A Doisy Department of Biochemistry and Molecular Biology; School of Medicine; St Louis University; St Louis, MO USA; Departments of Radiation Oncology and Cell Biology & Physiology; School of Medicine; Washington University; St Louis, MO USA; Buck Institute for Research on Aging; Novato, CA USA; Institute of Medical Biology; Biopolis; Singapore
| | | | | | | | | | | | | | | | | |
Collapse
|
204
|
Scherfer C, Han VC, Wang Y, Anderson AE, Galko MJ. Autophagy drives epidermal deterioration in a Drosophila model of tissue aging. Aging (Albany NY) 2013; 5:276-87. [PMID: 23599123 PMCID: PMC3651520 DOI: 10.18632/aging.100549] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Organismal lifespan has been the primary readout in aging research. However, how longevity genes control tissue-specific aging remains an open question. To examine the crosstalk between longevity programs and specific tissues during aging, biomarkers of organ-specific aging are urgently needed. Since the earliest signs of aging occur in the skin, we sought to examine skin aging in a genetically tractable model. Here we introduce a Drosophila model of skin aging. The epidermis undergoes a dramatic morphological deterioration with age that includes membrane and nuclear loss. These changes were decelerated in a long-lived mutant and accelerated in a short-lived mutant. An increase in autophagy markers correlated with epidermal aging. Finally, the epidermis of Atg7 mutants retained younger characteristics, suggesting that autophagy is a critical driver of epidermal aging. This is surprising given that autophagy is generally viewed as protective during aging. Since Atg7 mutants are short-lived, the deceleration of epidermal aging in this mutant suggests that in the epidermis healthspan can be uncoupled from longevity. Because the aging readout we introduce here has an early onset and is easily visualized, genetic dissection using our model should identify other novel mechanisms by which lifespan genes feed into tissue-specific aging.
Collapse
Affiliation(s)
- Christoph Scherfer
- Department of Biochemistry and Molecular Biology, The University of Texas MD Anderson Cancer Center, Unit 1000, Houston, TX 77030, USA
| | | | | | | | | |
Collapse
|
205
|
Reales-Calderón JA, Sylvester M, Strijbis K, Jensen ON, Nombela C, Molero G, Gil C. Candida albicans induces pro-inflammatory and anti-apoptotic signals in macrophages as revealed by quantitative proteomics and phosphoproteomics. J Proteomics 2013; 91:106-35. [DOI: 10.1016/j.jprot.2013.06.026] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2013] [Accepted: 06/16/2013] [Indexed: 12/11/2022]
|
206
|
Li L, Du Y, Kong X, Li Z, Jia Z, Cui J, Gao J, Wang G, Xie K. Lamin B1 is a novel therapeutic target of betulinic acid in pancreatic cancer. Clin Cancer Res 2013; 19:4651-61. [PMID: 23857605 PMCID: PMC3800003 DOI: 10.1158/1078-0432.ccr-12-3630] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
PURPOSE Betulinic acid, a naturally occurring pentacyclic triterpenoid, exhibits potent antitumor activities, whereas the underlying mechanisms remain unclear. In the current study, we sought to determine the role and regulation of lamin B1 expression in human pancreatic cancer pathogenesis and betulinic acid-based therapy. EXPERIMENTAL DESIGN We used cDNA microarray to identify betulinic acid target genes and used tissue microarray to determine the expression levels of lamin B1 in pancreatic cancer tissues and to define their relationship with the clinicopathologic characteristics of pancreatic cancer. We also used in vitro and in vivo models to determine the biologic impacts of altered lamin B1 expression on and mechanisms underlying lamin B1 overexpression in human pancreatic cancer. RESULTS We found that lamin B1 was significantly downregulated by betulinic acid treatment in pancreatic cancer in both in vitro culture and xenograft models. Overexpression of lamin B1 was pronounced in human pancreatic cancer, and increased lamin B1 expression was directly associated with low-grade differentiation, increased incidence of distant metastasis, and poor prognosis of patients with pancreatic cancer. Furthermore, knockdown of lamin B1 significantly attenuated the proliferation, invasion, and tumorigenicity of pancreatic cancer cells. CONCLUSIONS Lamin B1 plays an important role in pancreatic cancer pathogenesis and is a novel therapeutic target of betulinic acid treatment.
Collapse
Affiliation(s)
- Lei Li
- Departments of Gastroenterology, Shanghai Changhai Hospital, Second Military Medical University, Shanghai, The People's Republic of China
| | - Yiqi Du
- Departments of Gastroenterology, Shanghai Changhai Hospital, Second Military Medical University, Shanghai, The People's Republic of China
| | - Xiangyu Kong
- Departments of Gastroenterology, Shanghai Changhai Hospital, Second Military Medical University, Shanghai, The People's Republic of China
- Department of Gastroenterology and Hepatology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Zhaoshen Li
- Departments of Gastroenterology, Shanghai Changhai Hospital, Second Military Medical University, Shanghai, The People's Republic of China
| | - Zhiliang Jia
- Department of Gastroenterology and Hepatology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jiujie Cui
- Department of Gastroenterology and Hepatology, The University of Texas MD Anderson Cancer Center, Houston, Texas
- Department of Oncology, Shanghai Jiaotong University Affiliated First People's Hospital, Shanghai, The People's Republic of China
| | - Jun Gao
- Departments of Gastroenterology, Shanghai Changhai Hospital, Second Military Medical University, Shanghai, The People's Republic of China
| | - Guokun Wang
- Departments of Cardiology, Shanghai Changhai Hospital, Second Military Medical University, Shanghai, The People's Republic of China
| | - Keping Xie
- Department of Gastroenterology and Hepatology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
207
|
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: 1.7] [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
|
208
|
Rivera-Torres J, Acín-Perez R, Cabezas-Sánchez P, Osorio FG, Gonzalez-Gómez C, Megias D, Cámara C, López-Otín C, Enríquez JA, Luque-García JL, Andrés V. Identification of mitochondrial dysfunction in Hutchinson-Gilford progeria syndrome through use of stable isotope labeling with amino acids in cell culture. J Proteomics 2013; 91:466-77. [PMID: 23969228 DOI: 10.1016/j.jprot.2013.08.008] [Citation(s) in RCA: 96] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2013] [Revised: 08/01/2013] [Accepted: 08/08/2013] [Indexed: 01/03/2023]
Abstract
UNLABELLED Hutchinson-Gilford progeria syndrome (HGPS) is a rare segmental premature aging disorder that recapitulates some biological and physical aspects of physiological aging. The disease is caused by a sporadic dominant mutation in the LMNA gene that leads to the expression of progerin, a mutant form of lamin A that lacks 50 amino acids and retains a toxic farnesyl modification in its carboxy-terminus. However, the mechanisms underlying cellular damage and senescence and accelerated aging in HGPS are incompletely understood. Here, we analyzed fibroblasts from healthy subjects and HGPS patients using SILAC (stable isotope labeling with amino acids in cell culture). We found in HGPS cells a marked downregulation of mitochondrial oxidative phosphorylation proteins accompanied by mitochondrial dysfunction, a process thought to provoke broad organ decline during normal aging. We also found mitochondrial dysfunction in fibroblasts from adult progeroid mice expressing progerin (Lmna(G609G/G609G) knock-in mice) or prelamin A (Zmpste24-null mice). Analysis of tissues from these mouse models revealed that the damaging effect of these proteins on mitochondrial function is time- and dose-dependent. Mitochondrial alterations were not observed in the brain, a tissue with extremely low progerin expression that seems to be unaffected in HGPS. Remarkably, mitochondrial function was restored in progeroid mouse fibroblasts treated with the isoprenylation inhibitors FTI-277 or pravastatin plus zoledronate, which are being tested in HGPS clinical trials. Our results suggest that mitochondrial dysfunction contributes to premature organ decline and aging in HGPS. Beyond its effects on progeria, prelamin A and progerin may also contribute to mitochondrial dysfunction and organ damage during normal aging, since these proteins are expressed in cells and tissues from non-HGPS individuals, most prominently at advanced ages. BIOLOGICAL SIGNIFICANCE Mutations in LMNA or defective processing of prelamin A causes premature aging disorders, including Hutchinson-Gilford progeria syndrome (HGPS). Most HGPS patients carry in heterozygosis a de-novo point mutation (c.1824C>T: GGC>GGT; p.G608G) which causes the expression of the lamin A mutant protein called progerin. Despite the importance of progerin and prelamin A in accelerated aging, the underlying molecular mechanisms remain largely unknown. To tackle this question, we compared the proteome of skin-derived dermal fibroblast from HGPS patients and age-matched controls using quantitative stable isotope labeling with amino acids in cell culture (SILAC). Our results show a pronounced down-regulation of several components of the mitochondrial ATPase complex accompanied by up-regulation of some glycolytic enzymes. Accordingly, functional studies demonstrated mitochondrial dysfunction in HGPS fibroblasts. Moreover, our expression and functional studies using cellular and animal models confirmed that mitochondrial dysfunction is a feature of progeria which develops in a time- and dose-dependent manner. Finally, we demonstrate improved mitochondrial function in progeroid mouse cells treated with a combination of statins and aminobisphosphonates, two drugs that are being evaluated in ongoing HGPS clinical trials. Although further studies are needed to unravel the mechanisms through which progerin and prelamin A provoke mitochondrial abnormalities, our findings may pave the way to improved treatments of HGPS. These studies may also improve our knowledge of the mechanisms leading to mitochondrial dysfunction during normal aging, since both progerin and prelamin A have been found to accumulate during normal aging.
Collapse
Key Words
- ATP synthase, H+ transporting, mitochondrial F0 complex, subunit B1
- ATP synthase, H+ transporting, mitochondrial F1 complex, O subunit
- ATP synthase, H+ transporting, mitochondrial F1 complex, alpha subunit 1
- ATP synthase, H+ transporting, mitochondrial F1 complex, beta polypeptide
- ATP synthase, H+ transporting, mitochondrial F1 complex, gamma polypeptide
- ATP5A1
- ATP5B
- ATP5C1
- ATP5F1
- ATP5O
- Accelerated aging
- COX
- CS
- ENO2
- FTI
- FpSDH
- HGPS
- Hutchinson–Gilford progeria syndrome
- Lamin A
- MAF
- Mitochondrial dysfunction
- Molecular biology of aging
- OXPHOS
- PKM
- Progerin
- SILAC
- Zmpste24
- citrate synthase
- cytochrome c oxidase
- eIF2
- eIF4
- enolase 2
- eukaryotic translation initiation factor 2
- eukaryotic translation initiation factor 4
- farnesyltransferase inhibitor
- flavoprotein subunit of succinate dehydrogenase
- mTOR
- mammalian target of rapamycin
- mouse adult fibroblast
- oxidative phosphorylation
- p70S6K
- pyruvate kinase, muscle
- ribosomal protein S6 kinase, 70kDa, polypeptide 1
- stable isotope labeling with amino acids in cell culture
- zinc metalloproteinase STE24 homolog
Collapse
Affiliation(s)
- José Rivera-Torres
- Department of Epidemiology, Atherothrombosis and Imaging, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
209
|
Holland A, Dowling P, Zweyer M, Swandulla D, Henry M, Clynes M, Ohlendieck K. Proteomic profiling of cardiomyopathic tissue from the aged mdx model of Duchenne muscular dystrophy reveals a drastic decrease in laminin, nidogen and annexin. Proteomics 2013; 13:2312-23. [PMID: 23713012 DOI: 10.1002/pmic.201200578] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2012] [Revised: 04/11/2013] [Accepted: 04/24/2013] [Indexed: 01/07/2023]
Abstract
The majority of patients afflicted with Duchenne muscular dystrophy develop cardiomyopathic complications, warranting large-scale proteomic studies of global cardiac changes for the identification of new protein markers of dystrophinopathy. The aged heart from the X-linked dystrophic mdx mouse has been shown to exhibit distinct pathological aspects of cardiomyopathy. In order to establish age-related alterations in the proteome of dystrophin-deficient hearts, cardiomyopathic tissue from young versus aged mdx mice was examined by label-free LC-MS/MS. Significant age-dependent alterations were established for 67 proteins, of which 28 proteins were shown to exhibit a lower abundance and 39 proteins were found to be increased in their expression levels. Drastic changes were demonstrated for 17 proteins, including increases in Ig chains and transferrin, and drastic decreases in laminin, nidogen and annexin. An immunblotting survey of young and old wild-type versus mdx hearts confirmed these proteomic findings and illustrated the effects of natural aging versus dystrophin deficiency. These proteome-wide alterations suggest a disintegration of the basal lamina structure and cytoskeletal network in dystrophin-deficient cardiac fibres, increased levels of antibodies in a potential autoimmune reaction of the degenerating heart, compensatory binding of excess iron and a general perturbation of metabolic pathways in dystrophy-associated cardiomyopathy.
Collapse
Affiliation(s)
- Ashling Holland
- Department of Biology, National University of Ireland, Maynooth, Ireland
| | | | | | | | | | | | | |
Collapse
|
210
|
Saha B, Zitnik G, Johnson S, Nguyen Q, Risques RA, Martin GM, Oshima J. DNA damage accumulation and TRF2 degradation in atypical Werner syndrome fibroblasts with LMNA mutations. Front Genet 2013; 4:129. [PMID: 23847654 PMCID: PMC3701863 DOI: 10.3389/fgene.2013.00129] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2013] [Accepted: 06/16/2013] [Indexed: 02/01/2023] Open
Abstract
Segmental progeroid syndromes are groups of disorders with multiple features suggestive of accelerated aging. One subset of adult-onset progeroid syndromes, referred to as atypical Werner syndrome, is caused by mutations in the LMNA gene, which encodes a class of nuclear intermediate filaments, lamin A/C. We previously described rapid telomere attrition and accelerated replicative senescence in cultured fibroblasts overexpressing mutant lamin A. In this study, we investigated the cellular phenotypes associated with accelerated telomere shortening in LMNA mutant primary fibroblasts. In early passage primary fibroblasts with R133L or L140R LMNA mutations, shelterin protein components were already reduced while cells still retained telomere lengths comparable to those of controls. There was a significant inverse correlation between the degree of abnormal nuclear morphology and the level of TRF2, a shelterin subunit, suggesting a potential causal relationship. Stabilization of the telomeres via the introduction of the catalytic subunit of human telomerase, hTERT (human telomerase reverse transcriptase), did not prevent degradation of shelterin components, indicating that reduced TRF2 in LMNA mutants is not mediated by short telomeres. Interestingly, γ-H2AX foci (reflecting double strand DNA damage) in early passage LMNA mutant primary fibroblasts and LMNA mutant hTERT fibroblasts were markedly increased in non-telomeric regions of DNA. Our results raise the possibility that mutant lamin A/C causes global genomic instability with accumulation of non-telomeric DNA damage as an early event, followed by TRF2 degradation and telomere shortening.
Collapse
Affiliation(s)
- Bidisha Saha
- Department of Pathology, University of Washington Seattle, WA, USA
| | | | | | | | | | | | | |
Collapse
|
211
|
Al-Haggar M, Shams A, Madej-Pilarczyk A, Barakat T, Puzianowska-Kuznicka M. Ultrastructural skin changes in Egyptian mandibuloacral dysplasia patients with p.Arg527Leu LMNA mutation and in their asymptomatic heterozygotic mothers. J Clin Pathol 2013; 66:1000-4. [PMID: 23775434 DOI: 10.1136/jclinpath-2013-201690] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Affiliation(s)
- Mohammad Al-Haggar
- Genetics Unit, Pediatrics Department, Faculty of Medicine, Mansoura University, , Mansoura, Egypt
| | | | | | | | | |
Collapse
|
212
|
Bhattacharjee P, Banerjee A, Banerjee A, Dasgupta D, Sengupta K. Structural Alterations of Lamin A Protein in Dilated Cardiomyopathy. Biochemistry 2013; 52:4229-41. [DOI: 10.1021/bi400337t] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Pritha Bhattacharjee
- Biophysics
Division, Saha Institute of Nuclear Physics, 1/AF Bidhannagar, Kolkata 700064, India
| | - Avinanda Banerjee
- Biophysics
Division, Saha Institute of Nuclear Physics, 1/AF Bidhannagar, Kolkata 700064, India
| | - Amrita Banerjee
- Biophysics
Division, Saha Institute of Nuclear Physics, 1/AF Bidhannagar, Kolkata 700064, India
| | - Dipak Dasgupta
- Biophysics
Division, Saha Institute of Nuclear Physics, 1/AF Bidhannagar, Kolkata 700064, India
| | - Kaushik Sengupta
- Biophysics
Division, Saha Institute of Nuclear Physics, 1/AF Bidhannagar, Kolkata 700064, India
| |
Collapse
|
213
|
Heng MY, Lin ST, Verret L, Huang Y, Kamiya S, Padiath QS, Tong Y, Palop JJ, Huang EJ, Ptácχek LJ, Fu YH. Lamin B1 mediates cell-autonomous neuropathology in a leukodystrophy mouse model. J Clin Invest 2013; 123:2719-29. [PMID: 23676464 PMCID: PMC3668844 DOI: 10.1172/jci66737] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2012] [Accepted: 03/19/2013] [Indexed: 01/20/2023] Open
Abstract
Adult-onset autosomal-dominant leukodystrophy (ADLD) is a progressive and fatal neurological disorder characterized by early autonomic dysfunction, cognitive impairment, pyramidal tract and cerebellar dysfunction, and white matter loss in the central nervous system. ADLD is caused by duplication of the LMNB1 gene, which results in increased lamin B1 transcripts and protein expression. How duplication of LMNB1 leads to myelin defects is unknown. To address this question, we developed a mouse model of ADLD that overexpresses lamin B1. These mice exhibited cognitive impairment and epilepsy, followed by age-dependent motor deficits. Selective overexpression of lamin B1 in oligodendrocytes also resulted in marked motor deficits and myelin defects, suggesting these deficits are cell autonomous. Proteomic and genome-wide transcriptome studies indicated that lamin B1 overexpression is associated with downregulation of proteolipid protein, a highly abundant myelin sheath component that was previously linked to another myelin-related disorder, Pelizaeus-Merzbacher disease. Furthermore, we found that lamin B1 overexpression leads to reduced occupancy of Yin Yang 1 transcription factor at the promoter region of proteolipid protein. These studies identify a mechanism by which lamin B1 overexpression mediates oligodendrocyte cell-autonomous neuropathology in ADLD and implicate lamin B1 as an important regulator of myelin formation and maintenance during aging.
Collapse
Affiliation(s)
- Mary Y. Heng
- Department of Neurology, UCSF, San Francisco, California, USA.
Gladstone Institute of Neurological Disease, San Francisco,
California, USA. Howard Hughes Medical Institute, San Francisco,
California, USA. Department of Pathology, UCSF, San Francisco,
California, USA. Veterans Affairs Medical Center, San Francisco,
California, USA
| | - Shu-Ting Lin
- Department of Neurology, UCSF, San Francisco, California, USA.
Gladstone Institute of Neurological Disease, San Francisco,
California, USA. Howard Hughes Medical Institute, San Francisco,
California, USA. Department of Pathology, UCSF, San Francisco,
California, USA. Veterans Affairs Medical Center, San Francisco,
California, USA
| | - Laure Verret
- Department of Neurology, UCSF, San Francisco, California, USA.
Gladstone Institute of Neurological Disease, San Francisco,
California, USA. Howard Hughes Medical Institute, San Francisco,
California, USA. Department of Pathology, UCSF, San Francisco,
California, USA. Veterans Affairs Medical Center, San Francisco,
California, USA
| | - Yong Huang
- Department of Neurology, UCSF, San Francisco, California, USA.
Gladstone Institute of Neurological Disease, San Francisco,
California, USA. Howard Hughes Medical Institute, San Francisco,
California, USA. Department of Pathology, UCSF, San Francisco,
California, USA. Veterans Affairs Medical Center, San Francisco,
California, USA
| | - Sherry Kamiya
- Department of Neurology, UCSF, San Francisco, California, USA.
Gladstone Institute of Neurological Disease, San Francisco,
California, USA. Howard Hughes Medical Institute, San Francisco,
California, USA. Department of Pathology, UCSF, San Francisco,
California, USA. Veterans Affairs Medical Center, San Francisco,
California, USA
| | - Quasar S. Padiath
- Department of Neurology, UCSF, San Francisco, California, USA.
Gladstone Institute of Neurological Disease, San Francisco,
California, USA. Howard Hughes Medical Institute, San Francisco,
California, USA. Department of Pathology, UCSF, San Francisco,
California, USA. Veterans Affairs Medical Center, San Francisco,
California, USA
| | - Ying Tong
- Department of Neurology, UCSF, San Francisco, California, USA.
Gladstone Institute of Neurological Disease, San Francisco,
California, USA. Howard Hughes Medical Institute, San Francisco,
California, USA. Department of Pathology, UCSF, San Francisco,
California, USA. Veterans Affairs Medical Center, San Francisco,
California, USA
| | - Jorge J. Palop
- Department of Neurology, UCSF, San Francisco, California, USA.
Gladstone Institute of Neurological Disease, San Francisco,
California, USA. Howard Hughes Medical Institute, San Francisco,
California, USA. Department of Pathology, UCSF, San Francisco,
California, USA. Veterans Affairs Medical Center, San Francisco,
California, USA
| | - Eric J. Huang
- Department of Neurology, UCSF, San Francisco, California, USA.
Gladstone Institute of Neurological Disease, San Francisco,
California, USA. Howard Hughes Medical Institute, San Francisco,
California, USA. Department of Pathology, UCSF, San Francisco,
California, USA. Veterans Affairs Medical Center, San Francisco,
California, USA
| | - Louis J. Ptácχek
- Department of Neurology, UCSF, San Francisco, California, USA.
Gladstone Institute of Neurological Disease, San Francisco,
California, USA. Howard Hughes Medical Institute, San Francisco,
California, USA. Department of Pathology, UCSF, San Francisco,
California, USA. Veterans Affairs Medical Center, San Francisco,
California, USA
| | - Ying-Hui Fu
- Department of Neurology, UCSF, San Francisco, California, USA.
Gladstone Institute of Neurological Disease, San Francisco,
California, USA. Howard Hughes Medical Institute, San Francisco,
California, USA. Department of Pathology, UCSF, San Francisco,
California, USA. Veterans Affairs Medical Center, San Francisco,
California, USA
| |
Collapse
|
214
|
Renes J, Mariman E. Application of proteomics technology in adipocyte biology. MOLECULAR BIOSYSTEMS 2013; 9:1076-91. [PMID: 23629546 DOI: 10.1039/c3mb25596d] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Obesity and its associated complications have reached epidemic proportions in Western-type societies. Concomitantly, the obesity incidence in developing countries is increasing. One hallmark of obesity is the differentiation of pre-adipocytes into mature triglyceride-loaded adipocytes present in subcutaneous and visceral adipose tissue depots. This may ultimately lead to dysfunctional adipose tissue together with detrimental changes in the profiles of (pre-)adipocyte-secreted proteins, known as adipokines. Obesity-induced alterations in adipokine profiles contribute to the development of obesity-associated disorders. Consequently, the interest in the molecular events responsible for adipose tissue modifications during weight gain and weight loss as well as in the aetiology of obesity-associated disorders is growing. Molecular mechanisms involved in pre-adipocyte differentiation and alterations in adipokine profiles have been examined at the gene and protein level by high-throughput technologies. Independent proteomics studies have contributed significantly to further insight into adipocyte biology, particularly with respect to adipokine profiling. In this review novel findings obtained with adipo-proteomics studies are highlighted and the relevance of proteomics technologies to further understand molecular aspects of adipocyte biology is discussed.
Collapse
Affiliation(s)
- Johan Renes
- Department of Human Biology, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands.
| | | |
Collapse
|
215
|
Cattin ME, Bertrand AT, Schlossarek S, Le Bihan MC, Skov Jensen S, Neuber C, Crocini C, Maron S, Lainé J, Mougenot N, Varnous S, Fromes Y, Hansen A, Eschenhagen T, Decostre V, Carrier L, Bonne G. Heterozygous LmnadelK32 mice develop dilated cardiomyopathy through a combined pathomechanism of haploinsufficiency and peptide toxicity. Hum Mol Genet 2013; 22:3152-64. [DOI: 10.1093/hmg/ddt172] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
|
216
|
Sehgal P, Chaturvedi P, Kumaran RI, Kumar S, Parnaik VK. Lamin A/C haploinsufficiency modulates the differentiation potential of mouse embryonic stem cells. PLoS One 2013; 8:e57891. [PMID: 23451281 PMCID: PMC3581495 DOI: 10.1371/journal.pone.0057891] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2012] [Accepted: 01/28/2013] [Indexed: 02/06/2023] Open
Abstract
Background Lamins are structural proteins that are the major determinants of nuclear architecture and play important roles in various nuclear functions including gene regulation and cell differentiation. Mutations in the human lamin A gene cause a spectrum of genetic diseases that affect specific tissues. Most available mouse models for laminopathies recapitulate disease symptoms for muscle diseases and progerias. However, loss of human lamin A/C also has highly deleterious effects on fetal development. Hence it is important to understand the impact of lamin A/C expression levels on embryonic differentiation pathways. Methodology and Principal Findings We have investigated the differentiation potential of mouse embryonic stem cells containing reduced levels of lamin A/C by detailed lineage analysis of embryoid bodies derived from these cells by in vitro culture. We initially carried out a targeted disruption of one allele of the mouse lamin A/C gene (Lmna). Undifferentiated wild-type and Lmna+/− embryonic stem cells showed similar expression of pluripotency markers and cell cycle profiles. Upon spontaneous differentiation into embryoid bodies, markers for visceral endoderm such as α-fetoprotein were highly upregulated in haploinsufficient cells. However, neuronal markers such as β-III tubulin and nestin were downregulated. Furthermore, we observed a reduction in the commitment of Lmna+/− cells into the myogenic lineage, but no discernible effects on cardiac, adipocyte or osteocyte lineages. In the next series of experiments, we derived embryonic stem cell clones expressing lamin A/C short hairpin RNA and examined their differentiation potential. These cells expressed pluripotency markers and, upon differentiation, the expression of lineage-specific markers was altered as observed with Lmna+/− embryonic stem cells. Conclusions We have observed significant effects on embryonic stem cell differentiation to visceral endoderm, neuronal and myogenic lineages upon depletion of lamin A/C. Hence our results implicate lamin A/C level as an important determinant of lineage-specific differentiation during embryonic development.
Collapse
Affiliation(s)
- Poonam Sehgal
- CSIR-Centre for Cellular and Molecular Biology, Hyderabad, India
| | | | - R. Ileng Kumaran
- CSIR-Centre for Cellular and Molecular Biology, Hyderabad, India
| | - Satish Kumar
- CSIR-Centre for Cellular and Molecular Biology, Hyderabad, India
| | - Veena K. Parnaik
- CSIR-Centre for Cellular and Molecular Biology, Hyderabad, India
- * E-mail:
| |
Collapse
|
217
|
Lamin A/C depletion enhances DNA damage-induced stalled replication fork arrest. Mol Cell Biol 2013; 33:1210-22. [PMID: 23319047 DOI: 10.1128/mcb.01676-12] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
The human LMNA gene encodes the essential nuclear envelope proteins lamin A and C (lamin A/C). Mutations in LMNA result in altered nuclear morphology, but how this impacts the mechanisms that maintain genomic stability is unclear. Here, we report that lamin A/C-deficient cells have a normal response to ionizing radiation but are sensitive to agents that cause interstrand cross-links (ICLs) or replication stress. In response to treatment with ICL agents (cisplatin, camptothecin, and mitomycin), lamin A/C-deficient cells displayed normal γ-H2AX focus formation but a higher frequency of cells with delayed γ-H2AX removal, decreased recruitment of the FANCD2 repair factor, and a higher frequency of chromosome aberrations. Similarly, following hydroxyurea-induced replication stress, lamin A/C-deficient cells had an increased frequency of cells with delayed disappearance of γ-H2AX foci and defective repair factor recruitment (Mre11, CtIP, Rad51, RPA, and FANCD2). Replicative stress also resulted in a higher frequency of chromosomal aberrations as well as defective replication restart. Taken together, the data can be interpreted to suggest that lamin A/C has a role in the restart of stalled replication forks, a prerequisite for initiation of DNA damage repair by the homologous recombination pathway, which is intact in lamin A/C-deficient cells. We propose that lamin A/C is required for maintaining genomic stability following replication fork stalling, induced by either ICL damage or replicative stress, in order to facilitate fork regression prior to DNA damage repair.
Collapse
|
218
|
Bonne G, Quijano-Roy S. Emery-Dreifuss muscular dystrophy, laminopathies, and other nuclear envelopathies. HANDBOOK OF CLINICAL NEUROLOGY 2013; 113:1367-76. [PMID: 23622360 DOI: 10.1016/b978-0-444-59565-2.00007-1] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
The nuclear envelopathies, more frequently known as laminopathies are a rapidly expanding group of human hereditary diseases caused by mutations of genes that encode proteins of the nuclear envelope. The most frequent and best known form is Emery-Dreifuss muscular dystrophy (EDMD), a skeletal myopathy characterized by progressive muscular weakness, joint contractures, and cardiac disease. EMD gene, encoding emerin, causes the X-linked form of EDMD, while LMNA gene encoding lamins A and C, is responsible for autosomal forms, usually with a dominant transmission. In the last years, the spectrum of conditions has been extraordinarily enlarged, from a congenital muscular dystrophy with severe paralytic or rapidly progressive picture due to de novo mutations in LMNA (L-CMD) to a limb-girdle muscular dystrophy with adult onset and much milder weakness (LGMD1B). LMNA has also been involved in a form of isolated cardiomyopathy associated with cardiac conduction disease and in an axonal form of hereditary neuropathy. Identification of this gene has been reported also in a number of non-neuromuscular disorders including lipodystrophy syndromes and a wide spectrum of premature aging syndromes ranging from mandibuloacral dysplasia to restrictive dermopathy. Mutations in other genes implicated in the processing or maturation of nuclear lamins have also been found. The extraordinary complexity of the molecular and pathophysiological mechanisms of these diseases is still not well known and the occurrence of modifying factors or genes is highly suspected. Identification of new genes and investigation of new therapeutic approaches are in progress.
Collapse
Affiliation(s)
- Gisèle Bonne
- Inserm, U974; Université Pierre et Marie Curie - Paris 6, UM 76; CNRS, UMR 7215; Institut de Myologie, and AP-HP - U.F. Cardiogénétique et Myogénétique, Groupe Hospitalier Pitié-Salpêtrière, Paris, France.
| | | |
Collapse
|
219
|
Tamiello C, Kamps MAF, van den Wijngaard A, Verstraeten VLRM, Baaijens FPT, Broers JLV, Bouten CCV. Soft substrates normalize nuclear morphology and prevent nuclear rupture in fibroblasts from a laminopathy patient with compound heterozygous LMNA mutations. Nucleus 2013; 4:61-73. [PMID: 23324461 PMCID: PMC3585029 DOI: 10.4161/nucl.23388] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Laminopathies, mainly caused by mutations in the LMNA gene, are a group of inherited diseases with a highly variable penetrance; i.e., the disease spectrum in persons with identical LMNA mutations range from symptom-free conditions to severe cardiomyopathy and progeria, leading to early death. LMNA mutations cause nuclear abnormalities and cellular fragility in response to cellular mechanical stress, but the genotype/phenotype correlations in these diseases remain unclear. Consequently, tools such as mutation analysis are not adequate for predicting the course of the disease.
Here, we employ growth substrate stiffness to probe nuclear fragility in cultured dermal fibroblasts from a laminopathy patient with compound progeroid syndrome. We show that culturing of these cells on substrates with stiffness higher than 10 kPa results in malformations and even rupture of the nuclei, while culture on a soft substrate (3 kPa) protects the nuclei from morphological alterations and ruptures. No malformations were seen in healthy control cells at any substrate stiffness. In addition, analysis of the actin cytoskeleton organization in this laminopathy cells demonstrates that the onset of nuclear abnormalities correlates to an increase in cytoskeletal tension.
Together, these data indicate that culturing of these LMNA mutated cells on substrates with a range of different stiffnesses can be used to probe the degree of nuclear fragility. This assay may be useful in predicting patient-specific phenotypic development and in investigations on the underlying mechanisms of nuclear and cellular fragility in laminopathies.
Collapse
Affiliation(s)
- Chiara Tamiello
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands.
| | | | | | | | | | | | | |
Collapse
|
220
|
Perrin S, Cremer J, Faucher O, Reynes J, Dellamonica P, Micallef J, Solas C, Lacarelle B, Stretti C, Kaspi E, Robaglia-Schlupp A, Tamalet CNBC, Lévy N, Poizot-Martin I, Cau P, Roll P. HIV protease inhibitors do not cause the accumulation of prelamin A in PBMCs from patients receiving first line therapy: the ANRS EP45 "aging" study. PLoS One 2012; 7:e53035. [PMID: 23285253 PMCID: PMC3532351 DOI: 10.1371/journal.pone.0053035] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2012] [Accepted: 11/22/2012] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND The ANRS EP45 "Aging" study investigates the cellular mechanisms involved in the accelerated aging of HIV-1 infected and treated patients. The present report focuses on lamin A processing, a pathway known to be altered in systemic genetic progeroid syndromes. METHODS 35 HIV-1 infected patients being treated with first line antiretroviral therapy (ART, mean duration at inclusion: 2.7±1.3 years) containing boosted protease inhibitors (PI/r) (comprising lopinavir/ritonavir in 65% of patients) were recruited together with 49 seronegative age- and sex-matched control subjects (http://clinicaltrials.gov/, NCT01038999). In more than 88% of patients, the viral load was <40 copies/ml and the CD4+ cell count was >500/mm³. Prelamin A processing in peripheral blood mononuclear cells (PBMCs) from patients and controls was analysed by western blotting at inclusion. PBMCs from patients were also investigated at 12 and 24 months after enrolment in the study. PBMCs from healthy controls were also incubated with boosted lopinavir in culture medium containing various concentrations of proteins (4 to 80 g/L). RESULTS Lamin A precursor was not observed in cohort patient PBMC regardless of the PI/r used, the dose and the plasma concentration. Prelamin A was detected in PBMC incubated in culture medium containing a low protein concentration (4 g/L) but not in plasma (60-80 g/L) or in medium supplemented with BSA (40 g/L), both of which contain a high protein concentration. CONCLUSIONS Prelamin A processing abnormalities were not observed in PBMCs from patients under the PI/r first line regimen. Therefore, PI/r do not appear to contribute to lamin A-related aging in PBMCs. In cultured PBMCs from healthy donors, prelamin A processing abnormalities were only observed when the protein concentration in the culture medium was low, thus increasing the amount of PI available to enter cells. ClinicalTrials.gov NCT01038999 http://clinicaltrials.gov/ct2/show/NCT01038999.
Collapse
Affiliation(s)
- Sophie Perrin
- Inserm UMR_S 910, Aix-Marseille Université, Marseille, France
- Laboratoire de Biologie Cellulaire, Centre Hospitalier Universitaire (CHU) La Timone Assistance Publique des Hôpitaux de Marseille (APHM), Marseille, France
| | - Jonathan Cremer
- Inserm UMR_S 910, Aix-Marseille Université, Marseille, France
- Laboratoire de Biologie Cellulaire, Centre Hospitalier Universitaire (CHU) La Timone Assistance Publique des Hôpitaux de Marseille (APHM), Marseille, France
| | - Olivia Faucher
- Service d’Immuno-Hématologie Clinique, Centre Hospitalier Universitaire (CHU) Sainte Marguerite Assistance Publique des Hôpitaux de Marseille (APHM), Marseille, France
| | - Jacques Reynes
- Département des Maladies Infectieuses et Tropicales, Centre Hospitalier Régional et Universitaire (CHRU) Gui-de-Chauliac, Montpellier, France
| | - Pierre Dellamonica
- Service d’Infectiologie, Centre Hospitalier Universitaire (CHU) L’Archet 1, Sophia-Antipolis Université, Nice, France
| | - Joëlle Micallef
- Centre d’Investigation Clinique - Unité de Pharmacologie Clinique et d’Evaluations Thérapeutiques (CIC-UPCET), Centre Hospitalier Universitaire (CHU) La Timone Assistance Publique des Hôpitaux de Marseille (APHM), Marseille, France
| | - Caroline Solas
- Laboratoire de Pharmacocinétique et de Toxicologie, Centre Hospitalier Universitaire (CHU) La Timone Assistance Publique des Hôpitaux de Marseille (APHM), Marseille, France
- Inserm UMR_S 911, Aix-Marseille Université, Marseille, France
| | - Bruno Lacarelle
- Laboratoire de Pharmacocinétique et de Toxicologie, Centre Hospitalier Universitaire (CHU) La Timone Assistance Publique des Hôpitaux de Marseille (APHM), Marseille, France
- Inserm UMR_S 911, Aix-Marseille Université, Marseille, France
| | - Charlotte Stretti
- Inserm UMR_S 910, Aix-Marseille Université, Marseille, France
- Laboratoire de Biologie Cellulaire, Centre Hospitalier Universitaire (CHU) La Timone Assistance Publique des Hôpitaux de Marseille (APHM), Marseille, France
| | - Elise Kaspi
- Inserm UMR_S 910, Aix-Marseille Université, Marseille, France
- Laboratoire de Biologie Cellulaire, Centre Hospitalier Universitaire (CHU) La Timone Assistance Publique des Hôpitaux de Marseille (APHM), Marseille, France
| | - Andrée Robaglia-Schlupp
- Inserm UMR_S 910, Aix-Marseille Université, Marseille, France
- Laboratoire de Biologie Cellulaire, Centre Hospitalier Universitaire (CHU) La Timone Assistance Publique des Hôpitaux de Marseille (APHM), Marseille, France
| | | | - Nicolas Lévy
- Inserm UMR_S 910, Aix-Marseille Université, Marseille, France
- Laboratoire de Génetique Moléculaire, Centre Hospitalier Universitaire (CHU) La Timone Assistance Publique des Hôpitaux de Marseille (APHM), Marseille, France
| | - Isabelle Poizot-Martin
- Département des Maladies Infectieuses et Tropicales, Centre Hospitalier Régional et Universitaire (CHRU) Gui-de-Chauliac, Montpellier, France
| | - Pierre Cau
- Inserm UMR_S 910, Aix-Marseille Université, Marseille, France
- Laboratoire de Biologie Cellulaire, Centre Hospitalier Universitaire (CHU) La Timone Assistance Publique des Hôpitaux de Marseille (APHM), Marseille, France
| | - Patrice Roll
- Inserm UMR_S 910, Aix-Marseille Université, Marseille, France
- Laboratoire de Biologie Cellulaire, Centre Hospitalier Universitaire (CHU) La Timone Assistance Publique des Hôpitaux de Marseille (APHM), Marseille, France
- * E-mail:
| |
Collapse
|
221
|
Abstract
Lamins are the major components of the nuclear lamina, a filamentous layer found at the interphase between chromatin and the inner nuclear membrane. The lamina supports the nuclear envelope and provides anchorage sites for chromatin. Lamins and their associated proteins are required for most nuclear activities, mitosis, and for linking the nucleoskeleton to the network of cytoskeletal filaments. Mutations in lamins and their associated proteins give rise to a wide range of diseases, collectively called laminopathies. This review focuses on the evolution of the lamin protein family. Evolution from basal metazoans to man will be described on the basis of protein sequence comparisons and analyses of their gene structure. Lamins are the founding members of the family of intermediate filament proteins. How genes encoding cytoplasmic IF proteins could have arisen from the archetypal lamin gene progenitor, can be inferred from a comparison of the respective gene structures. The lamin/IF protein family seems to be restricted to the metazoans. In general, invertebrate genomes harbor only a single lamin gene encoding a B-type lamin. The archetypal lamin gene structure found in basal metazoans is conserved up to the vertebrate lineage. The completely different structure of lamin genes in Caenorhabditis and Drosophila are exceptions rather than the rule within their systematic groups. However, variation in the length of the coiled-coil forming central domain might be more common than previously anticipated. The increase in the number of lamin genes in vertebrates can be explained by two rounds of genome duplication. The origin of lamin A by exon shuffling might explain the processing of prelamin A to the mature non-isoprenylated form of lamin A. By alternative splicing the number of vertebrate lamin proteins has increased even further. Lamin C, an alternative splice form of the LMNA gene, is restricted to mammals. Amphibians and mammals express germline-specific lamins that differ in their protein structure from that of somatic lamins. Evidence is provided that there exist lamin-like proteins outside the metazoan lineage.
Collapse
Affiliation(s)
- Annette Peter
- Department for Cell Biology, University of Bremen, Bremen, Germany
| | | |
Collapse
|
222
|
Zuela N, Bar DZ, Gruenbaum Y. Lamins in development, tissue maintenance and stress. EMBO Rep 2012; 13:1070-8. [PMID: 23146893 PMCID: PMC3512410 DOI: 10.1038/embor.2012.167] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2012] [Accepted: 10/01/2012] [Indexed: 12/24/2022] Open
Abstract
Lamins are nuclear intermediate filament proteins. They provide mechanical stability, organize chromatin and regulate transcription, replication, nuclear assembly and nuclear positioning. Recent studies provide new insights into the role of lamins in development, differentiation and tissue response to mechanical, reactive oxygen species and thermal stresses. These studies also propose the existence of separate filament networks for A- and B-type lamins and identify new roles for the different networks. Furthermore, they show changes in lamin composition in different cell types, propose explanations for the more than 14 distinct human diseases caused by lamin A and lamin C mutations and propose a role for lamin B1 in these diseases.
Collapse
Affiliation(s)
- Noam Zuela
- Department of Genetics, Institute of Life Sciences, Hebrew University of Jerusalem, Jerusalem 91904, Israel
| | - Daniel Z Bar
- Department of Genetics, Institute of Life Sciences, Hebrew University of Jerusalem, Jerusalem 91904, Israel
| | - Yosef Gruenbaum
- Department of Genetics, Institute of Life Sciences, Hebrew University of Jerusalem, Jerusalem 91904, Israel
| |
Collapse
|
223
|
Affiliation(s)
- Chin Yee Ho
- Cornell University, Weill Institute for Cell and Molecular Biology, Department of Biomedical Engineering, Ithaca, NY 14853, USA
| | | |
Collapse
|
224
|
Chaturvedi P, Khanna R, Parnaik VK. Ubiquitin ligase RNF123 mediates degradation of heterochromatin protein 1α and β in lamin A/C knock-down cells. PLoS One 2012; 7:e47558. [PMID: 23077635 PMCID: PMC3471868 DOI: 10.1371/journal.pone.0047558] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2012] [Accepted: 09/17/2012] [Indexed: 11/19/2022] Open
Abstract
Background The nuclear lamina is a key determinant of nuclear architecture, integrity and functionality in metazoan nuclei. Mutations in the human lamin A gene lead to highly debilitating genetic diseases termed as laminopathies. Expression of lamin A mutations or reduction in levels of endogenous A-type lamins leads to nuclear defects such as abnormal nuclear morphology and disorganization of heterochromatin. This is accompanied by increased proteasomal degradation of certain nuclear proteins such as emerin, nesprin-1α, retinoblastoma protein and heterochromatin protein 1 (HP1). However, the pathways of proteasomal degradation have not been well characterized. Methodology/Principal Findings To investigate the mechanisms underlying the degradation of HP1 proteins upon lamin misexpression, we analyzed the effects of shRNA-mediated knock-down of lamins A and C in HeLa cells. Cells with reduced levels of expression of lamins A and C exhibited proteasomal degradation of HP1α and HP1β but not HP1γ. Since specific ubiquitin ligases are upregulated in lamin A/C knock-down cells, further studies were carried out with one of these ligases, RNF123, which has a putative HP1-binding motif. Ectopic expression of GFP-tagged RNF123 directly resulted in degradation of HP1α and HP1β. Mutational analysis showed that the canonical HP1-binding pentapeptide motif PXVXL in the N-terminus of RNF123 was required for binding to HP1 proteins and targeting them for degradation. The role of endogenous RNF123 in the degradation of HP1 isoforms was confirmed by RNF123 RNAi experiments. Furthermore, FRAP analysis suggested that HP1β was displaced from chromatin in laminopathic cells. Conclusions/Significance Our data support a role for RNF123 ubiquitin ligase in the degradation of HP1α and HP1β upon lamin A/C knock-down. Hence lamin misexpression can cause degradation of mislocalized proteins involved in key nuclear processes by induction of specific components of the ubiquitin-proteasome system.
Collapse
Affiliation(s)
| | - Richa Khanna
- Centre for Cellular and Molecular Biology (CSIR), Hyderabad, India
| | - Veena K. Parnaik
- Centre for Cellular and Molecular Biology (CSIR), Hyderabad, India
- * E-mail:
| |
Collapse
|
225
|
Jung HJ, Lee JM, Yang SH, Young SG, Fong LG. Nuclear lamins in the brain - new insights into function and regulation. Mol Neurobiol 2012; 47:290-301. [PMID: 23065386 DOI: 10.1007/s12035-012-8350-1] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2012] [Accepted: 09/12/2012] [Indexed: 11/28/2022]
Abstract
The nuclear lamina is an intermediate filament meshwork composed largely of four nuclear lamins - lamins A and C (A-type lamins) and lamins B1 and B2 (B-type lamins). Located immediately adjacent to the inner nuclear membrane, the nuclear lamina provides a structural scaffolding for the cell nucleus. It also interacts with both nuclear membrane proteins and the chromatin and is thought to participate in many important functions within the cell nucleus. Defects in A-type lamins cause cardiomyopathy, muscular dystrophy, peripheral neuropathy, lipodystrophy, and progeroid disorders. In contrast, the only bona fide link between the B-type lamins and human disease is a rare demyelinating disease of the central nervous system - adult-onset autosomal-dominant leukoencephalopathy, caused by a duplication of the gene for lamin B1. However, this leukoencephalopathy is not the only association between the brain and B-type nuclear lamins. Studies of conventional and tissue-specific knockout mice have demonstrated that B-type lamins play essential roles in neuronal migration in the developing brain and in neuronal survival. The importance of A-type lamin expression in the brain is unclear, but it is intriguing that the adult brain preferentially expresses lamin C rather than lamin A, very likely due to microRNA-mediated removal of prelamin A transcripts. Here, we review recent studies on nuclear lamins, focusing on the function and regulation of the nuclear lamins in the central nervous system.
Collapse
Affiliation(s)
- Hea-Jin Jung
- Molecular Biology Institute, University of California-Los Angeles, Los Angeles, CA 90095, USA
| | | | | | | | | |
Collapse
|
226
|
Abstract
Sunlight that reaches the human skin contains solar energy composed of 6.8% ultraviolet (UV), 38.9% visible light and 54.3% infrared radiation. In addition to natural near-infrared (NIR), human skin is increasingly exposed to artificial NIR from medical devices and electrical appliances. Thus, we are exposed to tremendous amounts of NIR. Many studies have proven the effects of UV exposure on human skin and skin cancers but have not investigated well the effects of NIR exposure. Furthermore, many of the previous NIR studies have used NIR resources without a water filter or a contact cooling. With these resources, a substantial amount of NIR energy is absorbed in the superficial layers and only limited NIR energy can be delivered to deeper tissues. Thus, they could not sufficiently evaluate the effects of incident solar NIR. In order to simulate solar NIR that reaches the skin, a water filter is essential because solar NIR is filtered by atmospheric water. In reality, NIR increases the surface temperature and induces thermal effects so a contact cooling is needed to pursue the properties of NIR. I clarify that NIR can penetrate the skin and non-thermally affect the subcutaneous tissues, including muscle and bone marrow, using a NIR resource with a water filter and a cooling system. I would like to emphasize the biological effects of NIR which have both merits and demerits. Appropriate NIR irradiation induces dermal heating thermally and non-thermally induces collagen and elastin stimulation, which results in skin tightening. NIR also induces non-thermal DNA damage of mitotic cells, which may have the potential application for treating cancer. However, as continuous NIR exposure may induce photoaging and potentially photocarcinogenesis, we should consider the effect of, not only UV, but also NIR and the necessity for protection against solar NIR. Here, this paper introduces the new aspects of the biological effects of NIR radiation.
Collapse
|
227
|
Lu W, Schneider M, Neumann S, Jaeger VM, Taranum S, Munck M, Cartwright S, Richardson C, Carthew J, Noh K, Goldberg M, Noegel AA, Karakesisoglou I. Nesprin interchain associations control nuclear size. Cell Mol Life Sci 2012; 69:3493-509. [PMID: 22653047 PMCID: PMC11114684 DOI: 10.1007/s00018-012-1034-1] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2011] [Revised: 04/26/2012] [Accepted: 05/14/2012] [Indexed: 12/12/2022]
Abstract
Nesprins-1/-2/-3/-4 are nuclear envelope proteins, which connect nuclei to the cytoskeleton. The largest nesprin-1/-2 isoforms (termed giant) tether F-actin through their N-terminal actin binding domain (ABD). Nesprin-3, however, lacks an ABD and associates instead to plectin, which binds intermediate filaments. Nesprins are integrated into the outer nuclear membrane via their C-terminal KASH-domain. Here, we show that nesprin-1/-2 ABDs physically and functionally interact with nesprin-3. Thus, both ends of nesprin-1/-2 giant are integrated at the nuclear surface: via the C-terminal KASH-domain and the N-terminal ABD-nesprin-3 association. Interestingly, nesprin-2 ABD or KASH-domain overexpression leads to increased nuclear areas. Conversely, nesprin-2 mini (contains the ABD and KASH-domain but lacks the massive nesprin-2 giant rod segment) expression yields smaller nuclei. Nuclear shrinkage is further enhanced upon nesprin-3 co-expression or microfilament depolymerization. Our findings suggest that multivariate intermolecular nesprin interactions with the cytoskeleton form a lattice-like filamentous network covering the outer nuclear membrane, which determines nuclear size.
Collapse
Affiliation(s)
- Wenshu Lu
- School of Biological and Biomedical Sciences, University of Durham, Durham, DH1 3LE UK
- Center for Biochemistry, Medical Faculty, University of Cologne, Joseph-Stelzmann-Strasse 52, 50931 Cologne, Germany
| | - Maria Schneider
- School of Biological and Biomedical Sciences, University of Durham, Durham, DH1 3LE UK
- Center for Biochemistry, Medical Faculty, University of Cologne, Joseph-Stelzmann-Strasse 52, 50931 Cologne, Germany
| | - Sascha Neumann
- Center for Biochemistry, Medical Faculty, University of Cologne, Joseph-Stelzmann-Strasse 52, 50931 Cologne, Germany
| | - Verena-Maren Jaeger
- School of Biological and Biomedical Sciences, University of Durham, Durham, DH1 3LE UK
- Center for Biochemistry, Medical Faculty, University of Cologne, Joseph-Stelzmann-Strasse 52, 50931 Cologne, Germany
| | - Surayya Taranum
- Center for Biochemistry, Medical Faculty, University of Cologne, Joseph-Stelzmann-Strasse 52, 50931 Cologne, Germany
| | - Martina Munck
- Center for Biochemistry, Medical Faculty, University of Cologne, Joseph-Stelzmann-Strasse 52, 50931 Cologne, Germany
| | - Sarah Cartwright
- School of Biological and Biomedical Sciences, University of Durham, Durham, DH1 3LE UK
| | - Christine Richardson
- School of Biological and Biomedical Sciences, University of Durham, Durham, DH1 3LE UK
| | - James Carthew
- School of Biological and Biomedical Sciences, University of Durham, Durham, DH1 3LE UK
| | - Kowoon Noh
- School of Biological and Biomedical Sciences, University of Durham, Durham, DH1 3LE UK
| | - Martin Goldberg
- School of Biological and Biomedical Sciences, University of Durham, Durham, DH1 3LE UK
| | - Angelika A. Noegel
- Center for Biochemistry, Medical Faculty, University of Cologne, Joseph-Stelzmann-Strasse 52, 50931 Cologne, Germany
- Center for Molecular Medicine Cologne, Medical Faculty, University of Cologne, Joseph-Stelzmann-Strasse 52, 50931 Cologne, Germany
| | | |
Collapse
|
228
|
Sarge KD, Park-Sarge OK. WITHDRAWN: Protein sumoylation and human diseases. Biochimie 2012:S0300-9084(12)00371-9. [PMID: 23022145 DOI: 10.1016/j.biochi.2012.09.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2012] [Accepted: 09/13/2012] [Indexed: 11/26/2022]
Abstract
This review has been withdrawn at the request of the author(s) and/or editor. The Publisher apologizes for any inconvenience this may cause. The full Elsevier Policy on Article Withdrawal can be found at http://www.elsevier.com/locate/withdrawalpolicy.
Collapse
Affiliation(s)
- Kevin D Sarge
- Department of Molecular and Cellular Biochemistry, Chandler Medical Center, University of Kentucky, Lexington, KY 40536, USA.
| | | |
Collapse
|
229
|
Clinical trial of a farnesyltransferase inhibitor in children with Hutchinson-Gilford progeria syndrome. Proc Natl Acad Sci U S A 2012; 109:16666-71. [PMID: 23012407 DOI: 10.1073/pnas.1202529109] [Citation(s) in RCA: 258] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Hutchinson-Gilford progeria syndrome (HGPS) is an extremely rare, fatal, segmental premature aging syndrome caused by a mutation in LMNA that produces the farnesylated aberrant lamin A protein, progerin. This multisystem disorder causes failure to thrive and accelerated atherosclerosis leading to early death. Farnesyltransferase inhibitors have ameliorated disease phenotypes in preclinical studies. Twenty-five patients with HGPS received the farnesyltransferase inhibitor lonafarnib for a minimum of 2 y. Primary outcome success was predefined as a 50% increase over pretherapy in estimated annual rate of weight gain, or change from pretherapy weight loss to statistically significant on-study weight gain. Nine patients experienced a ≥50% increase, six experienced a ≥50% decrease, and 10 remained stable with respect to rate of weight gain. Secondary outcomes included decreases in arterial pulse wave velocity and carotid artery echodensity and increases in skeletal rigidity and sensorineural hearing within patient subgroups. All patients improved in one or more of these outcomes. Results from this clinical treatment trial for children with HGPS provide preliminary evidence that lonafarnib may improve vascular stiffness, bone structure, and audiological status.
Collapse
|
230
|
Lamine C2 et spermatogenèse. Basic Clin Androl 2012. [DOI: 10.1007/s12610-012-0188-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Résumé
Les lamines A/C sont des filaments intermédiaires présents dans les noyaux des cellules. Leurs rôles sont multiples et des mutations du gène LMNA sont à l’origine de nombreuses maladies appelées laminopathies. Dans les cellules germinales masculines, cette famille de protéines n’est représentée que par la lamine C2. Les données obtenues chez la souris démontrent l’importance de ces filaments dans le déroulement de la méiose masculine et présagent de l’existence d’un nouveau domaine d’infertilité d’origine masculine lié à des mutations de ce filament intermédiaire ou de ses protéines associées.
Collapse
|
231
|
Zuo B, Yang J, Wang F, Wang L, Yin Y, Dan J, Liu N, Liu L. Influences of lamin A levels on induction of pluripotent stem cells. Biol Open 2012; 1:1118-27. [PMID: 23213392 PMCID: PMC3507184 DOI: 10.1242/bio.20121586] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2012] [Accepted: 08/08/2012] [Indexed: 01/08/2023] Open
Abstract
Lamin A is an inner nuclear membrane protein that maintains nuclear structure integrity, is involved in transcription, DNA damage response and genomic stability, and also links to cell differentiation, senescence, premature aging and associated diseases. Induced pluripotent stem (iPS) cells have been successfully generated from various types of cells and used to model human diseases. It remains unclear whether levels of lamin A influence reprogramming of somatic cells to pluripotent states during iPS induction. Consistently, lamin A is expressed more in differentiated than in relatively undifferentiated somatic cells, and increases in expression levels with age. Somatic cells with various expression levels of lamin A differ in their dynamics and efficiency during iPS cell induction. Cells with higher levels of lamin A show slower reprogramming and decreased efficiency to iPS cells. Furthermore, depletion of lamin A by transient shRNA accelerates iPS cell induction from fibroblasts. Reduced levels of lamin A are associated with increased expression of pluripotent genes Oct4 and Nanog, and telomerase genes Tert and Terc. On the contrary, overexpression of lamin A retards somatic cell reprogramming to iPS-like colony formation. Our data suggest that levels of lamin A influence reprogramming of somatic cells to pluripotent stem cells and that artificial silencing of lamin A facilitates iPS cell induction. These findings may have implications in enhancing rejuvenation of senescent or older cells by iPS technology and manipulating lamin A levels.
Collapse
Affiliation(s)
- Bingfeng Zuo
- State Key Laboratory of Medicinal Chemical Biology, Department of Genetics and Cell Biology, College of Life Sciences, Nankai University , Tianjin 300071 , China ; Tianjin-Oxford Joint Laboratory of Gene Therapy, Tianjin Research Centre of Basic Medical Science, Tianjin Medical University , Tianjin 300070 , China
| | | | | | | | | | | | | | | |
Collapse
|
232
|
Barascu A, Le Chalony C, Pennarun G, Genet D, Zaarour N, Bertrand P. Oxydative stress alters nuclear shape through lamins dysregulation: a route to senescence. Nucleus 2012; 3:411-7. [PMID: 22895091 PMCID: PMC3474660 DOI: 10.4161/nucl.21674] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Progeroid phenotypes are mainly encountered in 2 types of syndromes: in laminopathies, which are characterized by nuclear shape abnormalities due to lamin A alteration, and in DNA damage response defect syndromes. Because lamin A dysregulation leads to DNA damages, it has been proposed that senescence occurs in both types of syndromes through the accumulation of damages. We recently showed that elevated oxidative stress is responsible for lamin B1 accumulation, nuclear shape alteration and senescence in the DDR syndrome, ataxia telangiectasia (A-T). Interestingly, overexpression of lamin B1 in wild type cells is sufficient to induce senescence without the induction of DNA damages. Here, we will discuss the importance of controlling the lamins level in order for maintenance nuclear architecture and we will comment the relationships of lamins with other senescence mechanisms. Finally, we will describe emerging data reporting redox control by lamins, leading us to propose a general mechanism by which reactive oxygen species can induce senescence through lamin dysregulation and NSA.
Collapse
Affiliation(s)
| | - Catherine Le Chalony
- CEA, DSV; Institut de Radiobiologie Cellulaire et Moléculaire; Laboratoire Réparation et Vieillissement; Fontenay-aux-Roses, France
| | - Gaëlle Pennarun
- CEA, DSV; Institut de Radiobiologie Cellulaire et Moléculaire; Laboratoire Réparation et Vieillissement; Fontenay-aux-Roses, France
| | - Diane Genet
- CEA, DSV; Institut de Radiobiologie Cellulaire et Moléculaire; Laboratoire Réparation et Vieillissement; Fontenay-aux-Roses, France
| | - Nancy Zaarour
- CEA, DSV; Institut de Radiobiologie Cellulaire et Moléculaire; Laboratoire Réparation et Vieillissement; Fontenay-aux-Roses, France
| | - Pascale Bertrand
- CEA, DSV; Institut de Radiobiologie Cellulaire et Moléculaire; Laboratoire Réparation et Vieillissement; Fontenay-aux-Roses, France
| |
Collapse
|
233
|
Novelli G, D'Apice MR. Protein farnesylation and disease. J Inherit Metab Dis 2012; 35:917-26. [PMID: 22307208 DOI: 10.1007/s10545-011-9445-y] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2011] [Revised: 12/16/2011] [Accepted: 12/22/2011] [Indexed: 12/19/2022]
Abstract
Prenylation consists of the addition of an isoprenoid group to a cysteine residue located near the carboxyl terminal of a protein. This enzymatic posttranslational modification is important for the maturation and processing of proteins. Both processes are necessary to mediate protein-protein and membrane-protein associations, in addition to regulating the localisation and function of proteins. The severe phenotype of animals deficient in enzymes involved in both prenylation and maturation highlights the significance of these processes. Moreover, alterations in the genes coding for isoprenylated proteins or enzymes that are involved in both prenylation and maturation processes have been found to be the basis of severe human diseases, such as cancer, neurodegenerative disorders, retinitis pigmentosa, and premature ageing syndromes. Recent studies on isoprenylation and postprenylation processing in pathological conditions have unveiled surprising aspects of these modifications and their roles in different cellular pathways. The identification of these enzymes as therapeutic targets has led researchers to validate their effects in vitro and in vivo as antitumour or antiageing agents. This review attempts to summarise the basic aspects of protein isoprenylation and postprenylation, integrating our data with that observed in other studies to provide a comprehensive scenario of progeroid syndromes and the therapeutic avenues.
Collapse
Affiliation(s)
- Giuseppe Novelli
- Department of Biopathology and Diagnostic Imaging, University of Tor Vergata, Via Montpellier 1, 00133 Rome, Italy.
| | | |
Collapse
|
234
|
Houben F, De Vos WH, Krapels IPC, Coorens M, Kierkels GJJ, Kamps MAF, Verstraeten VLRM, Marcelis CLM, van den Wijngaard A, Ramaekers FCS, Broers JLV. Cytoplasmic localization of PML particles in laminopathies. Histochem Cell Biol 2012; 139:119-34. [PMID: 22918509 DOI: 10.1007/s00418-012-1005-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/23/2012] [Indexed: 01/01/2023]
Abstract
There is growing evidence that laminopathies, diseases associated with mutations in the LMNA gene, are caused by a combination of mechanical and gene regulatory distortions. Strikingly, there is a large variability in disease symptoms between individual patients carrying an identical LMNA mutation. This is why classical genetic screens for mutations appear to have limited predictive value for disease development. Recently, the widespread occurrence of repetitive nuclear ruptures has been described in fibroblast cultures from various laminopathy patients. Since this phenomenon was strongly correlated with disease severity, the identification of biomarkers that report on these rupture events could have diagnostic relevance. One such candidate marker is the PML nuclear body, a structure that is normally confined to the nuclear interior, but leaks out of the nucleus upon nuclear rupture. Here, we show that a variety of laminopathies shows the presence of these cytoplasmic PML particles (PML CPs), and that the amount of these protein aggregates increases with severity of the disease. In addition, between clinically healthy individuals, carrying LMNA mutations, significant differences can be found. Therefore, we postulate that detection of PML CPs in patient fibroblasts could become a valuable marker for diagnosis of disease development.
Collapse
Affiliation(s)
- F Houben
- Department of Molecular Cell Biology, CARIM, School for Cardiovascular Diseases, Maastricht University Medical Center, UNS50 Box 17, P.O. Box 616, NL-6200 MD, Maastricht, The Netherlands
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
235
|
Byrne RD. The nuclear membrane as a lipid 'sink'-linking cell cycle progression to lipid synthesis. J Chem Biol 2012; 5:141-2. [PMID: 23943675 DOI: 10.1007/s12154-012-0082-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2012] [Accepted: 07/30/2012] [Indexed: 01/28/2023] Open
Affiliation(s)
- Richard D Byrne
- Cell Biophysics Laboratory, Cancer Research UK, London Research Institute, 44 Lincoln's Inn Fields, London, WC2A 3LY UK
| |
Collapse
|
236
|
Coutance G, Labombarda F, Cauderlier E, Belin A, Richard P, Bonne G, Chapon F. Hypoplasia of the aorta in a patient diagnosed with LMNA gene mutation. CONGENIT HEART DIS 2012; 8:E127-9. [PMID: 22883396 DOI: 10.1111/j.1747-0803.2012.00695.x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/30/2012] [Indexed: 12/01/2022]
Abstract
Hypoplasia of the aorta is a rare entity comprising tubular hypotrophy of a large segment of the thoracic and the abdominal aorta. We report for the first time the case of a 26-year-old man with Emery-Dreifuss muscular dystrophy presenting severe and diffuse hypoplasia of the aorta.
Collapse
Affiliation(s)
- Guillaume Coutance
- Department of Cardiology, University Teaching Hospital of Caen, Caen, France
| | | | | | | | | | | | | |
Collapse
|
237
|
Candelario J, Chen LY, Marjoram P, Reddy S, Comai L. A filtering strategy identifies FOXQ1 as a potential effector of lamin A dysfunction. Aging (Albany NY) 2012; 4:567-77. [PMID: 22948034 PMCID: PMC3461344 DOI: 10.18632/aging.100483] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2012] [Accepted: 08/29/2012] [Indexed: 11/25/2022]
Abstract
Small increases in the expression of wild-type prelamin A are sufficient to recapitulate the reduced cell proliferation and altered nuclear membrane morphology observed in cells expressing progerin, the mutant lamin A associated with progeria. We hypothesized that the manifestation of these phenotypes in cells expressing elevated levels of wild-type prelamin A or progerin is caused by the same molecular effectors, which play a central role in the onset of the progeroid phenotype. To experimentally test this hypothesis, we compared the transcriptomes of isogenic diploid fibroblasts expressing progerin or elevated levels of wild-type prelamin A with that of wild-type fibroblasts. We subsequently used the reversion towards normal of two phenotypes, reduced cell growth and dismorphic nuclei, by treatment with farnesyltransferase inhibitor (FTI) or overexpression of ZMPSTE24, as a filtering strategy to identify genes linked to the onset of these two phenotypes. Through this analysis we identified the gene encoding for the transcription factor FOXQ1, as a gene whose expression is induced in both cells expressing progerin and elevated levels of wild-type prelamin A, and subsequently reduced in both cell types upon conditions that ameliorate the phenotypes. We overexpressed FOXQ1 in normal fibroblasts and demonstrated that increased levels of this factor lead to the development of both features that were used in the filtering strategy. These findings suggest a potential link between this transcription factor and cell dysfunction induced by altered prelamin A metabolism.
Collapse
Affiliation(s)
- Jose Candelario
- Department of Molecular Microbiology & Immunology, University of Southern California, Los Angeles, CA 90033 USA
- Institute for Genetic Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033 USA
- La Jolla Bioengineering Institute, San Diego, CA 9212
| | - Leng-Ying Chen
- Department of Molecular Microbiology & Immunology, University of Southern California, Los Angeles, CA 90033 USA
- Institute for Genetic Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033 USA
| | - Paul Marjoram
- Department of Preventive Medicine, University of Southern California, Los Angeles, CA 90033 USA
| | - Sita Reddy
- Department of Molecular Biology & Biochemistry, University of Southern California, Los Angeles, CA 90033 USA
- Institute for Genetic Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033 USA
| | - Lucio Comai
- Department of Molecular Microbiology & Immunology, University of Southern California, Los Angeles, CA 90033 USA
- Institute for Genetic Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033 USA
| |
Collapse
|
238
|
Pathological features in the LmnaDhe/+ mutant mouse provide a novel model of human otitis media and laminopathies. THE AMERICAN JOURNAL OF PATHOLOGY 2012; 181:761-74. [PMID: 22819531 DOI: 10.1016/j.ajpath.2012.05.031] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/29/2012] [Revised: 05/07/2012] [Accepted: 05/17/2012] [Indexed: 11/21/2022]
Abstract
Genetic predisposition is recognized as an important pathogenetic factor in otitis media (OM) and associated diseases. Mutant Lmna mice heterozygous for the disheveled hair and ears allele (Lmna(Dhe/+)) exhibit early-onset, profound hearing deficits and other pathological features mimicking human laminopathy associated with the LMNA mutation. We assessed the effects of the Lmna(Dhe/+) mutation on development of OM and pathological abnormalities characteristic of laminopathy. Malformation and abnormal positioning of the eustachian tube, accompanied by OM, were observed in all of the Lmna(Dhe/+) mice (100% penetrance) as early as postnatal day P12. Scanning electronic microscopy revealed ultrastructural damage to the cilia in middle ears that exhibited OM. Hearing assessment revealed significant hearing loss, paralleling that in human OM. Expression of NF-κB, TNF-α, and TGF-β, which correlated with inflammation and/or bony development, was up-regulated in the ears or in the peritoneal macrophages of Lmna(Dhe/+) mice. Rugous, disintegrative, and enlarged nuclear morphology of peritoneal macrophages and hyperphosphatemia were found in Lmna(Dhe/+) mutant mice. Taken together, these features resemble the pathology of human laminopathies, possibly revealing some profound pathology, beyond OM, associated with the mutation. The Lmna(Dhe/+) mutant mouse provides a novel model of human OM and laminopathy.
Collapse
|
239
|
Lanzuolo C. Epigenetic alterations in muscular disorders. Comp Funct Genomics 2012; 2012:256892. [PMID: 22761545 PMCID: PMC3385594 DOI: 10.1155/2012/256892] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2012] [Revised: 04/11/2012] [Accepted: 04/19/2012] [Indexed: 11/18/2022] Open
Abstract
Epigenetic mechanisms, acting via chromatin organization, fix in time and space different transcriptional programs and contribute to the quality, stability, and heritability of cell-specific transcription programs. In the last years, great advances have been made in our understanding of mechanisms by which this occurs in normal subjects. However, only a small part of the complete picture has been revealed. Abnormal gene expression patterns are often implicated in the development of different diseases, and thus epigenetic studies from patients promise to fill an important lack of knowledge, deciphering aberrant molecular mechanisms at the basis of pathogenesis and diseases progression. The identification of epigenetic modifications that could be used as targets for therapeutic interventions could be particularly timely in the light of pharmacologically reversion of pathological perturbations, avoiding changes in DNA sequences. Here I discuss the available information on epigenetic mechanisms that, altered in neuromuscular disorders, could contribute to the progression of the disease.
Collapse
Affiliation(s)
- Chiara Lanzuolo
- CNR Institute of Cellular Biology and Neurobiology, IRCCS Santa Lucia Foundation, Via Del Fosso di Fiorano 64, 00143 Rome, Italy
| |
Collapse
|
240
|
Stallmeyer B, Koopmann M, Schulze-Bahr E. Identification of Novel Mutations in LMNA Associated with Familial Forms of Dilated Cardiomyopathy. Genet Test Mol Biomarkers 2012; 16:543-9. [DOI: 10.1089/gtmb.2011.0214] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Affiliation(s)
- Birgit Stallmeyer
- Institut für Genetik von Herzerkrankungen (IfGH), Universitätsklinik Münster, Münster, Germany
- Interdisziplinäres Zentrum für klinische Forschung (IZKF), Münster, Germany
| | - Matthias Koopmann
- Department für Kardiology und Angiology, Universitätsklinik Münster, Münster, Germany
| | - Eric Schulze-Bahr
- Institut für Genetik von Herzerkrankungen (IfGH), Universitätsklinik Münster, Münster, Germany
- Interdisziplinäres Zentrum für klinische Forschung (IZKF), Münster, Germany
- Department für Kardiology und Angiology, Universitätsklinik Münster, Münster, Germany
| |
Collapse
|
241
|
Araújo-Vilar D, Victoria B, González-Méndez B, Barreiro F, Fernández-Rodríguez B, Cereijo R, Gallego-Escuredo JM, Villarroya F, Pañeda-Menéndez A. Histological and molecular features of lipomatous and nonlipomatous adipose tissue in familial partial lipodystrophy caused by LMNA mutations. Clin Endocrinol (Oxf) 2012; 76:816-24. [PMID: 21883346 DOI: 10.1111/j.1365-2265.2011.04208.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
OBJECTIVES Type 2 familial partial lipodystrophy (FPLD2) is a rare adipose tissue (AT) disease caused by mutations in LMNA, in which lipomas appear occasionally. In this study, we aimed to histologically characterize FPLD2-associated lipomatosis and study the expression of genes and proteins involved in cell cycle control, mitochondrial function, inflammation and adipogenesis. DESIGN AND PATIENTS One lipoma and perilipoma fat from each of four subjects with FPLD2 and 10 control subjects were analysed by optical microscopy. The presence of inflammatory cells was evaluated by immunohistochemistry. Real-time RT-PCR and Western blot were used to evaluate gene and protein levels. RESULTS Adipocytes from lipodystrophic patients were significantly larger than those of controls, in both the lipomas and perilipoma fat. Lipodystrophic AT exhibited CD68(+) macrophages and CD3(+) lymphocytes infiltration. TP53 expression was reduced in all types of lipomas. At protein level, C/EBPβ, p53 and pRb were severely disturbed in both lipodystrophic lipomas and perilipoma fat coming from lipoatrophic areas, whereas the expression of CEBPα was normal. Mitochondrial function genes were less expressed in lipoatrophic fat. In both lipomas and perilipoma fat from lipoatrophic areas, the expression of adipogenes was lower than controls. CONCLUSIONS Even in lipomas, the adipogenic machinery is impaired in lipodystrophic fat coming from lipoatrophic regions in FPLD2, although the histological phenotype is near-normal, exhibiting low-grade inflammatory features. Our results suggest that the p53 pathway and some adipogenic proteins, such as CEBPα, could contribute to the maintenance of this near normal phenotype in the remnant AT present in these patients.
Collapse
Affiliation(s)
- D Araújo-Vilar
- Thyroid and Metabolic Diseases Unit (U.E.T.eM.), Department of Medicine, University of Santiago de Compostela, Spain.
| | | | | | | | | | | | | | | | | |
Collapse
|
242
|
Kubben N, Adriaens M, Meuleman W, Voncken JW, van Steensel B, Misteli T. Mapping of lamin A- and progerin-interacting genome regions. Chromosoma 2012; 121:447-64. [PMID: 22610065 PMCID: PMC3443488 DOI: 10.1007/s00412-012-0376-7] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2012] [Revised: 04/23/2012] [Accepted: 04/26/2012] [Indexed: 12/15/2022]
Abstract
Mutations in the A-type lamins A and C, two major components of the nuclear lamina, cause a large group of phenotypically diverse diseases collectively referred to as laminopathies. These conditions often involve defects in chromatin organization. However, it is unclear whether A-type lamins interact with chromatin in vivo and whether aberrant chromatin–lamin interactions contribute to disease. Here, we have used an unbiased approach to comparatively map genome-wide interactions of gene promoters with lamin A and progerin, the mutated lamin A isoform responsible for the premature aging disorder Hutchinson–Gilford progeria syndrome (HGPS) in mouse cardiac myoytes and embryonic fibroblasts. We find that lamin A-associated genes are predominantly transcriptionally silent and that loss of lamin association leads to the relocation of peripherally localized genes, but not necessarily to their activation. We demonstrate that progerin induces global changes in chromatin organization by enhancing interactions with a specific subset of genes in addition to the identified lamin A-associated genes. These observations demonstrate disease-related changes in higher order genome organization in HGPS and provide novel insights into the role of lamin–chromatin interactions in chromatin organization.
Collapse
Affiliation(s)
- Nard Kubben
- Genome Cell Biology Group, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | | | | | |
Collapse
|
243
|
Gentil BJ, Cooper L. Molecular basis of axonal dysfunction and traffic impairments in CMT. Brain Res Bull 2012; 88:444-53. [PMID: 22595495 DOI: 10.1016/j.brainresbull.2012.05.003] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2011] [Revised: 05/01/2012] [Accepted: 05/04/2012] [Indexed: 12/17/2022]
Abstract
Charcot-Marie-Tooth disease (CMT) is one of the most common inherited neurological disorders. It comprises a group of diseases caused by mutations in genes involved in Schwann cells homeostasis and neuronal function that affect the peripheral nerves. So far mutations in more than 33 genes have been identified causing either the demyelinating form (CMT1) or the axonal form (CMT2). Genes involving a large variety of unrelated functions may lead to the same phenotype when mutated. Our review will focus on the common link between genes causing axonal phenotypes like MFN2, KIF1B, DYNC1H1, Rab7, TRPV4, ARSs, NEFL, HSPB1, MPZ, and HSPB8. While KIF1B and DYNC1H1, two genes coding for molecular motors, are directly linked to axonal transport, the involvement of the other CMT2-causing genes in this function is less obvious. However, the last years have seen a growing list of evidence demonstrating that intracellular trafficking and mitochondrial dynamics might be dysfunctional in CMT2, and these mechanisms might present a common link between dissimilar CMT2-causing genes. The involvement of impaired transport in the pathogenesis of other rare neurological diseases or recessive CMT2 is also discussed.
Collapse
Affiliation(s)
- Benoit J Gentil
- Department of Neurology/Neurosurgery and Montreal Neurological Institute, McGill University, Montreal, QC H3A 2B4 Canada.
| | | |
Collapse
|
244
|
Young SG, Jung HJ, Coffinier C, Fong LG. Understanding the roles of nuclear A- and B-type lamins in brain development. J Biol Chem 2012; 287:16103-10. [PMID: 22416132 PMCID: PMC3351360 DOI: 10.1074/jbc.r112.354407] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The nuclear lamina is composed mainly of lamins A and C (A-type lamins) and lamins B1 and B2 (B-type lamins). Dogma has held that lamins B1 and B2 play unique and essential roles in the nucleus of every eukaryotic cell. Recent studies have raised doubts about that view but have uncovered crucial roles for lamins B1 and B2 in neuronal migration during the development of the brain. The relevance of lamins A and C in the brain remains unclear, but it is intriguing that prelamin A expression in the brain is low and is regulated by miR-9, a brain-specific microRNA.
Collapse
Affiliation(s)
| | - Hea-Jin Jung
- the Molecular Biology Institute, UCLA, Los Angeles, California 90095
| | | | | |
Collapse
|
245
|
Righolt CH, van 't Hoff MLR, Vermolen BJ, Young IT, Raz V. Robust nuclear lamina-based cell classification of aging and senescent cells. Aging (Albany NY) 2012; 3:1192-201. [PMID: 22199022 PMCID: PMC3273899 DOI: 10.18632/aging.100414] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Changes in the shape of the nuclear lamina are exhibited in senescent cells, as well as in cells expressing mutations in lamina genes. To identify cells with defects in the nuclear lamina we developed an imaging method that quantifies the intensity and curvature of the nuclear lamina. We show that this method accurately describes changes in the nuclear lamina. Spatial changes in nuclear lamina coincide with redistribution of lamin A proteins and local reduction in protein mobility in senescent cell. We suggest that local accumulation of lamin A in the nuclear envelope leads to bending of the structure. A quantitative distinction of the nuclear lamina shape in cell populations was found between fresh and senescent cells, and between primary myoblasts from young and old donors. Moreover, with this method mutations in lamina genes were significantly distinct from cells with wild-type genes. We suggest that this method can be applied to identify abnormal cells during aging, in in vitro propagation, and in lamina disorders.
Collapse
|
246
|
A novel homozygous p.Arg527Leu LMNA mutation in two unrelated Egyptian families causes overlapping mandibuloacral dysplasia and progeria syndrome. Eur J Hum Genet 2012; 20:1134-40. [PMID: 22549407 DOI: 10.1038/ejhg.2012.77] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Mandibuloacral dysplasia (MAD) is a rare disease resulting from a mutation of LMNA gene encoding lamins A and C. The most common mutation associated with this disease is a homozygous arginine 527 replacement by histidine. Three female patients originating from two unrelated families from Northeast Egypt were examined. Their growth was retarded; they had microcephaly, widened cranial sutures, prominent eyes and cheeks, micrognathia, dental crowding, hypoplastic mandible, acro-osteolysis of distal phalanges, and joint contractures. In addition, they presented some progeroid features, such as pinched nose, premature loss of teeth, loss of hair, scleroderma-like skin atrophy, spine rigidity, and waddling gait. The clinical presentation of the disease varied between the patient originating from Family 1 and patients from Family 2, suggesting that unknown, possibly epigenetic factors, modify the course of the disease. The first symptoms of the disease appeared at the age of 2.5 (a girl from Family 1), 5, and 3 years (girls from Family 2). All patients had the same, novel homozygous c.1580G>T LMNA mutation, resulting in the replacement of arginine 527 by leucine. Computational predictions of such substitution effects suggested that it might alter protein stability and increase the tendency for protein aggregation, and as a result, might influence its interaction with other proteins. In addition, restriction fragment-length polymorphism analysis performed in 178 unrelated individuals showed that up to 1.12% of inhabitants of Northeast Egypt might be heterozygous carriers of this mutation, suggesting the presence of a founder effect in this area.
Collapse
|
247
|
Lindeman RE, Pelegri F. Localized products of futile cycle/lrmp promote centrosome-nucleus attachment in the zebrafish zygote. Curr Biol 2012; 22:843-51. [PMID: 22542100 DOI: 10.1016/j.cub.2012.03.058] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2011] [Revised: 02/16/2012] [Accepted: 03/12/2012] [Indexed: 10/28/2022]
Abstract
BACKGROUND The centrosome has a well-established role as a microtubule organizer during mitosis and cytokinesis. In addition, it facilitates the union of parental haploid genomes following fertilization by nucleating a microtubule aster along which the female pronucleus migrates toward the male pronucleus. Stable associations between the sperm aster and the pronuclei are essential during this directed movement. RESULTS Our studies reveal that the zebrafish gene futile cycle (fue) is required in the zygote for male pronucleus-centrosome attachment and female pronuclear migration. We show that fue encodes a novel, maternally-provided long form of lymphoid-restricted membrane protein (lrmp), a vertebrate-specific gene of unknown function. Both maternal lrmp messenger RNA (mRNA) and protein are highly localized in the zygote, in a largely overlapping pattern at nuclear membranes, centrosomes, and spindles. Truncated Lrmp-EGFP fusion proteins identified subcellular targeting signals in the C terminus of Lrmp; however, endogenous mRNA localization is likely important to ensure strict spatial expression of the protein. Localization of both Lrmp protein and lrmp RNA is defective in fue mutant embryos, indicating that correct targeting of lrmp gene products is dependent on Lrmp function. CONCLUSIONS Lrmp is a conserved vertebrate gene whose maternally inherited products are essential for nucleus-centrosome attachment and pronuclear congression during fertilization. Precise subcellular localization of lrmp products also suggests a requirement for strict spatiotemporal regulation of their function in the early embryo.
Collapse
Affiliation(s)
- Robin E Lindeman
- Laboratory of Genetics, University of Wisconsin-Madison, Madison, WI 53706, USA
| | | |
Collapse
|
248
|
Sieprath T, Darwiche R, De Vos WH. Lamins as mediators of oxidative stress. Biochem Biophys Res Commun 2012; 421:635-9. [PMID: 22538370 DOI: 10.1016/j.bbrc.2012.04.058] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2012] [Accepted: 04/10/2012] [Indexed: 01/29/2023]
Abstract
The nuclear lamina defines both structural and functional properties of the eukaryotic cell nucleus. Mutations in the LMNA gene, encoding A-type lamins, lead to a broad spectrum of diseases termed laminopathies. While different hypotheses have been postulated to explain disease development, there is still no unified view on the mechanistic basis of laminopathies. Recent observations indicate that laminopathies are often accompanied by altered levels of reactive oxygen species and a higher susceptibility to oxidative stress at the cellular level. In this review, we highlight the role of reactive oxygen species for cell function and disease development in the context of laminopathies and present a framework of non-exclusive mechanisms to explain the reciprocal interactions between a dysfunctional lamina and altered redox homeostasis.
Collapse
Affiliation(s)
- Tom Sieprath
- Cell Systems and Cellular Imaging Group, Department of Molecular Biotechnology, Ghent University, Ghent, Belgium
| | | | | |
Collapse
|
249
|
Kubben N, Voncken JW, Konings G, van Weeghel M, van den Hoogenhof MM, Gijbels M, van Erk A, Schoonderwoerd K, van den Bosch B, Dahlmans V, Calis C, Houten SM, Misteli T, Pinto YM. Post-natal myogenic and adipogenic developmental: defects and metabolic impairment upon loss of A-type lamins. Nucleus 2012; 2:195-207. [PMID: 21818413 DOI: 10.4161/nucl.2.3.15731] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2011] [Revised: 04/01/2011] [Accepted: 04/05/2011] [Indexed: 12/23/2022] Open
Abstract
A-type lamins are a major component of the nuclear lamina. Mutations in the LMNA gene, which encodes the A-type lamins A and C, cause a set of phenotypically diverse diseases collectively called laminopathies. While adult LMNA null mice show various symptoms typically associated with laminopathies, the effect of loss of lamin A/C on early post-natal development is poorly understood. Here we developed a novel LMNA null mouse (LMNA(GT-/-)) based on genetrap technology and analyzed its early post-natal development. We detect LMNA transcripts in heart, the outflow tract, dorsal aorta, liver and somites during early embryonic development. Loss of A-type lamins results in severe growth retardation and developmental defects of the heart, including impaired myocyte hypertrophy, skeletal muscle hypotrophy, decreased amounts of subcutaneous adipose tissue and impaired ex vivo adipogenic differentiation. These defects cause death at 2 to 3 weeks post partum associated with muscle weakness and metabolic complications, but without the occurrence of dilated cardiomyopathy or an obvious progeroid phenotype. Our results indicate that defective early post-natal development critically contributes to the disease phenotypes in adult laminopathies.
Collapse
Affiliation(s)
- Nard Kubben
- Heart Failure Research Center and Department of Cardiology, Maastricht University Medical Centre, The Netherlands
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
250
|
Ruiz de Eguino G, Infante A, Schlangen K, Aransay AM, Fullaondo A, Soriano M, García-Verdugo JM, Martín AG, Rodríguez CI. Sp1 transcription factor interaction with accumulated prelamin a impairs adipose lineage differentiation in human mesenchymal stem cells: essential role of sp1 in the integrity of lipid vesicles. Stem Cells Transl Med 2012. [PMID: 23197810 DOI: 10.5966/sctm.2011-0010] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Lamin A (LMNA)-linked lipodystrophies may be either genetic (associated with LMNA mutations) or acquired (associated with the use of human immunodeficiency virus protease inhibitors [PIs]), and in both cases they share clinical features such as anomalous distribution of body fat or generalized loss of adipose tissue, metabolic alterations, and early cardiovascular complications. Both LMNA-linked lipodystrophies are characterized by the accumulation of the lamin A precursor prelamin A. The pathological mechanism by which prelamin A accumulation induces the lipodystrophy associated phenotypes remains unclear. Since the affected tissues in these disorders are of mesenchymal origin, we have generated an LMNA-linked experimental model using human mesenchymal stem cells treated with a PI, which recapitulates the phenotypes observed in patient biopsies. This model has been demonstrated to be a useful tool to unravel the pathological mechanism of the LMNA-linked lipodystrophies, providing an ideal system to identify potential targets to generate new therapies for drug discovery screening. We report for the first time that impaired adipogenesis is a consequence of the interaction between accumulated prelamin A and Sp1 transcription factor, sequestration of which results in altered extracellular matrix gene expression. In fact, our study shows a novel, essential, and finely tuned role for Sp1 in adipose lineage differentiation in human mesenchymal stem cells. These findings define a new physiological experimental model to elucidate the pathological mechanisms LMNA-linked lipodystrophies, creating new opportunities for research and treatment not only of LMNA-linked lipodystrophies but also of other adipogenesis-associated metabolic diseases.
Collapse
Affiliation(s)
- Garbiñe Ruiz de Eguino
- Stem Cells and Cell Therapy Laboratory, BioCruces, Hospital Universitario Cruces, Barakaldo, Spain
| | | | | | | | | | | | | | | | | |
Collapse
|