51
|
Kuga T, Nie H, Kazami T, Satoh M, Matsushita K, Nomura F, Maeshima K, Nakayama Y, Tomonaga T. Lamin B2 prevents chromosome instability by ensuring proper mitotic chromosome segregation. Oncogenesis 2014; 3:e94. [PMID: 24637494 PMCID: PMC4038388 DOI: 10.1038/oncsis.2014.6] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2013] [Revised: 12/27/2013] [Accepted: 01/27/2014] [Indexed: 12/20/2022] Open
Abstract
The majority of human cancer shows chromosomal instability (CIN). Although the precise mechanism remains largely uncertain, proper progression of mitosis is crucial. B-type lamins were suggested to be components of the spindle matrix of mitotic cells and to be involved in mitotic spindle assembly; thus, B-type lamins may contribute to the maintenance of chromosome integrity. Here, using a proteomic approach, we identified lamin B2 as a novel protein involved in CIN. Lamin B2 expression decreased in colorectal cancer cell lines exhibiting CIN, as compared with colorectal cancer cell lines exhibiting microsatellite instability (MIN), which is mutually exclusive to CIN. Importantly, lamin B2 knockdown in MIN-type colorectal cancer cells induced CIN phenotypes such as aneuploidy, chromosome mis-segregation and aberrant spindle assembly, whereas ectopic expression of lamin B2 in CIN-type colorectal cancer cells prevented their CIN phenotypes. Additionally, immunohistochemical analysis showed a lower expression of lamin B2 in cancer tissues extracted from patients with sporadic colorectal cancer (CIN-type) than that from patients with hereditary non-polyposis colorectal cancer (HNPCC; MIN type). Intriguingly, mitotic lamin B2 in MIN cancer cells was localized outside the spindle poles and mitotic lamin B2 localization was diminished in CIN cancer cells, suggesting an important role of lamin B2 in proper mitotic spindle formation. The obtained results suggest that lamin B2 maintains chromosome integrity by ensuring proper spindle assembly and that its downregulation causes CIN in colorectal cancer.
Collapse
Affiliation(s)
- T Kuga
- 1] Laboratory of Proteome Research, National Institute of Biomedical Innovation, Osaka, Japan [2] Department of Molecular Diagnosis (F8), Graduate School of Medicine, Chiba University, Chiba, Japan [3] Department of Biochemistry and Molecular Biology, Kyoto Pharmaceutical University, Kyoto, Japan
| | - H Nie
- Department of Molecular Diagnosis (F8), Graduate School of Medicine, Chiba University, Chiba, Japan
| | - T Kazami
- Department of Molecular Diagnosis (F8), Graduate School of Medicine, Chiba University, Chiba, Japan
| | - M Satoh
- Department of Molecular Diagnosis (F8), Graduate School of Medicine, Chiba University, Chiba, Japan
| | - K Matsushita
- Department of Molecular Diagnosis (F8), Graduate School of Medicine, Chiba University, Chiba, Japan
| | - F Nomura
- Department of Molecular Diagnosis (F8), Graduate School of Medicine, Chiba University, Chiba, Japan
| | - K Maeshima
- Biological Macromolecules Laboratory, Structural Biology Center, National Institute of Genetics, Shizuoka, Japan
| | - Y Nakayama
- Department of Biochemistry and Molecular Biology, Kyoto Pharmaceutical University, Kyoto, Japan
| | - T Tomonaga
- 1] Laboratory of Proteome Research, National Institute of Biomedical Innovation, Osaka, Japan [2] Department of Molecular Diagnosis (F8), Graduate School of Medicine, Chiba University, Chiba, Japan
| |
Collapse
|
52
|
RB and Lamins in Cell Cycle Regulation and Aging. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2014; 773:127-42. [DOI: 10.1007/978-1-4899-8032-8_6] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
53
|
Hatch EM, Fischer AH, Deerinck TJ, Hetzer MW. Catastrophic nuclear envelope collapse in cancer cell micronuclei. Cell 2013; 154:47-60. [PMID: 23827674 DOI: 10.1016/j.cell.2013.06.007] [Citation(s) in RCA: 550] [Impact Index Per Article: 45.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2013] [Revised: 04/24/2013] [Accepted: 06/07/2013] [Indexed: 10/26/2022]
Abstract
During mitotic exit, missegregated chromosomes can recruit their own nuclear envelope (NE) to form micronuclei (MN). MN have reduced functioning compared to primary nuclei in the same cell, although the two compartments appear to be structurally comparable. Here we show that over 60% of MN undergo an irreversible loss of compartmentalization during interphase due to NE collapse. This disruption of the MN, which is induced by defects in nuclear lamina assembly, drastically reduces nuclear functions and can trigger massive DNA damage. MN disruption is associated with chromatin compaction and invasion of endoplasmic reticulum (ER) tubules into the chromatin. We identified disrupted MN in both major subtypes of human non-small-cell lung cancer, suggesting that disrupted MN could be a useful objective biomarker for genomic instability in solid tumors. Our study shows that NE collapse is a key event underlying MN dysfunction and establishes a link between aberrant NE organization and aneuploidy.
Collapse
Affiliation(s)
- Emily M Hatch
- Molecular and Cell Biology Laboratory, Salk Institute for Biological Studies, 10010 N. Torrey Pines Road, La Jolla, CA 92037, USA
| | | | | | | |
Collapse
|
54
|
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: 2.0] [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
|
55
|
Zwerger M, Jaalouk DE, Lombardi ML, Isermann P, Mauermann M, Dialynas G, Herrmann H, Wallrath LL, Lammerding J. Myopathic lamin mutations impair nuclear stability in cells and tissue and disrupt nucleo-cytoskeletal coupling. Hum Mol Genet 2013; 22:2335-49. [PMID: 23427149 DOI: 10.1093/hmg/ddt079] [Citation(s) in RCA: 137] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Lamins are intermediate filament proteins that assemble into a meshwork underneath the inner nuclear membrane, the nuclear lamina. Mutations in the LMNA gene, encoding lamins A and C, cause a variety of diseases collectively called laminopathies. The disease mechanism for these diverse conditions is not well understood. Since lamins A and C are fundamental determinants of nuclear structure and stability, we tested whether defects in nuclear mechanics could contribute to the disease development, especially in laminopathies affecting mechanically stressed tissue such as muscle. Using skin fibroblasts from laminopathy patients and lamin A/C-deficient mouse embryonic fibroblasts stably expressing a broad panel of laminopathic lamin A mutations, we found that several mutations associated with muscular dystrophy and dilated cardiomyopathy resulted in more deformable nuclei; in contrast, lamin mutants responsible for diseases without muscular phenotypes did not alter nuclear deformability. We confirmed our results in intact muscle tissue, demonstrating that nuclei of transgenic Drosophila melanogaster muscle expressing myopathic lamin mutations deformed more under applied strain than controls. In vivo and in vitro studies indicated that the loss of nuclear stiffness resulted from impaired assembly of mutant lamins into the nuclear lamina. Although only a subset of lamin mutations associated with muscular diseases caused increased nuclear deformability, almost all mutations tested had defects in force transmission between the nucleus and cytoskeleton. In conclusion, our results indicate that although defective nuclear stability may play a role in the development of muscle diseases, other factors, such as impaired nucleo-cytoskeletal coupling, likely contribute to the muscle phenotype.
Collapse
Affiliation(s)
- Monika Zwerger
- Department of Medicine, Brigham and Women's Hospital/Harvard Medical School, Boston, MA 02115, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
56
|
Abstract
Budding yeast, like other eukaryotes, carries its genetic information on chromosomes that are sequestered from other cellular constituents by a double membrane, which forms the nucleus. An elaborate molecular machinery forms large pores that span the double membrane and regulate the traffic of macromolecules into and out of the nucleus. In multicellular eukaryotes, an intermediate filament meshwork formed of lamin proteins bridges from pore to pore and helps the nucleus reform after mitosis. Yeast, however, lacks lamins, and the nuclear envelope is not disrupted during yeast mitosis. The mitotic spindle nucleates from the nucleoplasmic face of the spindle pole body, which is embedded in the nuclear envelope. Surprisingly, the kinetochores remain attached to short microtubules throughout interphase, influencing the position of centromeres in the interphase nucleus, and telomeres are found clustered in foci at the nuclear periphery. In addition to this chromosomal organization, the yeast nucleus is functionally compartmentalized to allow efficient gene expression, repression, RNA processing, genomic replication, and repair. The formation of functional subcompartments is achieved in the nucleus without intranuclear membranes and depends instead on sequence elements, protein-protein interactions, specific anchorage sites at the nuclear envelope or at pores, and long-range contacts between specific chromosomal loci, such as telomeres. Here we review the spatial organization of the budding yeast nucleus, the proteins involved in forming nuclear subcompartments, and evidence suggesting that the spatial organization of the nucleus is important for nuclear function.
Collapse
|
57
|
Affiliation(s)
- Chin Yee Ho
- Cornell University, Weill Institute for Cell and Molecular Biology, Department of Biomedical Engineering, Ithaca, NY 14853, USA
| | | |
Collapse
|
58
|
Uchino R, Nonaka YK, Horigome T, Sugiyama S, Furukawa K. Loss of Drosophila A-type lamin C initially causes tendon abnormality including disintegration of cytoskeleton and nuclear lamina in muscular defects. Dev Biol 2012; 373:216-27. [PMID: 22982669 DOI: 10.1016/j.ydbio.2012.08.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2012] [Revised: 07/19/2012] [Accepted: 08/01/2012] [Indexed: 11/25/2022]
Abstract
Lamins are the major components of nuclear envelope architecture, being required for both the structural and informational roles of the nuclei. Mutations of lamins cause a spectrum of diseases in humans, including muscular dystrophy. We report here that the loss of the A-type lamin gene, lamin C in Drosophila resulted in pupal metamorphic lethality caused by tendon defects, matching the characteristics of human A-type lamin revealed by Emery-Dreifuss muscular dystrophy (EDMD). In tendon cells lacking lamin C activity, overall cell morphology was affected and organization of the spectraplakin family cytoskeletal protein Shortstop which is prominently expressed in tendon cells gradually disintegrated, notably around the nucleus and in a manner correlating well with the degradation of musculature. Furthermore, lamin C null mutants were efficiently rescued by restoring lamin C expression to shortstop-expressing cells, which include tendon cells but exclude skeletal muscle cells. Thus the critical function of A-type lamin C proteins in Drosophila musculature is to maintain proper function and morphology of tendon cells.
Collapse
Affiliation(s)
- Ryo Uchino
- Department of Chemistry, Faculty of Science, Niigata University, Niigata 950-2181, Japan
| | | | | | | | | |
Collapse
|
59
|
Frock RL, Chen SC, Da DF, Frett E, Lau C, Brown C, Pak DN, Wang Y, Muchir A, Worman HJ, Santana LF, Ladiges WC, Rabinovitch PS, Kennedy BK. Cardiomyocyte-specific expression of lamin a improves cardiac function in Lmna-/- mice. PLoS One 2012; 7:e42918. [PMID: 22905185 PMCID: PMC3419749 DOI: 10.1371/journal.pone.0042918] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2011] [Accepted: 07/16/2012] [Indexed: 11/18/2022] Open
Abstract
Lmna(-/-) mice display multiple tissue defects and die by 6-8 weeks of age reportedly from dilated cardiomyopathy with associated conduction defects. We sought to determine whether restoration of lamin A in cardiomyocytes improves cardiac function and extends the survival of Lmna(-/-) mice. We observed increased total desmin protein levels and disorganization of the cytoplasmic desmin network in ~20% of Lmna(-/-) ventricular myocytes, rescued in a cell-autonomous manner in Lmna(-/-) mice expressing a cardiac-specific lamin A transgene (Lmna(-/-); Tg). Lmna(-/-); Tg mice displayed significantly increased contractility and preservation of myocardial performance compared to Lmna(-/-) mice. Lmna(-/-); Tg mice attenuated ERK1/2 phosphorylation relative to Lmna(-/-) mice, potentially underlying the improved localization of connexin43 to the intercalated disc. Electrocardiographic recordings from Lmna(-/-) mice revealed arrhythmic events and increased frequency of PR interval prolongation, which is partially rescued in Lmna(-/-); Tg mice. These findings support our observation that Lmna(-/-); Tg mice have a 12% median extension in lifespan compared to Lmna(-/-) mice. While significant, Lmna(-/-); Tg mice only have modest improvement in cardiac function and survival likely stemming from the observation that only 40% of Lmna(-/-); Tg cardiomyocytes have detectable lamin A expression. Cardiomyocyte-specific restoration of lamin A in Lmna(-/-) mice improves heart-specific pathology and extends lifespan, demonstrating that the cardiac pathology of Lmna(-/-) mice limits survival. The expression of lamin A is sufficient to rescue certain cellular defects associated with loss of A-type lamins in cardiomyocytes in a cell-autonomous fashion.
Collapse
Affiliation(s)
- Richard L. Frock
- Department of Biochemistry, University of Washington, Seattle, Washington, United States of America
| | - Steven C. Chen
- Department of Biochemistry, University of Washington, Seattle, Washington, United States of America
| | - Dao-Fu Da
- Department of Pathology, University of Washington, Seattle, Washington, United States of America
| | - Ellie Frett
- Department of Pharmacology and Physiology, University of Rochester, Rochester, New York, United States of America
| | - Carmen Lau
- Department of Biochemistry, University of Washington, Seattle, Washington, United States of America
| | - Christina Brown
- Department of Biochemistry, University of Washington, Seattle, Washington, United States of America
| | - Diana N. Pak
- Department of Biochemistry, University of Washington, Seattle, Washington, United States of America
| | - Yuexia Wang
- Department of Medicine and Department of Pathology and Cell Biology, College of Physicians and Surgeons, Columbia University, New York, New York, United States of America
| | - Antoine Muchir
- Department of Medicine and Department of Pathology and Cell Biology, College of Physicians and Surgeons, Columbia University, New York, New York, United States of America
| | - Howard J. Worman
- Department of Medicine and Department of Pathology and Cell Biology, College of Physicians and Surgeons, Columbia University, New York, New York, United States of America
| | - Luis F. Santana
- Department of Physiology and Biophysics, University of Washington, Seattle, Washington, United States of America
| | - Warren C. Ladiges
- Department of Comparative Medicine, University of Washington, Seattle, Washington, United States of America
| | - Peter S. Rabinovitch
- Department of Pathology, University of Washington, Seattle, Washington, United States of America
| | - Brian K. Kennedy
- Department of Biochemistry, University of Washington, Seattle, Washington, United States of America
- Buck Institute for Age Research, Novato, California, United States of America
- * E-mail:
| |
Collapse
|
60
|
Deshmukh RS, Østrup O, Strejcek F, Vejlsted M, Lucas-Hahn A, Petersen B, Li J, Callesen H, Niemann H, Hyttel P. Early aberrations in chromatin dynamics in embryos produced under in vitro conditions. Cell Reprogram 2012; 14:225-34. [PMID: 22468997 DOI: 10.1089/cell.2011.0069] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
In vitro production of porcine embryos by means of in vitro fertilization (IVF) or somatic cell nuclear transfer (SCNT) is limited by great inefficienciy. The present study investigated chromatin and nucleolar dynamics in porcine embryos developed in vivo (IV) and compared this physiological standard to that of embryos produced by IVF, parthenogenetic activation (PA), or SCNT. In contrast to IV embryos, chromatin spatial and temporal dynamics in PA, IVF, and SCNT embryos were altered; starting with aberrant chromatin-nuclear envelope interactions at the two-cell stage, delayed chromatin decondensation and nucleolar development at the four-cell stage, and ultimately culminating in failure of proper first lineage segregation at the blastocyst stage, demonstrated by poorly defined inner cell mass. Interestingly, in vitro produced (IVP) embryos also lacked a heterochromatin halo around nucleolar precursors, indicating imperfections in global chromatin remodeling after fertilization/activation. Porcine IV-produced zygotes and embryos display a well-synchronized pattern of chromatin dynamics compatible with genome activation and regular nucleolar formation at the four-cell stage. Production of porcine embryos under in vitro conditions by IVF, PA, or SCNT is associated with altered chromatin remodeling, delayed nucleolar formation, and poorly defined lineage segregation at the blastocyst stage, which in turn may impair their developmental capacity.
Collapse
Affiliation(s)
- Rahul S Deshmukh
- Department of Basic Animal and Veterinary Sciences, Faculty of Life Sciences, University of Copenhagen, Denmark
| | | | | | | | | | | | | | | | | | | |
Collapse
|
61
|
de Las Heras JI, Batrakou DG, Schirmer EC. Cancer biology and the nuclear envelope: a convoluted relationship. Semin Cancer Biol 2012; 23:125-37. [PMID: 22311402 DOI: 10.1016/j.semcancer.2012.01.008] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2011] [Revised: 01/16/2012] [Accepted: 01/19/2012] [Indexed: 12/23/2022]
Abstract
Although its properties have long been used for both typing and prognosis of various tumors, the nuclear envelope (NE) itself and its potential roles in tumorigenesis are only beginning to be understood. Historically viewed as merely a protective barrier, the nuclear envelope is now linked to a wide range of functions. Nuclear membrane proteins connect the nucleus to the cytoskeleton on one side and to chromatin on the other. Several newly identified nuclear envelope functions associated with these connections intersect with cancer pathways. For example, the nuclear envelope could affect genome stability by tethering chromatin. Some nuclear envelope proteins affect cell cycle regulation by directly binding to the master regulator pRb, others by interacting with TGF-ß and Smad signaling cascades, and others by affecting the mitotic spindle. Finally, the NE directly affects cytoskeletal organization and can also influence cell migration in metastasis. In this review we discuss the link between the nuclear envelope and cellular defects that are common in cancer cells, and we show that NE proteins are often aberrantly expressed in tumors. The NE represents a potential reservoir of diagnostic and prognostic markers in cancer.
Collapse
Affiliation(s)
- Jose I de Las Heras
- Wellcome Trust Centre for Cell Biology, University of Edinburgh, Edinburgh, UK
| | | | | |
Collapse
|
62
|
Anderson DD, Woeller CF, Chiang EP, Shane B, Stover PJ. Serine hydroxymethyltransferase anchors de novo thymidylate synthesis pathway to nuclear lamina for DNA synthesis. J Biol Chem 2012; 287:7051-62. [PMID: 22235121 DOI: 10.1074/jbc.m111.333120] [Citation(s) in RCA: 103] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The de novo thymidylate biosynthetic pathway in mammalian cells translocates to the nucleus for DNA replication and repair and consists of the enzymes serine hydroxymethyltransferase 1 and 2α (SHMT1 and SHMT2α), thymidylate synthase, and dihydrofolate reductase. In this study, we demonstrate that this pathway forms a multienzyme complex that is associated with the nuclear lamina. SHMT1 or SHMT2α is required for co-localization of dihydrofolate reductase, SHMT, and thymidylate synthase to the nuclear lamina, indicating that SHMT serves as scaffold protein that is essential for complex formation. The metabolic complex is enriched at sites of DNA replication initiation and associated with proliferating cell nuclear antigen and other components of the DNA replication machinery. These data provide a mechanism for previous studies demonstrating that SHMT expression is rate-limiting for de novo thymidylate synthesis and indicate that de novo thymidylate biosynthesis occurs at replication forks.
Collapse
Affiliation(s)
- Donald D Anderson
- Division of Nutritional Sciences, Cornell University, Ithaca, New York 14853, USA
| | | | | | | | | |
Collapse
|
63
|
Shimi T, Butin-Israeli V, Adam SA, Hamanaka RB, Goldman AE, Lucas CA, Shumaker DK, Kosak ST, Chandel NS, Goldman RD. The role of nuclear lamin B1 in cell proliferation and senescence. Genes Dev 2011; 25:2579-93. [PMID: 22155925 DOI: 10.1101/gad.179515.111] [Citation(s) in RCA: 417] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Nuclear lamin B1 (LB1) is a major structural component of the nucleus that appears to be involved in the regulation of many nuclear functions. The results of this study demonstrate that LB1 expression in WI-38 cells decreases during cellular senescence. Premature senescence induced by oncogenic Ras also decreases LB1 expression through a retinoblastoma protein (pRb)-dependent mechanism. Silencing the expression of LB1 slows cell proliferation and induces premature senescence in WI-38 cells. The effects of LB1 silencing on proliferation require the activation of p53, but not pRb. However, the induction of premature senescence requires both p53 and pRb. The proliferation defects induced by silencing LB1 are accompanied by a p53-dependent reduction in mitochondrial reactive oxygen species (ROS), which can be rescued by growth under hypoxic conditions. In contrast to the effects of LB1 silencing, overexpression of LB1 increases the proliferation rate and delays the onset of senescence of WI-38 cells. This overexpression eventually leads to cell cycle arrest at the G1/S boundary. These results demonstrate the importance of LB1 in regulating the proliferation and senescence of human diploid cells through a ROS signaling pathway.
Collapse
Affiliation(s)
- Takeshi Shimi
- Department of Cell and Molecular Biology, Division of Pulmonary and Critical Care Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
64
|
Worman HJ. Nuclear lamins and laminopathies. J Pathol 2011; 226:316-25. [PMID: 21953297 DOI: 10.1002/path.2999] [Citation(s) in RCA: 294] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2011] [Revised: 09/01/2011] [Accepted: 09/15/2011] [Indexed: 12/31/2022]
Abstract
Nuclear lamins are intermediate filament proteins that polymerize to form the nuclear lamina on the inner aspect of the inner nuclear membrane. Long known to be essential for maintaining nuclear structure and disassembling/reassembling during mitosis in metazoans, research over the past dozen years has shown that mutations in genes encoding nuclear lamins, particularly LMNA encoding the A-type lamins, cause a broad range of diverse diseases, often referred to as laminopathies. Lamins are expressed in all mammalian somatic cells but mutations in their genes lead to relatively tissue-selective disease phenotypes in most cases. While mutations causing laminopathies have been shown to produce abnormalities in nuclear morphology, how these disease-causing mutations or resultant alterations in nuclear structure lead to pathology is only starting to be understood. Despite the incomplete understanding of pathogenic mechanisms underlying the laminopathies, basic research in cellular and small animal models has produced promising leads for treatments of these rare diseases.
Collapse
Affiliation(s)
- Howard J Worman
- Departments of Medicine and of Pathology and Cell Biology, College of Physicians and Surgeons, Columbia University, New York, USA.
| |
Collapse
|
65
|
Wakabayashi T, Mori T, Hirahara Y, Koike T, Kubota Y, Takamori Y, Yamada H. Nuclear lamins are differentially expressed in retinal neurons of the adult rat retina. Histochem Cell Biol 2011; 136:427-36. [PMID: 21842415 DOI: 10.1007/s00418-011-0853-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/01/2011] [Indexed: 10/17/2022]
Abstract
Lamins are type V intermediate filament proteins that support nuclear membranes. They are divided into A-type lamins, which include lamin A and C, and B-type lamins, which include lamin B1 and B2. In the rat brain, lamin A and C are expressed in relatively equal amounts, while the expressions of lamin B1 and B2 vary depending on the cell type. Lamins play important roles in normal morphogenesis and function. In the nervous system, their abnormal expression causes several neurodegenerative diseases such as peripheral neuropathy, leukodystrophy and lissencephaly. The retina belongs to the central nervous system (CNS) and has widely been used as a source of CNS neurons. We investigated the expression patterns of lamin subtypes in the adult rat retina by immunohistochemistry and found that the staining patterns differed when compared with the brain. All retinal neurons expressed lamin B1 and B2 in relatively equal amounts. In addition, horizontal cells and a subpopulation of retinal ganglion cells expressed lamin A and C, while photoreceptor cells expressed neither lamin A nor C, and all other retinal neurons expressed lamin C only. This differential expression pattern of lamins in retinal neurons suggests that they may be involved in cellular differentiation and expression of cell-specific genes in individual retinal neurons.
Collapse
Affiliation(s)
- Taketoshi Wakabayashi
- Department of Anatomy and Cell Science, Kansai Medical University, Moriguchi, Osaka, Japan.
| | | | | | | | | | | | | |
Collapse
|
66
|
Disruption of nuclear organization during the initial phase of African swine fever virus infection. J Virol 2011; 85:8263-9. [PMID: 21680527 DOI: 10.1128/jvi.00704-11] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
African swine fever virus (ASFV), the causative agent of one of the most devastating swine diseases, has been considered exclusively cytoplasmic, even though some authors have shown evidence of an early stage of nuclear replication. In the present study, an increment of lamin A/C phosphorylation was observed in ASFV-infected cells as early as 4 h postinfection, followed by the disassembling of the lamina network close to the sites where the viral genome starts its replication. At later time points, this and other nuclear envelope markers were found in the cytoplasm of the infected cells. The effect of the infection on the cell nucleus was much more severe than previously expected, since a redistribution of other nuclear proteins, such as RNA polymerase II, the splicing speckle SC-35 marker, and the B-23 nucleolar marker, was observed from 4 h postinfection. All this evidence, together with the redistribution, dephosphorylation, and subsequent degradation of RNA polymerase II after ASFV infection, suggests the existence of sophisticated mechanisms to regulate the nuclear machinery during viral infection.
Collapse
|
67
|
Gehrig K, Ridgway ND. CTP:phosphocholine cytidylyltransferase α (CCTα) and lamins alter nuclear membrane structure without affecting phosphatidylcholine synthesis. BIOCHIMICA ET BIOPHYSICA ACTA 2011; 1811:377-85. [PMID: 21504799 DOI: 10.1016/j.bbalip.2011.04.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/17/2010] [Revised: 03/28/2011] [Accepted: 04/01/2011] [Indexed: 11/21/2022]
Abstract
CTP:phosphocholine cytidylyltransferase α (CCTα) is a nuclear enzyme that catalyzes the rate-limiting step in the CDP-choline pathway for phosphatidylcholine (PC) synthesis. Lipid activation of CCTα results in its translocation to the nuclear envelope and expansion of an intranuclear membrane network termed the nucleoplasmic reticulum (NR) by a mechanism involving membrane deformation. Nuclear lamins are also required for stability and proliferation of the NR, but whether this unique structure, or the nuclear lamina in general, is required for PC synthesis is not known. To examine this relationship, the nuclear lamina was depleted by RNAi or disrupted by expression of the Hutchinson-Gilford progeria syndrome (HGPS) mutant lamin A (progerin), and the effect on CCTα and choline metabolism was analyzed. siRNA-mediated silencing of lamin A/C or lamin B1 in CHO cells to diminish the NR had no effect on PC synthesis, while double knockdown non-specifically inhibited the pathway. Confirming this minor role in PC synthesis, only 10% of transiently overexpressed choline/ethanolamine phosphotransferase was detected in the NR. In CHO cells, CCTα was nucleoplasmic and co-localized with GFP-progerin in nuclear folds and invaginations; however, HGPS fibroblasts displayed an abnormal distribution of CCTα in the cytoplasm and nuclear envelope that was accompanied by a 2-fold reduction in PC synthesis. In spite of its altered localization, choline-labeling experiments showed that CCT activity was unaffected, and inhibition of PC synthesis was traced to reduced activity of a hemicholinium-sensitive choline transporter. We conclude that CCTα and lamins specifically cooperate to form the NR, but the overall structure of the nuclear envelope has a minimal impact on CCT activity and PC synthesis.
Collapse
Affiliation(s)
- Karsten Gehrig
- Atlantic Research Centre, Departments of Pediatrics and Biochemistry and Molecular Biology, Dalhousie University, Halifax, Nova Scotia, Canada
| | | |
Collapse
|
68
|
Abstract
The lamins are the major architectural proteins of the animal cell nucleus. Lamins line the inside of the nuclear membrane, where they provide a platform for the binding of proteins and chromatin and confer mechanical stability. They have been implicated in a wide range of nuclear functions, including higher-order genome organization, chromatin regulation, transcription, DNA replication and DNA repair. The lamins are members of the intermediate filament (IF) family of proteins, which constitute a major component of the cytoskeleton. Lamins are the only nuclear IFs and are the ancestral founders of the IF protein superfamily. Lamins polymerize into fibers forming a complex protein meshwork in vivo and, like all IF proteins, have a tripartite structure with two globular head and tail domains flanking a central α-helical rod domain, which supports the formation of higher-order polymers. Mutations in lamins cause a large number of diverse human diseases, collectively known as the laminopathies, underscoring their functional importance.
Collapse
Affiliation(s)
- Travis A Dittmer
- National Cancer Institute, National Institutes of Health, Bethesda, MD 20896, USA.
| | | |
Collapse
|
69
|
Cabeen MT, Herrmann H, Jacobs-Wagner C. The domain organization of the bacterial intermediate filament-like protein crescentin is important for assembly and function. Cytoskeleton (Hoboken) 2011; 68:205-19. [PMID: 21360832 PMCID: PMC3087291 DOI: 10.1002/cm.20505] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2011] [Revised: 02/01/2011] [Accepted: 02/02/2011] [Indexed: 01/08/2023]
Abstract
Crescentin is a bacterial filament-forming protein that exhibits domain organization features found in metazoan intermediate filament (IF) proteins. Structure-function studies of eukaryotic IFs have been hindered by a lack of simple genetic systems and easily quantifiable phenotypes. Here we exploit the characteristic localization of the crescentin structure along the inner curvature of Caulobacter crescentus cells and the loss of cell curvature associated with impaired crescentin function to analyze the importance of the domain organization of crescentin. By combining biochemistry and ultrastructural analysis in vitro with cellular localization and functional studies, we show that crescentin requires its distinctive domain organization, and furthermore that different structural elements have distinct structural and functional contributions. The head domain can be functionally subdivided into two subdomains; the first (amino-terminal) is required for function but not assembly, while the second is necessary for structure assembly. The rod domain is similarly required for structure assembly, and the linker L1 appears important to prevent runaway assembly into nonfunctional aggregates. The data also suggest that the stutter and the tail domain have critical functional roles in stabilizing crescentin structures against disassembly by monovalent cations in the cytoplasm. This study suggests that the IF-like behavior of crescentin is a consequence of its domain organization, implying that the IF protein layout is an adaptable cytoskeletal motif, much like the actin and tubulin folds, that is broadly exploited for various functions throughout life from bacteria to humans.
Collapse
Affiliation(s)
- Matthew T Cabeen
- Department of Molecular, Cellular and Developmental Biology, Yale UniversityNew Haven, Connecticut
| | - Harald Herrmann
- Functional Architecture of the Cell Group, German Cancer Research CenterHeidelberg, Germany
| | - Christine Jacobs-Wagner
- Department of Molecular, Cellular and Developmental Biology, Yale UniversityNew Haven, Connecticut
- Section of Microbial Pathogenesis, Yale School of MedicineNew Haven, Connecticut
- Howard Hughes Medical Institute, Yale UniversityNew Haven, Connecticut
| |
Collapse
|
70
|
Abstract
Trypanosomes are a group of protozoan eukaryotes, many of which are major parasites of humans and livestock. The genomes of trypanosomes and their modes of gene expression differ in several important aspects from those of other eukaryotic model organisms. Protein-coding genes are organized in large directional gene clusters on a genome-wide scale, and their polycistronic transcription is not generally regulated at initiation. Transcripts from these polycistrons are processed by global trans-splicing of pre-mRNA. Furthermore, in African trypanosomes, some protein-coding genes are transcribed by a multifunctional RNA polymerase I from a specialized extranucleolar compartment. The primary DNA sequence of the trypanosome genomes and their cellular organization have usually been treated as separate entities. However, it is becoming increasingly clear that in order to understand how a genome functions in a living cell, we will need to unravel how the one-dimensional genomic sequence and its trans-acting factors are arranged in the three-dimensional space of the eukaryotic nucleus. Understanding this cell biology of the genome will be crucial if we are to elucidate the genetic control mechanisms of parasitism. Here, we integrate the concepts of nuclear architecture, deduced largely from studies of yeast and mammalian nuclei, with recent developments in our knowledge of the trypanosome genome, gene expression, and nuclear organization. We also compare this nuclear organization to those in other systems in order to shed light on the evolution of nuclear architecture in eukaryotes.
Collapse
|
71
|
Sui L, Yang Y. Distinct effects of nuclear membrane localization on gene transcription silencing in Drosophila S2 cells and germ cells. J Genet Genomics 2011; 38:55-61. [PMID: 21356524 DOI: 10.1016/j.jcg.2011.01.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2010] [Revised: 01/06/2011] [Accepted: 01/07/2011] [Indexed: 11/29/2022]
Abstract
Nuclear envelope proteins have important roles in chromatin organization and signal-dependent transcriptional regulation. A previous study reported that the inner nuclear membrane protein, Otefin (Ote), was essential for germline stem cell (GSC) maintenance via interaction with Smad complex. The interaction of Ote with the Smad complex recruits the bam locus to the nuclear periphery and subsequently results in bam transcriptional silencing, revealing that nuclear peripheral localization is essential for bam gene regulation. However, it remains unknown whether the nuclear peripheral localization is sufficient for bam silencing. To address this issue, we have established a tethering system, in which the Gal4 DNA binding domain (DBD) of the Flag:Gal4 DBD:Ote▵LEM fusion protein physically interacts with the Gal4 binding sites upstream of bamP-gfp to artificially recruit the reporter gene gfp to the nuclear membrane. Our data demonstrated that the nuclear peripheral localization seemed to affect the expression of the target naked gene in S2 cells. By contrast, in Drosophila germ cells, the nuclear membrane localization was not sufficient for gene silencing.
Collapse
Affiliation(s)
- Lu Sui
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | | |
Collapse
|
72
|
Mitsuhashi H, Hayashi YK, Matsuda C, Noguchi S, Wakatsuki S, Araki T, Nishino I. Specific phosphorylation of Ser458 of A-type lamins in LMNA-associated myopathy patients. J Cell Sci 2010; 123:3893-900. [DOI: 10.1242/jcs.072157] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Mutations in LMNA, which encodes A-type nuclear lamins, cause various human diseases, including myopathy, cardiomyopathy, lipodystrophy and progeria syndrome. To date, little is known about how mutations in a single gene cause a wide variety of diseases. Here, by characterizing an antibody that specifically recognizes the phosphorylation of Ser458 of A-type lamins, we uncover findings that might contribute to our understanding of laminopathies. This antibody only reacts with nuclei in muscle biopsies from myopathy patients with mutations in the Ig-fold motif of A-type lamins. Ser458 phosphorylation is not seen in muscles from control patients or patients with any other neuromuscular diseases. In vitro analysis confirmed that only lamin A mutants associated with myopathy induce phosphorylation of Ser458, whereas lipodystrophy- or progeria-associated mutants do not. We also found that Akt1 directly phosphorylates Ser458 of lamin A with myopathy-related mutations in vitro. These results suggest that Ser458 phosphorylation of A-type lamins correlates with striated muscle laminopathies; this might be useful for the early diagnosis of LMNA-associated myopathies. We propose that disease-specific phosphorylation of A-type lamins by Akt1 contributes to myopathy caused by LMNA mutations.
Collapse
Affiliation(s)
- Hiroaki Mitsuhashi
- Department of Neuromuscular Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, 4-1-1 Ogawa-higashi, Kodaira, Tokyo 187-8502, Japan
| | - Yukiko K. Hayashi
- Department of Neuromuscular Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, 4-1-1 Ogawa-higashi, Kodaira, Tokyo 187-8502, Japan
| | - Chie Matsuda
- Neuroscience Research Institute, AIST, Central 6, Tsukuba, Ibaraki 305-8566, Japan
| | - Satoru Noguchi
- Department of Neuromuscular Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, 4-1-1 Ogawa-higashi, Kodaira, Tokyo 187-8502, Japan
| | - Shuji Wakatsuki
- Department of Peripheral Nervous System Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Tokyo 187-8502, Japan
| | - Toshiyuki Araki
- Department of Peripheral Nervous System Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Tokyo 187-8502, Japan
| | - Ichizo Nishino
- Department of Neuromuscular Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, 4-1-1 Ogawa-higashi, Kodaira, Tokyo 187-8502, Japan
| |
Collapse
|
73
|
Lebofsky R, van Oijen AM, Walter JC. DNA is a co-factor for its own replication in Xenopus egg extracts. Nucleic Acids Res 2010; 39:545-55. [PMID: 20861001 PMCID: PMC3025559 DOI: 10.1093/nar/gkq739] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Soluble Xenopus egg extracts efficiently replicate added plasmids using a physiological mechanism, and thus represent a powerful system to understand vertebrate DNA replication. Surprisingly, DNA replication in this system is highly sensitive to plasmid concentration, being undetectable below ∼10 pM and highly efficient above ∼75 pM. DNA replication at the high plasmid concentration does not require plasmid-plasmid contacts, since replication is not inhibited when plasmids are immobilized in agarose prior to addition of egg extract. The absence of replication at low plasmid concentration is due to a defect in the assembly of pre-replication complexes (pre-RCs). pre-RC assembly requires contact-independent communication between plasmids. Our results show that in Xenopus egg extracts, aggregation of multiple replication forks is not required for efficient replication of plasmid DNA, and they suggest that DNA functions as a co-factor for its own duplication.
Collapse
Affiliation(s)
- Ronald Lebofsky
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA
| | | | | |
Collapse
|
74
|
Kuga T, Nozaki N, Matsushita K, Nomura F, Tomonaga T. Phosphorylation statuses at different residues of lamin B2, B1, and A/C dynamically and independently change throughout the cell cycle. Exp Cell Res 2010; 316:2301-12. [PMID: 20580708 DOI: 10.1016/j.yexcr.2010.05.017] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2009] [Revised: 04/23/2010] [Accepted: 05/18/2010] [Indexed: 11/17/2022]
Abstract
Lamins, major components of the nuclear lamina, undergo phosphorylation at multiple residues during cell cycle progression, but their detailed phosphorylation kinetics remain largely undetermined. Here, we examined changes in the phosphorylation of major phosphorylation residues (Thr14, Ser17, Ser385, Ser387, and Ser401) of lamin B2 and the homologous residues of lamin B1, A/C during the cell cycle using novel antibodies to the site-specific phosphorylation. The phosphorylation levels of these residues independently changed during the cell cycle. Thr14 and Ser17 were phosphorylated during G(2)/M phase to anaphase/telophase. Ser385 was persistently phosphorylated during mitosis to G(1) phase, whereas Ser387 was phosphorylated discontinuously in prophase and G(1) phase. Ser401 phosphorylation was enhanced in the G(1)/S boundary. Immunoprecipitation using the phospho-antibodies suggested that metaphase-phosphorylation at Thr14, Ser17, and Ser385 of lamins occurred simultaneously, whereas G(1)-phase phosphorylation at Ser385 and Ser387 occurred in distinct pools or with different timings. Additionally, we showed that lamin B2 phosphorylated at Ser17, but not Ser385, Ser387 and Ser401, was exclusively non-ionic detergent soluble, depolymerized forms in growing cells, implicating specific involvement of Ser17 phosphorylation in lamin depolymerization and nuclear envelope breakdown. These results suggest that the phosphorylations at different residues of lamins might play specific roles throughout the cell cycle.
Collapse
Affiliation(s)
- Takahisa Kuga
- Laboratory of Proteome Research, National Institute of Biomedical Innovation, Ibaraki, Osaka 567-0085, Japan.
| | | | | | | | | |
Collapse
|
75
|
Dialynas G, Speese S, Budnik V, Geyer PK, Wallrath LL. The role of Drosophila Lamin C in muscle function and gene expression. Development 2010; 137:3067-77. [PMID: 20702563 DOI: 10.1242/dev.048231] [Citation(s) in RCA: 97] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The inner side of the nuclear envelope (NE) is lined with lamins, a meshwork of intermediate filaments that provides structural support for the nucleus and plays roles in many nuclear processes. Lamins, classified as A- or B-types on the basis of biochemical properties, have a conserved globular head, central rod and C-terminal domain that includes an Ig-fold structural motif. In humans, mutations in A-type lamins give rise to diseases that exhibit tissue-specific defects, such as Emery-Dreifuss muscular dystrophy. Drosophila is being used as a model to determine tissue-specific functions of A-type lamins in development, with implications for understanding human disease mechanisms. The GAL4-UAS system was used to express wild-type and mutant forms of Lamin C (the presumed Drosophila A-type lamin), in an otherwise wild-type background. Larval muscle-specific expression of wild type Drosophila Lamin C caused no overt phenotype. By contrast, larval muscle-specific expression of a truncated form of Lamin C lacking the N-terminal head (Lamin C DeltaN) caused muscle defects and semi-lethality, with adult 'escapers' possessing malformed legs. The leg defects were due to a lack of larval muscle function and alterations in hormone-regulated gene expression. The consequences of Lamin C association at a gene were tested directly by targeting a Lamin C DNA-binding domain fusion protein upstream of a reporter gene. Association of Lamin C correlated with localization of the reporter gene at the nuclear periphery and gene repression. These data demonstrate connections among the Drosophila A-type lamin, hormone-induced gene expression and muscle function.
Collapse
Affiliation(s)
- George Dialynas
- Department of Biochemistry, University of Iowa, Iowa City, IA 52241, USA
| | | | | | | | | |
Collapse
|
76
|
Musich PR, Zou Y. Genomic instability and DNA damage responses in progeria arising from defective maturation of prelamin A. Aging (Albany NY) 2010; 1:28-37. [PMID: 19851476 PMCID: PMC2765059 DOI: 10.18632/aging.100012] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Progeria syndromes have in common a premature aging phenotype and increased genome instability. The susceptibility to DNA damage arises from a compromised repair system, either in the repair proteins themselves or in the DNA damage response pathways. The most severe progerias stem from mutations affecting lamin A production, a filamentous protein of the nuclear lamina. Hutchinson-Gilford progeria syndrome (HGPS) patients are heterozygous for aLMNA gene mutation while Restrictive Dermopathy (RD) individuals have a homozygous deficiency in the processing protease Zmpste24. These mutations generate the mutant lamin A proteins progerin and FC-lamina A, respectively, which cause nuclear deformations and chromatin perturbations. Genome instability is observed even though genome maintenance and repair genes appear normal. The unresolved question is what features of the DNA damage response pathways are deficient in HGPS and RD cells. Here we review and discuss recent findings which resolve some mechanistic details of how the accumulation of progerin/FC-lamin A proteins may disrupt DNA damage response pathways in HGPS and RD cells. As the mutant lamin proteins accumulate they sequester replication and repair factors, leading to stalled replication forks which collapse into DNA double-strand beaks (DSBs). In a reaction unique to HGPS and RD cells these accessible DSB termini bind Xeroderma pigmentosum group A (XPA) protein which excludes normal binding by DNA DSB repair proteins. The bound XPA also signals activation of ATM and ATR, arresting cell cycle progression, leading to arrested growth. In addition, the effective sequestration of XPA at these DSB damage sites makes HGPS and RD cells more sensitive to ultraviolet light and other mutagens normally repaired by the nucleotide excision repair pathway of which XPA is a necessary and specific component.
Collapse
Affiliation(s)
- Phillip R Musich
- Department of Biochemistry & Molecular Biology, Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614-0581, USA
| | | |
Collapse
|
77
|
Abstract
A- and B-type lamins are the major intermediate filaments of the nucleus. Lamins engage in a plethora of stable and transient interactions, near the inner nuclear membrane and throughout the nucleus. Lamin-binding proteins serve an amazingly diverse range of functions. Numerous inner-membrane proteins help anchor lamin filaments to the nuclear envelope, serving as part of the nuclear "lamina" network that is essential for nuclear architecture and integrity. Certain lamin-binding proteins of the inner membrane bind partners in the outer membrane and mechanically link lamins to the cytoskeleton. Inside the nucleus, lamin-binding proteins appear to serve as the "adaptors" by which the lamina organizes chromatin, influences gene expression and epigenetic regulation, and modulates signaling pathways. Transient interactions of lamins with key components of the transcription and replication machinery may provide an additional level of regulation or support to these essential events.
Collapse
Affiliation(s)
- Katherine L Wilson
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| | | |
Collapse
|
78
|
Abstract
Nuclear intermediate filaments formed by A- and B-type lamins are central components of the nucleoskeleton and are required for the architecture and integrity of the nucleus. There is growing evidence that lamins are also involved in regulatory pathways controlling cell proliferation and differentiation. Lamins affect the activity of several transcription factors, such as retinoblastoma protein and c-Fos, and signalling pathways, such as the ERK1/2 (extracellular-signal-regulated kinase 1/2) and Notch pathways, which are key regulators of cell-cycle progression and differentiation. During mitosis, lamins are dynamically reorganized and play active roles in spindle matrix formation and in post-mitotic nuclear reassembly. Several of the cell-cycle-regulating functions of lamins may be impaired in the diseases linked to mutations in lamins and lamin-associated proteins, including striated muscle diseases, lipodystrophies and premature aging syndromes, and contribute to the tissue-specific disease pathologies.
Collapse
|
79
|
Abstract
The nuclear periphery is a specialized environment in the nucleus that contributes to genome organization and correspondingly to gene regulation. Mammalian chromosomes and certain genes occupy defined positions within the nucleus that are heritable and tissue specific. Genes located at the nuclear periphery tend to be inactive and this negative regulation can be reversed when they are released from the periphery in certain differentiation systems. Recent work using specially designed systems has shown that genes can be artificially tethered to the nuclear periphery by an affinity mechanism. The next important step will be to identify the endogenous NE (nuclear envelope) and chromatin proteins that participate in affinity-driven NE tethering and determine how they are regulated.
Collapse
|
80
|
Cortés R, Roselló-Lletí E, Rivera M, Martínez-Dolz L, Azorín I, Salvador A, Portolés M. The role of the nuclear lamins in the pathogenesis of heart failure in patients undergoing cardiac transplantation. Transplant Proc 2010; 41:2227-30. [PMID: 19715882 DOI: 10.1016/j.transproceed.2009.06.013] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
The nuclear lamins A/C play a critical role in maintaining the structure of the nuclear lamina and the organization of various proteins, such as emerin. These protein levels may play roles in the pathogenesis and clinical evolution of both ischemic (ICM) and dilated (DCM) cardiomyopathy. We evaluated the nuclear morphology of cardiomyocytes and determine lamins A/C and emerin levels among DCM and ICM heart failure patients compared with control human hearts. We determined protein levels by Western blots using mouse monoclonal antibodies in 23 explanted human hearts. Lamin A was increased in failing hearts but significantly different only among the DCM compared with the control group: mean, 236 +/- 51 vs 100 +/- 34; (P < .05). However, lamin C in the DCM group was near control values and significantly decreased in the ICM cohort compared to controls 75 +/- 7 versus 100 +/- 3 (P < .05). No alterations in emerin levels were observed in ICM or DCM, compared with controls. In conclusion, hearts with ICM or DCM showed different alterations in the nuclear morphology of cardiomyocytes; ICM patients had decreased lamin C, whereas DCM patients had increased lamin A. These changes affecting nuclear structure and function may have prognostic implications, for cardiomyopathy etiology.
Collapse
Affiliation(s)
- R Cortés
- Unidad de Cardiocirculación, Centro de Investigación, Hospital Universitario La Fe, Valencia, Spain
| | | | | | | | | | | | | |
Collapse
|
81
|
von Moeller F, Barendziak T, Apte K, Goldberg MW, Stick R. Molecular characterization of Xenopus lamin LIV reveals differences in the lamin composition of sperms in amphibians and mammals. Nucleus 2010; 1:85-95. [PMID: 21327107 PMCID: PMC3035121 DOI: 10.4161/nucl.1.1.10517] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2009] [Revised: 11/04/2009] [Accepted: 11/04/2009] [Indexed: 02/06/2023] Open
Abstract
Lamins are nuclear intermediate filament proteins. They are involved in most nuclear activities and are essential for retaining the mechano-elastic properties of the nucleus. Somatic cells of vertebrates express lamins A, B1 and B2 while lamin LIII, a major component of the amphibian oocyte lamina is absent in mammals. The organization of the lamina of germ cells differs significantly from that of somatic cells. Mammalian spermatogenic cells express two short lamins, C2 and B3, that are splice isoforms of lamin A and B2, respectively. Here we identify the previously described Xenopus lamin LIV as splice variant of the lamin LIII gene. LIV contains 40 extra residues in coil 2A of the rod domain, which results in altered assembly properties. Xenopus lamin LIV and mammalian B3 assemble into short structures rather than into long IF-like filaments. Expression of lamin LIV is restricted to male germ cells suggesting that it might be the functional equivalent of mammalian lamin B3. We provide evidence that lamins C2 and B3 are restricted to the mammalian lineage and describe the lamin composition of Xenopus sperm. Our results show that the evolution of germ cell-specific lamins followed separate and distinctly different paths in amphibians and mammals.
Collapse
Affiliation(s)
| | - Tanja Barendziak
- Department of Cell Biology; University of Bremen; Bremen, Germany
| | - Ketaki Apte
- Department of Cell Biology; University of Bremen; Bremen, Germany
| | - Martin W Goldberg
- School of Biological and Biomedical Sciences; The University of Durham; Durham, UK
| | - Reimer Stick
- Department of Cell Biology; University of Bremen; Bremen, Germany
| |
Collapse
|
82
|
Gonzalez-Suarez I, Gonzalo S. Nurturing the genome: A-type lamins preserve genomic stability. Nucleus 2009; 1:129-35. [PMID: 21326943 DOI: 10.4161/nucl.1.2.10797] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2009] [Revised: 11/24/2009] [Accepted: 11/29/2009] [Indexed: 01/17/2023] Open
Abstract
A-type lamins provide a scaffold for tethering chromatin and protein complexes regulating nuclear structure and function. Interest in lamins increased after mutations in the LMNA gene were found to be associated with a variety of human disorders termed laminopathies. These include muscular dystrophy, cardiomyopathy, lipodystrophy, peripheral neuropathy and premature aging syndromes such as progeria. In addition, altered expression of A-type lamins is emerging as a contributing factor to tumorigenesis. How different alterations in a gene that is ubiquitously expressed can cause such an array of systemic as well as tissue specific diseases remains an enigma. Several lines of evidence indicate that mutant forms of A-type lamins impact on genome function and integrity. A current model suggests that genomic instability plays a major part in the pathophysiology of some lamin-related diseases. However, this model remains to be fully investigated. Here we discuss recent studies revealing novel functions for A-type lamins in the maintenance of telomeres and in the DNA damage response (DDR) pathway. These findings have shed some light onto the putative molecular mechanisms by which alterations in A-type lamins induce genomic instability and contribute to disease.
Collapse
Affiliation(s)
- Ignacio Gonzalez-Suarez
- Radiation and Cancer Biology Division, Department of Radiation Oncology, Washington University School of Medicine, St. Louis, MO, USA
| | | |
Collapse
|
83
|
Schulze SR, Curio-Penny B, Speese S, Dialynas G, Cryderman DE, McDonough CW, Nalbant D, Petersen M, Budnik V, Geyer PK, Wallrath LL. A comparative study of Drosophila and human A-type lamins. PLoS One 2009; 4:e7564. [PMID: 19855837 PMCID: PMC2762312 DOI: 10.1371/journal.pone.0007564] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2009] [Accepted: 10/04/2009] [Indexed: 11/24/2022] Open
Abstract
Nuclear intermediate filament proteins, called lamins, form a meshwork that lines the inner surface of the nuclear envelope. Lamins contain three domains: an N-terminal head, a central rod and a C-terminal tail domain possessing an Ig-fold structural motif. Lamins are classified as either A- or B-type based on structure and expression pattern. The Drosophila genome possesses two genes encoding lamins, Lamin C and lamin Dm0, which have been designated A- and B-type, respectively, based on their expression profile and structural features. In humans, mutations in the gene encoding A-type lamins are associated with a spectrum of predominantly tissue-specific diseases known as laminopathies. Linking the disease phenotypes to cellular functions of lamins has been a major challenge. Drosophila is being used as a model system to identify the roles of lamins in development. Towards this end, we performed a comparative study of Drosophila and human A-type lamins. Analysis of transgenic flies showed that human lamins localize predictably within the Drosophila nucleus. Consistent with this finding, yeast two-hybrid data demonstrated conservation of partner-protein interactions. Drosophila lacking A-type lamin show nuclear envelope defects similar to those observed with human laminopathies. Expression of mutant forms of the A-type Drosophila lamin modeled after human disease-causing amino acid substitutions revealed an essential role for the N-terminal head and the Ig-fold in larval muscle tissue. This tissue-restricted sensitivity suggests a conserved role for lamins in muscle biology. In conclusion, we show that (1) localization of A-type lamins and protein-partner interactions are conserved between Drosophila and humans, (2) loss of the Drosophila A-type lamin causes nuclear defects and (3) muscle tissue is sensitive to the expression of mutant forms of A-type lamin modeled after those causing disease in humans. These studies provide new insights on the role of lamins in nuclear biology and support Drosophila as a model for studies of human laminopathies involving muscle dysfunction.
Collapse
Affiliation(s)
- Sandra R. Schulze
- Department of Biology, Western Washington University, Bellingham, Washington, United States of America
| | - Beatrice Curio-Penny
- Department of Biochemistry, University of Iowa, Iowa City, Iowa, United States of America
| | - Sean Speese
- Department of Neurobiology, University of Massachusetts, Wochester, Massachusetts, United States of America
| | - George Dialynas
- Department of Biochemistry, University of Iowa, Iowa City, Iowa, United States of America
| | - Diane E. Cryderman
- Department of Biochemistry, University of Iowa, Iowa City, Iowa, United States of America
| | - Caitrin W. McDonough
- Department of Biochemistry, University of Iowa, Iowa City, Iowa, United States of America
| | - Demet Nalbant
- Department of Biochemistry, University of Iowa, Iowa City, Iowa, United States of America
| | - Melissa Petersen
- Department of Biology, Western Washington University, Bellingham, Washington, United States of America
| | - Vivian Budnik
- Department of Neurobiology, University of Massachusetts, Wochester, Massachusetts, United States of America
| | - Pamela K. Geyer
- Department of Biochemistry, University of Iowa, Iowa City, Iowa, United States of America
| | - Lori L. Wallrath
- Department of Biochemistry, University of Iowa, Iowa City, Iowa, United States of America
- * E-mail:
| |
Collapse
|
84
|
Abstract
During mitosis in metazoans, the nuclear envelope (NE) breaks down at prophase and reassembles at telophase. The regulation of NE assembly is essential to correct cell functioning. The complex issue of the regulation of NE formation remains to be solved. It is still uncertain that a single mechanism depicts NE formation during mitosis. The aim of this review is to address some of the cytological, biophysical, and molecular aspects of models of NE formation. Our emphasis is on the role of lipids and their modifying enzymes in envelope assembly. We consider how the NE can be used as a model in characterizing membrane dynamics during membrane fusion. Fusion mechanisms that give insight into the formation of the double membrane of the envelope are summarized. We speculate on the possible roles of phosphoinositides in membrane fusion and NE formation.
Collapse
Affiliation(s)
- Banafshé Larijani
- Lincoln's Inn Fields Laboratories, Cancer Research UK, London WC2A 3PX, UK.
| | | |
Collapse
|
85
|
Prokocimer M, Davidovich M, Nissim-Rafinia M, Wiesel-Motiuk N, Bar DZ, Barkan R, Meshorer E, Gruenbaum Y. Nuclear lamins: key regulators of nuclear structure and activities. J Cell Mol Med 2009; 13:1059-85. [PMID: 19210577 PMCID: PMC4496104 DOI: 10.1111/j.1582-4934.2008.00676.x] [Citation(s) in RCA: 207] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2008] [Accepted: 01/19/2009] [Indexed: 11/27/2022] Open
Abstract
The nuclear lamina is a proteinaceous structure located underneath the inner nuclear membrane (INM), where it associates with the peripheral chromatin. It contains lamins and lamin-associated proteins, including many integral proteins of the INM, chromatin modifying proteins, transcriptional repressors and structural proteins. A fraction of lamins is also present in the nucleoplasm, where it forms stable complexes and is associated with specific nucleoplasmic proteins. The lamins and their associated proteins are required for most nuclear activities, mitosis and for linking the nucleoplasm to all major cytoskeletal networks in the cytoplasm. Mutations in nuclear lamins and their associated proteins cause about 20 different diseases that are collectively called laminopathies'. This review concentrates mainly on lamins, their structure and their roles in DNA replication, chromatin organization, adult stem cell differentiation, aging, tumorogenesis and the lamin mutations leading to laminopathic diseases.
Collapse
Affiliation(s)
- Miron Prokocimer
- Department of Genetics, The Institute of Life Sciences, The Hebrew University of JerusalemJerusalem, Israel
| | - Maya Davidovich
- Department of Genetics, The Institute of Life Sciences, The Hebrew University of JerusalemJerusalem, Israel
| | - Malka Nissim-Rafinia
- Department of Genetics, The Institute of Life Sciences, The Hebrew University of JerusalemJerusalem, Israel
| | - Naama Wiesel-Motiuk
- Department of Genetics, The Institute of Life Sciences, The Hebrew University of JerusalemJerusalem, Israel
| | - Daniel Z Bar
- Department of Genetics, The Institute of Life Sciences, The Hebrew University of JerusalemJerusalem, Israel
| | - Rachel Barkan
- Department of Genetics, The Institute of Life Sciences, The Hebrew University of JerusalemJerusalem, Israel
| | - Eran Meshorer
- Department of Genetics, The Institute of Life Sciences, The Hebrew University of JerusalemJerusalem, Israel
| | - Yosef Gruenbaum
- Department of Genetics, The Institute of Life Sciences, The Hebrew University of JerusalemJerusalem, Israel
| |
Collapse
|
86
|
Akgül B, Zigrino P, Frith D, Hanrahan S, Storey A. Proteomic analysis reveals the actin cytoskeleton as cellular target for the human papillomavirus type 8. Virology 2009; 386:1-5. [PMID: 19243805 DOI: 10.1016/j.virol.2009.01.036] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2008] [Revised: 09/25/2008] [Accepted: 01/16/2009] [Indexed: 10/21/2022]
Abstract
Recent studies strongly support a role of human papillomavirus type 8 (HPV8) in non-melanoma skin cancer development. In this study, a quantitative two-dimensional (2D) differential gene expression (DiGE) gel approach combined with mass spectrometry has been used to identify proteins that are abundantly deregulated in primary human epidermal keratinocytes expressing HPV8 sequences. Twenty six protein spots showed significant changes in the level of expression between keratinocytes expressing E7 or the complete early region (CER) of HPV8 compared to extracts from cells lacking HPV8 gene expression. No differences between HPV8 E7 alone and HPV8 CER expressing cells were observed. The 26 protein spots that were differentially expressed corresponded to 20 different proteins, of which 14 actin-associated proteins were downregulated except for calponin-2, which was the only actin-binding protein that was overexpressed. Besides changes in actin modulating proteins, an upregulation of cytokeratins (CK) 5, 6 and 14 was also noted. This study suggests that the actin and keratin cytoskeleton modulating proteins are targets for HPV8.
Collapse
Affiliation(s)
- Baki Akgül
- Institute of Cell and Molecular Science, Skin Tumour Laboratory, Cancer Research UK, 4 Newark Street, London E12AT, UK.
| | | | | | | | | |
Collapse
|
87
|
Ma L, Tsai MY, Wang S, Lu B, Chen R, Yates JR, Zhu X, Zheng Y. Requirement for Nudel and dynein for assembly of the lamin B spindle matrix. Nat Cell Biol 2009; 11:247-56. [PMID: 19198602 PMCID: PMC2699591 DOI: 10.1038/ncb1832] [Citation(s) in RCA: 93] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2008] [Accepted: 11/19/2008] [Indexed: 01/14/2023]
Abstract
The small guanosine triphosphatase Ran loaded with GTP (RanGTP) can stimulate assembly of the type V intermediate filament protein lamin B into a membranous lamin B spindle matrix, which is required for proper microtubule organization during spindle assembly. Microtubules in turn enhance assembly of the matrix. Here we report that the isolated matrix contains known spindle assembly factors such as dynein and Nudel. Using spindle assembly assays in Xenopus egg extracts, we show that Nudel regulates microtubule organization during spindle assembly independently of its function at kinetochores. Importantly, Nudel interacts directly with lamin B to facilitate the accumulation and assembly of lamin-B-containing matrix on microtubules in a dynein-dependent manner. Perturbing either Nudel or dynein inhibited the assembly of lamin B matrix. However, depleting lamin B still allowed the formation of matrices containing dynein and Nudel. Therefore, dynein and Nudel regulate assembly of the lamin B matrix. Interestingly, we found that whereas depleting lamin B resulted in disorganized spindle and spindle poles, disrupting the function of Nudel or dynein caused a complete lack of spindle pole focusing. We suggest that Nudel regulates microtubule organization in part by facilitating assembly of the lamin B spindle matrix in a dynein-dependent manner.
Collapse
Affiliation(s)
- Li Ma
- Laboratory of Molecular Cell Biology and Center of Cell Signaling, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
- Department of Embryology, Carnegie Institution for Science and Howard Hughes Medical Institute, Baltimore, Maryland, United States of America
| | - Ming-Ying Tsai
- Department of Embryology, Carnegie Institution for Science and Howard Hughes Medical Institute, Baltimore, Maryland, United States of America
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Shusheng Wang
- Department of Embryology, Carnegie Institution for Science and Howard Hughes Medical Institute, Baltimore, Maryland, United States of America
| | - Bingwen Lu
- Department of Cell Biology, The Scripps Research Institute, La Jolla, California, United States of America
| | - Rong Chen
- Department of Embryology, Carnegie Institution for Science and Howard Hughes Medical Institute, Baltimore, Maryland, United States of America
| | - John R. Yates
- Department of Cell Biology, The Scripps Research Institute, La Jolla, California, United States of America
| | - Xueliang Zhu
- Laboratory of Molecular Cell Biology and Center of Cell Signaling, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Yixian Zheng
- Laboratory of Molecular Cell Biology and Center of Cell Signaling, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
- Department of Embryology, Carnegie Institution for Science and Howard Hughes Medical Institute, Baltimore, Maryland, United States of America
| |
Collapse
|
88
|
Isokane M, Hieda M, Hirakawa S, Shudou M, Nakashiro K, Hashimoto K, Hamakawa H, Higashiyama S. Plasma-membrane-anchored growth factor pro-amphiregulin binds A-type lamin and regulates global transcription. J Cell Sci 2009; 121:3608-18. [PMID: 18946024 DOI: 10.1242/jcs.031443] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Amphiregulin (AR), a member of the EGF family, is synthesized as a type I transmembrane protein precursor (proAR) and expressed on the cell surface. Shedding of proAR yields a transmembrane-cytoplasmic fragment (AR-CTF), as well as a soluble AR. Here we demonstrate that the proAR-shedding stimuli trigger endocytosis of both AR-CTF and un-shed proAR. ProAR translocates from the plasma membrane to the inner nuclear membrane, whereas AR-CTF is translocated to the lysosome via retrograde membrane trafficking. Nuclear envelope localization of proAR involves truncation of the C-terminus, which subsequently activates the ER-retrieval signal. The truncated form of proAR interacts with A-type lamin and is retained at the inner nuclear membrane. Heterochromatin formation is then induced and global transcription is transiently suppressed. This study gives new insight into epigenetic chromatin organization in mammalian cells: a plasma-membrane-anchored growth factor is targeted to the inner nuclear membrane where it participates in dynamic chromatin organization and control of transcription.
Collapse
Affiliation(s)
- Mayumi Isokane
- Department of Biochemistry and Molecular Genetics, Ehime University Graduate School of Medicine, Toon, Ehime 791-0295, Japan
| | | | | | | | | | | | | | | |
Collapse
|
89
|
Dechat T, Adam SA, Goldman RD. Nuclear lamins and chromatin: when structure meets function. ADVANCES IN ENZYME REGULATION 2008; 49:157-66. [PMID: 19154754 PMCID: PMC3253622 DOI: 10.1016/j.advenzreg.2008.12.003] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Affiliation(s)
- Thomas Dechat
- Northwestern University Medical School, Department of Cell and Molecular Biology, Chicago, Illinois 60611, USA
| | - Stephen A. Adam
- Northwestern University Medical School, Department of Cell and Molecular Biology, Chicago, Illinois 60611, USA
| | - Robert D. Goldman
- Northwestern University Medical School, Department of Cell and Molecular Biology, Chicago, Illinois 60611, USA
| |
Collapse
|
90
|
Kudlow BA, Stanfel MN, Burtner CR, Johnston ED, Kennedy BK. Suppression of proliferative defects associated with processing-defective lamin A mutants by hTERT or inactivation of p53. Mol Biol Cell 2008; 19:5238-48. [PMID: 18843043 PMCID: PMC2592682 DOI: 10.1091/mbc.e08-05-0492] [Citation(s) in RCA: 99] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2008] [Revised: 09/12/2008] [Accepted: 10/01/2008] [Indexed: 01/11/2023] Open
Abstract
Hutchinson-Gilford progeria syndrome (HGPS) is a rare, debilitating disease with early mortality and rapid onset of aging-associated pathologies. It is linked to mutations in LMNA, which encodes A-type nuclear lamins. The most frequent HGPS-associated LMNA mutation results in a protein, termed progerin, with an internal 50 amino acid deletion and, unlike normal A-type lamins, stable farnesylation. The cellular consequences of progerin expression underlying the HGPS phenotype remain poorly understood. Here, we stably expressed lamin A mutants, including progerin, in otherwise identical primary human fibroblasts to compare the effects of different mutants on nuclear morphology and cell proliferation. We find that expression of progerin leads to inhibition of proliferation in a high percentage of cells and slightly premature senescence in the population. Expression of a stably farnesylated mutant of lamin A phenocopied the immediate proliferative defects but did not result in premature senescence. Either p53 inhibition or, more surprisingly, expression of the catalytic subunit of telomerase (hTERT) suppressed the early proliferative defects associated with progerin expression. These findings lead us to propose that progerin may interfere with telomere structure or metabolism in a manner suppressible by increased telomerase levels and possibly link mechanisms leading to progeroid phenotypes to those of cell immortalization.
Collapse
Affiliation(s)
- Brian A. Kudlow
- Department of Biochemistry, University of Washington, Seattle, WA 98195
| | | | | | | | - Brian K. Kennedy
- Department of Biochemistry, University of Washington, Seattle, WA 98195
| |
Collapse
|
91
|
Batrakou DG, Kerr ARW, Schirmer EC. Comparative proteomic analyses of the nuclear envelope and pore complex suggests a wide range of heretofore unexpected functions. J Proteomics 2008; 72:56-70. [PMID: 18852071 DOI: 10.1016/j.jprot.2008.09.004] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2008] [Revised: 08/26/2008] [Accepted: 09/18/2008] [Indexed: 12/31/2022]
Abstract
Since the discovery of several inherited diseases linked to the nuclear envelope the number of functions ascribed to this subcellular organelle has skyrocketed. However the molecular pathways underlying these functions are not clear in most cases, perhaps because of missing components. Several recent proteomic analyses of the nuclear envelope and nuclear pore complex proteomes have yielded not only enough missing components to potentially elucidate these pathways, but suggest an exponentially greater number of functions at the nuclear periphery than ever imagined. Many of these functions appear to derive from recapitulation of pathways utilized at the plasma membrane and from other membrane systems. Additionally, many proteins identified in the comparative nuclear envelope studies have sequence characteristics suggesting that they might also contribute to nuclear pore complex functions. In particular, the striking enrichment for proteins in the nuclear envelope fractions that carry phenylalanine-glycine (FG) repeats may be significant for the mechanism of nuclear transport. In retrospect, these findings are only surprising in context of the notion held for many years that the nuclear envelope was only a barrier protecting the genome. In fact, it is arguably the most complex membrane organelle in the cell.
Collapse
Affiliation(s)
- Dzmitry G Batrakou
- Wellcome Trust Centre for Cell Biology and Institute of Cell Biology, University of Edinburgh, Edinburgh EH9 3JR, UK
| | | | | |
Collapse
|
92
|
Schirmer EC. The epigenetics of nuclear envelope organization and disease. Mutat Res 2008; 647:112-21. [PMID: 18722388 DOI: 10.1016/j.mrfmmm.2008.07.012] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2008] [Revised: 07/16/2008] [Accepted: 07/23/2008] [Indexed: 01/09/2023]
Abstract
Mammalian chromosomes and some specific genes have non-random positions within the nucleus that are tissue-specific and heritable. Work in many organisms has shown that genes at the nuclear periphery tend to be inactive and altering their partitioning to the interior results in their activation. Proteins of the nuclear envelope can recruit chromatin with specific epigenetic marks and can also recruit silencing factors that add new epigenetic modifications to chromatin sequestered at the periphery. Together these findings indicate that the nuclear envelope is a significant epigenetic regulator. The importance of this function is emphasized by observations of aberrant distribution of peripheral heterochromatin in several human diseases linked to mutations in NE proteins. These debilitating inherited diseases range from muscular dystrophies to the premature aging progeroid syndromes and the heterochromatin changes are just one early clue for understanding the molecular details of how they work. The architecture of the nuclear envelope provides a unique environment for epigenetic regulation and as such a great deal of research will be required before we can ascertain the full range of its contributions to epigenetics.
Collapse
Affiliation(s)
- Eric C Schirmer
- Wellcome Trust Centre for Cell Biology, University of Edinburgh, Edinburgh EH9 3JR, UK.
| |
Collapse
|
93
|
Pekovic V, Hutchison CJ. Adult stem cell maintenance and tissue regeneration in the ageing context: the role for A-type lamins as intrinsic modulators of ageing in adult stem cells and their niches. J Anat 2008; 213:5-25. [PMID: 18638067 PMCID: PMC2475560 DOI: 10.1111/j.1469-7580.2008.00928.x] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/28/2008] [Indexed: 02/06/2023] Open
Abstract
Adult stem cells have been identified in most mammalian tissues of the adult body and are known to support the continuous repair and regeneration of tissues. A generalized decline in tissue regenerative responses associated with age is believed to result from a depletion and/or a loss of function of adult stem cells, which itself may be a driving cause of many age-related disease pathologies. Here we review the striking similarities between tissue phenotypes seen in many degenerative conditions associated with old age and those reported in age-related nuclear envelope disorders caused by mutations in the LMNA gene. The concept is beginning to emerge that nuclear filament proteins, A-type lamins, may act as signalling receptors in the nucleus required for receiving and/or transducing upstream cytosolic signals in a number of pathways central to adult stem cell maintenance as well as adaptive responses to stress. We propose that during ageing and in diseases caused by lamin A mutations, dysfunction of the A-type lamin stress-resistant signalling network in adult stem cells, their progenitors and/or stem cell niches leads to a loss of protection against growth-related stress. This in turn triggers an inappropriate activation or a complete failure of self-renewal pathways with the consequent initiation of stress-induced senescence. As such, A-type lamins should be regarded as intrinsic modulators of ageing within adult stem cells and their niches that are essential for survival to old age.
Collapse
Affiliation(s)
- Vanja Pekovic
- School of Biological and Biomedical Science, Integrated Cell Biology Laboratories, Durham University, South Road, Durham DH1 3LE, UK.
| | | |
Collapse
|
94
|
Shumaker DK, Solimando L, Sengupta K, Shimi T, Adam SA, Grunwald A, Strelkov SV, Aebi U, Cardoso MC, Goldman RD. The highly conserved nuclear lamin Ig-fold binds to PCNA: its role in DNA replication. ACTA ACUST UNITED AC 2008; 181:269-80. [PMID: 18426975 PMCID: PMC2315674 DOI: 10.1083/jcb.200708155] [Citation(s) in RCA: 94] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
This study provides insights into the role of nuclear lamins in DNA replication. Our data demonstrate that the Ig-fold motif located in the lamin C terminus binds directly to proliferating cell nuclear antigen (PCNA), the processivity factor necessary for the chain elongation phase of DNA replication. We find that the introduction of a mutation in the Ig-fold, which alters its structure and causes human muscular dystrophy, inhibits PCNA binding. Studies of nuclear assembly and DNA replication show that lamins, PCNA, and chromatin are closely associated in situ. Exposure of replicating nuclei to an excess of the lamin domain containing the Ig-fold inhibits DNA replication in a concentration-dependent fashion. This inhibitory effect is significantly diminished in nuclei exposed to the same domain bearing the Ig-fold mutation. Using the crystal structures of the lamin Ig-fold and PCNA, molecular docking simulations suggest probable interaction sites. These findings also provide insights into the mechanisms underlying the numerous disease-causing mutations located within the lamin Ig-fold.
Collapse
Affiliation(s)
- Dale K Shumaker
- Department of Cell and Molecular Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
95
|
The nuclear envelope as an integrator of nuclear and cytoplasmic architecture. FEBS Lett 2008; 582:2023-32. [PMID: 18474238 DOI: 10.1016/j.febslet.2008.05.001] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2008] [Revised: 05/02/2008] [Accepted: 05/05/2008] [Indexed: 12/11/2022]
Abstract
Initially perceived as little more than a container for the genome, our view of the nuclear envelope (NE) and its role in defining global nuclear architecture has evolved significantly in recent years. The recognition that certain human diseases arise from defects in NE components has provided new insight into its structural and regulatory functions. In particular, NE defects associated with striated muscle disease have been shown to cause structural perturbations not just of the nucleus itself but also of the cytoplasm. It is now becoming increasingly apparent that these two compartments display co-dependent mechanical properties. The identification of cytoskeletal binding complexes that localize to the NE now reveals a molecular framework that can seamlessly integrate nuclear and cytoplasmic architecture.
Collapse
|
96
|
Dechat T, Pfleghaar K, Sengupta K, Shimi T, Shumaker DK, Solimando L, Goldman RD. Nuclear lamins: major factors in the structural organization and function of the nucleus and chromatin. Genes Dev 2008; 22:832-53. [PMID: 18381888 PMCID: PMC2732390 DOI: 10.1101/gad.1652708] [Citation(s) in RCA: 746] [Impact Index Per Article: 43.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Over the past few years it has become evident that the intermediate filament proteins, the types A and B nuclear lamins, not only provide a structural framework for the nucleus, but are also essential for many aspects of normal nuclear function. Insights into lamin-related functions have been derived from studies of the remarkably large number of disease-causing mutations in the human lamin A gene. This review provides an up-to-date overview of the functions of nuclear lamins, emphasizing their roles in epigenetics, chromatin organization, DNA replication, transcription, and DNA repair. In addition, we discuss recent evidence supporting the importance of lamins in viral infections.
Collapse
Affiliation(s)
- Thomas Dechat
- Department of Cell and Molecular Biology, Northwestern University Medical School, Chicago, Illinois 60611, USA
| | - Katrin Pfleghaar
- Department of Cell and Molecular Biology, Northwestern University Medical School, Chicago, Illinois 60611, USA
| | - Kaushik Sengupta
- Department of Cell and Molecular Biology, Northwestern University Medical School, Chicago, Illinois 60611, USA
| | - Takeshi Shimi
- Department of Cell and Molecular Biology, Northwestern University Medical School, Chicago, Illinois 60611, USA
| | - Dale K. Shumaker
- Department of Cell and Molecular Biology, Northwestern University Medical School, Chicago, Illinois 60611, USA
| | - Liliana Solimando
- Department of Cell and Molecular Biology, Northwestern University Medical School, Chicago, Illinois 60611, USA
| | - Robert D. Goldman
- Department of Cell and Molecular Biology, Northwestern University Medical School, Chicago, Illinois 60611, USA
| |
Collapse
|
97
|
Håkelien AM, Delbarre E, Gaustad KG, Buendia B, Collas P. Expression of the myodystrophic R453W mutation of lamin A in C2C12 myoblasts causes promoter-specific and global epigenetic defects. Exp Cell Res 2008; 314:1869-80. [PMID: 18396274 DOI: 10.1016/j.yexcr.2008.02.018] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2008] [Revised: 02/22/2008] [Accepted: 02/26/2008] [Indexed: 10/22/2022]
Abstract
Autosomal dominant Emery-Dreifuss muscular dystrophy (EDMD) is characterized by muscle wasting and is caused by mutations in the LMNA gene encoding A-type lamins. Overexpression of the EDMD lamin A R453W mutation in C2C12 myoblasts impairs myogenic differentiation. We show here the influence of stable expression of the R453W and of the Dunnigan-type partial lipodystrophy R482W mutation of lamin A in C2C12 cells on transcription and epigenetic regulation of the myogenin (Myog) gene and on global chromatin organization. Expression of R453W-, but not R482W-lamin A, impairs activation of Myog and maintains a repressive chromatin state on the Myog promoter upon induction of differentiation, marked by H3 lysine (K) 9 dimethylation and failure to hypertrimethylate H3K4. Cells expressing WT-LaA also fail to hypertrimethylate H3K4. No defect occurs at the level of Myog promoter DNA methylation in any of the clones. Expression of R453W-lamin A and to a lesser extent R482W-lamin A in undifferentiated C2C12 cells redistributes H3K9me3 from pericentric heterochromatin. R453W-lamin A also elicits a redistribution of H3K27me3 from inactive X (Xi) and partial decondensation of Xi, but maintains Xist expression and coating of Xi, indicating that Xi remains inactivated. Our results argue that gene-specific and genome-wide chromatin rearrangements may constitute a molecular basis for laminopathies.
Collapse
Affiliation(s)
- Anne-Mari Håkelien
- Institute of Basic Medical Sciences, Department of Biochemistry, Faculty of Medicine, University of Oslo, PO Box 1112 Blindern, 0317 Oslo, Norway
| | | | | | | | | |
Collapse
|
98
|
Liu Y, Wang Y, Rusinol AE, Sinensky MS, Liu J, Shell SM, Zou Y. Involvement of xeroderma pigmentosum group A (XPA) in progeria arising from defective maturation of prelamin A. FASEB J 2008; 22:603-11. [PMID: 17848622 PMCID: PMC3116236 DOI: 10.1096/fj.07-8598com] [Citation(s) in RCA: 87] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Cellular accumulation of DNA damage has been widely implicated in cellular senescence, aging, and premature aging. In Hutchinson-Gilford progeria syndrome (HGPS) and restrictive dermopathy (RD), premature aging is linked to accumulation of DNA double-strand breaks (DSBs), which results in genome instability. However, how DSBs accumulate in cells despite the presence of intact DNA repair proteins remains unknown. Here we report that the recruitment of DSB repair factors Rad50 and Rad51 to the DSB sites, as marked by gamma-H2AX, was impaired in human HGPS and Zmpste24-deficient cells. Consistently, the progeria-associated DSBs appeared to be unrepairable although DSBs induced by camptothecin were efficiently removed in the progeroid cells. We also found that these progeroid cells exhibited nuclear foci of xeroderma pigmentosum group A (XPA), a unique nucleotide excision repair protein. Strikingly, these XPA foci colocalized with the DSB sites in the progeroid cells. This XPA-DSB association was further confirmed and found to be mediated by DNA, using a modified chromatin immunoprecipitation assay and coimmunoprecipitation. RNA interference (RNAi) knockdown of XPA in HGPS cells partially restored DSB repair as evidenced by Western blot analysis, immunofluorescence and comet assays. We propose that the uncharacteristic localization of XPA to or near DSBs inhibits DSB repair, thereby contributing to the premature aging phenotypes observed in progeria arising from genetic defects in prelamin A maturation.
Collapse
Affiliation(s)
- Yiyong Liu
- Department of Biochemistry and Molecular Biology, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee 37614
| | - Youjie Wang
- Department of Biochemistry and Molecular Biology, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee 37614
| | - Antonio E. Rusinol
- Department of Biochemistry and Molecular Biology, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee 37614
| | - Michael S. Sinensky
- Department of Biochemistry and Molecular Biology, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee 37614
| | - Ji Liu
- Department of Biochemistry and Molecular Biology, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee 37614
- Department of Biochemistry and Molecular Biology, Sichuan University, Chengdu, China
| | - Steven M. Shell
- Department of Biochemistry and Molecular Biology, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee 37614
| | - Yue Zou
- Department of Biochemistry and Molecular Biology, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee 37614
- Department of Biochemistry and Molecular Biology, Sichuan University, Chengdu, China
| |
Collapse
|
99
|
Parnaik VK. Role of Nuclear Lamins in Nuclear Organization, Cellular Signaling, and Inherited Diseases. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2008; 266:157-206. [DOI: 10.1016/s1937-6448(07)66004-3] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
100
|
Stewart CL, Roux KJ, Burke B. Blurring the Boundary: The Nuclear Envelope Extends Its Reach. Science 2007; 318:1408-12. [DOI: 10.1126/science.1142034] [Citation(s) in RCA: 208] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|