101
|
Butin-Israeli V, Adam SA, Goldman AE, Goldman RD. Nuclear lamin functions and disease. Trends Genet 2012; 28:464-71. [PMID: 22795640 DOI: 10.1016/j.tig.2012.06.001] [Citation(s) in RCA: 177] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2012] [Revised: 05/25/2012] [Accepted: 06/13/2012] [Indexed: 12/26/2022]
Abstract
Recent studies have shown that premature cellular senescence and normal organ development and function depend on the type V intermediate filament proteins, the lamins, which are major structural proteins of the nucleus. This review presents an up-to-date summary of the literature describing new findings on lamin functions in various cellular processes and emphasizes the relationship between the lamins and devastating diseases ranging from premature aging to cancer. Recent insights into the structure and function of the A- and B- type lamins in normal cells and their dysfunctions in diseased cells are providing novel targets for the development of new diagnostic procedures and disease intervention. We summarize these recent findings, focusing on data from mice and humans, and highlight the expanding knowledge of these proteins in both healthy and diseased cells.
Collapse
Affiliation(s)
- Veronika Butin-Israeli
- Department of Cell and Molecular Biology, Feinberg School of Medicine, Northwestern University, 303 East Chicago Avenue, Chicago, IL 60611, USA.
| | | | | | | |
Collapse
|
102
|
Khatau SB, Kusuma S, Hanjaya-Putra D, Mali P, Cheng L, Lee JSH, Gerecht S, Wirtz D. The differential formation of the LINC-mediated perinuclear actin cap in pluripotent and somatic cells. PLoS One 2012; 7:e36689. [PMID: 22574215 PMCID: PMC3344930 DOI: 10.1371/journal.pone.0036689] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2011] [Accepted: 04/12/2012] [Indexed: 12/22/2022] Open
Abstract
The actin filament cytoskeleton mediates cell motility and adhesion in somatic cells. However, whether the function and organization of the actin network are fundamentally different in pluripotent stem cells is unknown. Here we show that while conventional actin stress fibers at the basal surface of cells are present before and after onset of differentiation of mouse (mESCs) and human embryonic stem cells (hESCs), actin stress fibers of the actin cap, which wrap around the nucleus, are completely absent from undifferentiated mESCs and hESCs and their formation strongly correlates with differentiation. Similarly, the perinuclear actin cap is absent from human induced pluripotent stem cells (hiPSCs), while it is organized in the parental lung fibroblasts from which these hiPSCs are derived and in a wide range of human somatic cells, including lung, embryonic, and foreskin fibroblasts and endothelial cells. During differentiation, the formation of the actin cap follows the expression and proper localization of nuclear lamin A/C and associated linkers of nucleus and cytoskeleton (LINC) complexes at the nuclear envelope, which physically couple the actin cap to the apical surface of the nucleus. The differentiation of hESCs is accompanied by the progressive formation of a perinuclear actin cap while induced pluripotency is accompanied by the specific elimination of the actin cap, and that, through lamin A/C and LINC complexes, this actin cap is involved in progressively shaping the nucleus of hESCs undergoing differentiation. While, the localization of lamin A/C at the nuclear envelope is required for perinuclear actin cap formation, it is not sufficient to control nuclear shape.
Collapse
Affiliation(s)
- Shyam B. Khatau
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, Maryland, United States of America
- Johns Hopkins Physical Sciences – Oncology Center and Institute for NanoBioTechnology, The Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Sravanti Kusuma
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, Maryland, United States of America
- Johns Hopkins Physical Sciences – Oncology Center and Institute for NanoBioTechnology, The Johns Hopkins University, Baltimore, Maryland, United States of America
- Department of Biomedical Engineering, The Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Donny Hanjaya-Putra
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, Maryland, United States of America
- Johns Hopkins Physical Sciences – Oncology Center and Institute for NanoBioTechnology, The Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Prashant Mali
- Department of Biomedical Engineering, The Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Linzhao Cheng
- Institute for Cell Engineering and Department of Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland, United States of America
| | - Jerry S. H. Lee
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, Maryland, United States of America
- Center for Strategic Scientific Initiatives, Office of the Director, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Sharon Gerecht
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, Maryland, United States of America
- Johns Hopkins Physical Sciences – Oncology Center and Institute for NanoBioTechnology, The Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Denis Wirtz
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, Maryland, United States of America
- Johns Hopkins Physical Sciences – Oncology Center and Institute for NanoBioTechnology, The Johns Hopkins University, Baltimore, Maryland, United States of America
- * E-mail:
| |
Collapse
|
103
|
Kubben N, Voncken JW, Konings G, van Weeghel M, van den Hoogenhof MM, Gijbels M, van Erk A, Schoonderwoerd K, van den Bosch B, Dahlmans V, Calis C, Houten SM, Misteli T, Pinto YM. Post-natal myogenic and adipogenic developmental: defects and metabolic impairment upon loss of A-type lamins. Nucleus 2012; 2:195-207. [PMID: 21818413 DOI: 10.4161/nucl.2.3.15731] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2011] [Revised: 04/01/2011] [Accepted: 04/05/2011] [Indexed: 12/23/2022] Open
Abstract
A-type lamins are a major component of the nuclear lamina. Mutations in the LMNA gene, which encodes the A-type lamins A and C, cause a set of phenotypically diverse diseases collectively called laminopathies. While adult LMNA null mice show various symptoms typically associated with laminopathies, the effect of loss of lamin A/C on early post-natal development is poorly understood. Here we developed a novel LMNA null mouse (LMNA(GT-/-)) based on genetrap technology and analyzed its early post-natal development. We detect LMNA transcripts in heart, the outflow tract, dorsal aorta, liver and somites during early embryonic development. Loss of A-type lamins results in severe growth retardation and developmental defects of the heart, including impaired myocyte hypertrophy, skeletal muscle hypotrophy, decreased amounts of subcutaneous adipose tissue and impaired ex vivo adipogenic differentiation. These defects cause death at 2 to 3 weeks post partum associated with muscle weakness and metabolic complications, but without the occurrence of dilated cardiomyopathy or an obvious progeroid phenotype. Our results indicate that defective early post-natal development critically contributes to the disease phenotypes in adult laminopathies.
Collapse
Affiliation(s)
- Nard Kubben
- Heart Failure Research Center and Department of Cardiology, Maastricht University Medical Centre, The Netherlands
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
104
|
Abstract
Over the past two decades, the biomechanical properties of cells have emerged as key players in a broad range of cellular functions, including migration, proliferation, and differentiation. Although much of the attention has focused on the cytoskeletal networks and the cell's microenvironment, relatively little is known about the contribution of the cell nucleus. Here, we present an overview of the structural elements that determine the physical properties of the nucleus and discuss how changes in the expression of nuclear components or mutations in nuclear proteins can not only affect nuclear mechanics but also modulate cytoskeletal organization and diverse cellular functions. These findings illustrate that the nucleus is tightly integrated into the surrounding cellular structure. Consequently, changes in nuclear structure and composition are highly relevant to normal development and physiology and can contribute to many human diseases, such as muscular dystrophy, dilated cardiomyopathy, (premature) aging, and cancer.
Collapse
Affiliation(s)
- Monika Zwerger
- Department of Medicine, Brigham and Women's Hospital/Harvard Medical School, Boston, MA 02115, USA.
| | | | | |
Collapse
|
105
|
Abstract
Hutchinson-Gilford Progeria (HGPS) and Werner syndromes are diseases that clinically resemble some aspects of accelerated aging. HGPS is caused by mutations in theLMNA gene resulting in post-translational processing defects that trigger Progeria in children. Werner syndrome, arising from mutations in the WRN helicase gene, causes premature aging in young adults. What are the molecular mechanism(s) underlying these disorders and what aspects of the diseases resemble physiological human aging? Much of what we know stems from the study of patient derived fibroblasts with both mutations resulting in increased DNA damage, primarily at telomeres. However, in vivo patients with Werner's develop arteriosclerosis, among other pathologies. In HGPS patients, including iPS derived cells from HGPS patients, as well as some mouse models for Progeria, vascular smooth muscle (VSM) appears to be among the most severely affected tissues. Defective Lamin processing, associated with DNA damage, is present in VSM from old individuals, indicating processing defects may be a factor in normal aging. Whether persistent DNA damage, particularly at telomeres, is the root cause for these pathologies remains to be established, since not all progeroid Lmna mutations result in DNA damage and genome instability.
Collapse
|
106
|
Inner nuclear membrane proteins: impact on human disease. Chromosoma 2012; 121:153-67. [DOI: 10.1007/s00412-012-0360-2] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2011] [Revised: 01/02/2012] [Accepted: 01/03/2012] [Indexed: 02/01/2023]
|
107
|
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
|
108
|
Kim Y, Sharov AA, McDole K, Cheng M, Hao H, Fan CM, Gaiano N, Ko MSH, Zheng Y. Mouse B-type lamins are required for proper organogenesis but not by embryonic stem cells. Science 2011; 334:1706-10. [PMID: 22116031 PMCID: PMC3306219 DOI: 10.1126/science.1211222] [Citation(s) in RCA: 202] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
B-type lamins, the major components of the nuclear lamina, are believed to be essential for cell proliferation and survival. We found that mouse embryonic stem cells (ESCs) do not need any lamins for self-renewal and pluripotency. Although genome-wide lamin-B binding profiles correlate with reduced gene expression, such binding is not directly required for gene silencing in ESCs or trophectoderm cells. However, B-type lamins are required for proper organogenesis. Defects in spindle orientation in neural progenitor cells and migration of neurons probably cause brain disorganizations found in lamin-B null mice. Thus, our studies not only disprove several prevailing views of lamin-Bs but also establish a foundation for redefining the function of the nuclear lamina in the context of tissue building and homeostasis.
Collapse
Affiliation(s)
- Youngjo Kim
- Department of Embryology, Carnegie Institution for Science, Baltimore, MD 21218, USA
- Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
| | - Alexei A. Sharov
- Developmental Genomics and Aging Section, Laboratory of Genetics, National Institute on Aging, NIH, Baltimore, MD 21224, USA
| | - Katie McDole
- Department of Embryology, Carnegie Institution for Science, Baltimore, MD 21218, USA
- Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
- Department of Biology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Melody Cheng
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Haiping Hao
- Microarray Core Facility, Johns Hopkins University School of Medicine, Baltimore, MD 21209, USA
| | - Chen-Ming Fan
- Department of Embryology, Carnegie Institution for Science, Baltimore, MD 21218, USA
- Department of Biology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Nicholas Gaiano
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Minoru S. H. Ko
- Developmental Genomics and Aging Section, Laboratory of Genetics, National Institute on Aging, NIH, Baltimore, MD 21224, USA
| | - Yixian Zheng
- Department of Embryology, Carnegie Institution for Science, Baltimore, MD 21218, USA
- Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
- Department of Biology, Johns Hopkins University, Baltimore, MD 21218, USA
| |
Collapse
|
109
|
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
|
110
|
Abstract
The B-type lamins are widely assumed to be essential for mammalian cells. In part, this assumption is based on a highly cited study that found that RNAi-mediated knockdown of lamin B1 or lamin B2 in HeLa cells arrested cell growth and led to apoptosis. Studies indicating that B-type lamins play roles in DNA replication, the formation of the mitotic spindle, chromatin organization and regulation of gene expression have fueled the notion that B-type lamins must be essential. But surprisingly, this idea had never been tested with genetic approaches. Earlier this year, a research group from UCLA reported the development of genetically modified mice that lack expression of both Lmnb1 and Lmnb2 in skin keratinocytes (a cell type that proliferates rapidly and participates in complex developmental programs). They reasoned that if lamins B1 and B2 were truly essential, then keratinocyte-specific lamin B1/lamin B2 knockout mice would exhibit severe pathology. Contrary to expectations, the skin and hair of lamin B1/lamin B2-deficient mice were quite normal, indicating that the B-type lamins are dispensable in some cell types. The same UCLA research group has gone on to show that lamin B1 and lamin B2 are critical for neuronal migration in the developing brain and for neuronal survival. The absence of either lamin B1 or lamin B2, or the absence of both B-type lamins, results in severe neurodevelopmental abnormalities.
Collapse
Affiliation(s)
- Shao H. Yang
- Department of Medicine; David Geffen School of Medicine; University of California; Los Angeles, CA USA
| | - Hea-Jin Jung
- Molecular Biology Institute; University of California; Los Angeles, CA USA
| | - Catherine Coffinier
- Department of Medicine; David Geffen School of Medicine; University of California; Los Angeles, CA USA
| | - Loren G. Fong
- Department of Medicine; David Geffen School of Medicine; University of California; Los Angeles, CA USA
| | - Stephen G. Young
- Department of Medicine; David Geffen School of Medicine; University of California; Los Angeles, CA USA
- Molecular Biology Institute; University of California; Los Angeles, CA USA
| |
Collapse
|
111
|
Moiseeva O, Bourdeau V, Vernier M, Dabauvalle MC, Ferbeyre G. Retinoblastoma-independent regulation of cell proliferation and senescence by the p53-p21 axis in lamin A /C-depleted cells. Aging Cell 2011; 10:789-97. [PMID: 21535365 DOI: 10.1111/j.1474-9726.2011.00719.x] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The expression of A-type lamin is downregulated in several cancers, and lamin defects are the cause of several diseases including a form of accelerated aging. We report that depletion of lamin A/C expression in normal human cells leads to a dramatic downregulation of the Rb family of tumor suppressors and a defect in cell proliferation. Lamin A/C-depleted cells exhibited a flat morphology and accumulated markers of cellular senescence. This senescent phenotype was accompanied by engagement of the p53 tumor suppressor and induction of the p53 target gene p21 and was prevented by small hairpin RNAs against p53, p21, or by the oncoprotein Mdm2. The expression of E2F target genes, normally required for cell cycle progression, was downregulated after lamin A/C depletion but restored after the inactivation of p53. A similar senescence response was observed in myoblasts from a patient with a lamin A mutation causing muscular dystrophy. We thus reveal a previously unnoticed mechanism of controlling cell cycle genes expression, which depends on p53 but does not require the retinoblastoma family of tumor suppressors and that can be relevant to understand the pathogenesis of laminopathies and perhaps aging.
Collapse
Affiliation(s)
- Olga Moiseeva
- Biochemistry Department, Université de Montréal, Montréal, QC, Canada
| | | | | | | | | |
Collapse
|
112
|
Hasebe T, Kajita M, Iwabuchi M, Ohsumi K, Ishizuya-Oka A. Thyroid hormone-regulated expression of nuclear lamins correlates with dedifferentiation of intestinal epithelial cells during Xenopus laevis metamorphosis. Dev Genes Evol 2011; 221:199-208. [PMID: 21866414 DOI: 10.1007/s00427-011-0371-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2011] [Accepted: 07/04/2011] [Indexed: 11/26/2022]
Abstract
In the Xenopus laevis intestine during metamorphosis, which is triggered by thyroid hormone (TH), the adult epithelium develops and replaces the larval one undergoing apoptosis. We have previously shown that progenitor/stem cells of the adult epithelium originate from some differentiated larval epithelial cells. To investigate molecular mechanisms underlying larval epithelial dedifferentiation into the adult progenitor/stem cells, we here focused on nuclear lamin A (LA) and lamin LIII (LIII), whose expression is generally known to be correlated with the state of cell differentiation. We analyzed the spatiotemporal expression of LA and LIII during X. laevis intestinal remodeling by reverse transcription PCR, Western blotting, and immunohistochemistry. At the onset of natural metamorphosis, when the adult epithelial progenitor cells appear as small islets, the expression of LA is down-regulated, but that of LIII is up-regulated only in the islets. Then, as the adult progenitor cells differentiate, the expression of LA is up-regulated, whereas that of LIII is down-regulated in the adult cells. As multiple intestinal folds form, adult epithelial cells positive for LIII become restricted only to the troughs of the folds. In addition, we have shown that TH up- or down-regulates the expression of these lamins in the premetamorphic intestine as during natural metamorphosis. These results indicate that TH-regulated expression of LA and LIII closely correlates with dedifferentiation of the epithelial cells in the X. laevis intestine, suggesting the involvement of the lamins in the process of dedifferentiation during amphibian metamorphosis.
Collapse
Affiliation(s)
- Takashi Hasebe
- Department of Biology, Nippon Medical School, 2-297-2 Kosugi-cho, Nakahara-ku, Kawasaki, Kanagawa 211-0063, Japan
| | | | | | | | | |
Collapse
|
113
|
Yang SH, Chang SY, Yin L, Tu Y, Hu Y, Yoshinaga Y, de Jong PJ, Fong LG, Young SG. An absence of both lamin B1 and lamin B2 in keratinocytes has no effect on cell proliferation or the development of skin and hair. Hum Mol Genet 2011; 20:3537-44. [PMID: 21659336 PMCID: PMC3159554 DOI: 10.1093/hmg/ddr266] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2011] [Revised: 05/24/2011] [Accepted: 06/06/2011] [Indexed: 11/14/2022] Open
Abstract
Nuclear lamins are usually classified as A-type (lamins A and C) or B-type (lamins B1 and B2). A-type lamins have been implicated in multiple genetic diseases but are not required for cell growth or development. In contrast, B-type lamins have been considered essential in eukaryotic cells, with crucial roles in DNA replication and in the formation of the mitotic spindle. Knocking down the genes for B-type lamins (LMNB1, LMNB2) in HeLa cells has been reported to cause apoptosis. In the current study, we created conditional knockout alleles for mouse Lmnb1 and Lmnb2, with the goal of testing the hypothesis that B-type lamins are crucial for the growth and viability of mammalian cells in vivo. Using the keratin 14-Cre transgene, we bred mice lacking the expression of both Lmnb1 and Lmnb2 in skin keratinocytes (Lmnb1(Δ/Δ)Lmnb2(Δ/Δ)). Lmnb1 and Lmnb2 transcripts were absent in keratinocytes of Lmnb1(Δ/Δ)Lmnb2(Δ/Δ) mice, and lamin B1 and lamin B2 proteins were undetectable. But despite an absence of B-type lamins in keratinocytes, the skin and hair of Lmnb1(Δ/Δ)Lmnb2(Δ/Δ) mice developed normally and were free of histological abnormalities, even in 2-year-old mice. After an intraperitoneal injection of bromodeoxyuridine (BrdU), similar numbers of BrdU-positive keratinocytes were observed in the skin of wild-type and Lmnb1(Δ/Δ)Lmnb2(Δ/Δ) mice. Lmnb1(Δ/Δ)Lmnb2(Δ/Δ) keratinocytes did not exhibit aneuploidy, and their growth rate was normal in culture. These studies challenge the concept that B-type lamins are essential for proliferation and vitality of eukaryotic cells.
Collapse
Affiliation(s)
- Shao H. Yang
- Department of Medicine, Division of Cardiology and
| | | | - Liya Yin
- Department of Medicine, Division of Cardiology and
| | - Yiping Tu
- Department of Medicine, Division of Cardiology and
| | - Yan Hu
- Department of Medicine, Division of Cardiology and
| | - Yuko Yoshinaga
- Children's Hospital Oakland Research Institute, Oakland, CA 94609, USA
| | - Pieter J. de Jong
- Children's Hospital Oakland Research Institute, Oakland, CA 94609, USA
| | | | - Stephen G. Young
- Department of Medicine, Division of Cardiology and
- Department of Human Genetics, University of California, Los Angeles, CA 90095, USA and
| |
Collapse
|
114
|
Pillarisetti A, Desai JP, Ladjal H, Schiffmacher A, Ferreira A, Keefer CL. Mechanical phenotyping of mouse embryonic stem cells: increase in stiffness with differentiation. Cell Reprogram 2011; 13:371-80. [PMID: 21728815 DOI: 10.1089/cell.2011.0028] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Atomic force microscopy (AFM) has emerged as a promising tool to characterize the mechanical properties of biological materials and cells. In our studies, undifferentiated and early differentiating mouse embryonic stem cells (mESCs) were assessed individually using an AFM system to determine if we could detect changes in their mechanical properties by surface probing. Probes with pyramidal and spherical tips were assessed, as were different analytical models for evaluating the data. The combination of AFM probing with a spherical tip and analysis using the Hertz model provided the best fit to the experimental data obtained and thus provided the best approximation of the elastic modulus. Our results showed that after only 6 days of differentiation, individual cell stiffness increased significantly with early differentiating mESCs having an elastic modulus two- to threefold higher than undifferentiated mESCs, regardless of cell line (R1 or D3 mESCs) or treatment. Single-touch (indentation) probing of individual cells is minimally invasive compared to other techniques. Therefore, this method of mechanical phenotyping should prove to be a valuable tool in the development of improved methods of identification and targeted cellular differentiation of embryonic, adult, and induced-pluripotent stem cells for therapeutic and diagnostic purposes.
Collapse
Affiliation(s)
- Anand Pillarisetti
- Robotics, Automation, Medical Systems (RAMS) Laboratory, University of Maryland, College Park, Maryland, USA
| | | | | | | | | | | |
Collapse
|
115
|
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
|
116
|
Abstract
The nuclear lamins are type V intermediate filament proteins that are critically important for the structural properties of the nucleus. In addition, they are involved in the regulation of numerous nuclear processes, including DNA replication, transcription and chromatin organization. The developmentally regulated expression of lamins suggests that they are involved in cellular differentiation. Their assembly dynamic properties throughout the cell cycle, particularly in mitosis, are influenced by posttranslational modifications. Lamins may regulate nuclear functions by direct interactions with chromatin and determining the spatial organization of chromosomes within the nuclear space. They may also regulate chromatin functions by interacting with factors that epigenetically modify the chromatin or directly regulate replication or transcription.
Collapse
Affiliation(s)
- Thomas Dechat
- Department of Cell and Molecular Biology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611, USA
| | | | | | | | | |
Collapse
|
117
|
Hernandez L, Roux KJ, Wong ESM, Mounkes LC, Mutalif R, Navasankari R, Rai B, Cool S, Jeong JW, Wang H, Lee HS, Kozlov S, Grunert M, Keeble T, Jones CM, Meta MD, Young SG, Daar IO, Burke B, Perantoni AO, Stewart CL. Functional coupling between the extracellular matrix and nuclear lamina by Wnt signaling in progeria. Dev Cell 2010; 19:413-25. [PMID: 20833363 PMCID: PMC2953243 DOI: 10.1016/j.devcel.2010.08.013] [Citation(s) in RCA: 141] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2009] [Revised: 07/07/2010] [Accepted: 08/23/2010] [Indexed: 12/14/2022]
Abstract
The segmental premature aging disease Hutchinson-Gilford Progeria (HGPS) is caused by a truncated and farnesylated form of Lamin A. In a mouse model for HGPS, a similar Lamin A variant causes the proliferative arrest and death of postnatal, but not embryonic, fibroblasts. Arrest is due to an inability to produce a functional extracellular matrix (ECM), because growth on normal ECM rescues proliferation. The defects are associated with inhibition of canonical Wnt signaling, due to reduced nuclear localization and transcriptional activity of Lef1, but not Tcf4, in both mouse and human progeric cells. Defective Wnt signaling, affecting ECM synthesis, may be critical to the etiology of HGPS because mice exhibit skeletal defects and apoptosis in major blood vessels proximal to the heart. These results establish a functional link between the nuclear envelope/lamina and the cell surface/ECM and may provide insights into the role of Wnt signaling and the ECM in aging.
Collapse
Affiliation(s)
- Lidia Hernandez
- Cancer and Developmental Biology Laboratory, NCI, Frederick, MD 21702
- Molecular Signaling Section, Medical Oncology Branch, Center for Cancer Research NCI, Bethesda, MD 20892
| | - Kyle J. Roux
- Dept. of Anatomy and Cell Biology, University of Florida College of Medicine, Gainesville, FL 32606
| | | | - Leslie C. Mounkes
- Cancer and Developmental Biology Laboratory, NCI, Frederick, MD 21702
| | - Rafidah Mutalif
- Institute of Medical Biology, Immunos, 8A Biomedical Grove, Singapore 138648
| | - Raju Navasankari
- Institute of Medical Biology, Immunos, 8A Biomedical Grove, Singapore 138648
| | - Bina Rai
- Institute of Medical Biology, Immunos, 8A Biomedical Grove, Singapore 138648
| | - Simon Cool
- Institute of Medical Biology, Immunos, 8A Biomedical Grove, Singapore 138648
| | - Jae-Wook Jeong
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030
| | - Honghe Wang
- Cancer and Developmental Biology Laboratory, NCI, Frederick, MD 21702
| | - Hyun-Shik Lee
- Laboratory of Cell and Developmental Signaling, NCI, Frederick, MD 21702
| | - Serguei Kozlov
- Cancer and Developmental Biology Laboratory, NCI, Frederick, MD 21702
| | - Martin Grunert
- Institute of Medical Biology, Immunos, 8A Biomedical Grove, Singapore 138648
| | - Thomas Keeble
- Institute of Medical Biology, Immunos, 8A Biomedical Grove, Singapore 138648
| | - C. Michael Jones
- Institute of Medical Biology, Immunos, 8A Biomedical Grove, Singapore 138648
| | - Margarita D. Meta
- Dept. of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA90095
| | - Stephen G. Young
- Dept. of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA90095
| | - Ira O. Daar
- Laboratory of Cell and Developmental Signaling, NCI, Frederick, MD 21702
| | - Brian Burke
- Dept. of Anatomy and Cell Biology, University of Florida College of Medicine, Gainesville, FL 32606
| | - Alan O. Perantoni
- Cancer and Developmental Biology Laboratory, NCI, Frederick, MD 21702
| | - Colin L. Stewart
- Cancer and Developmental Biology Laboratory, NCI, Frederick, MD 21702
- Institute of Medical Biology, Immunos, 8A Biomedical Grove, Singapore 138648
| |
Collapse
|
118
|
Kelly RDW, Alberio R, Campbell KHS. A-type lamin dynamics in bovine somatic cell nuclear transfer embryos. Reprod Fertil Dev 2010; 22:956-65. [PMID: 20591330 DOI: 10.1071/rd09264] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2009] [Accepted: 01/20/2010] [Indexed: 01/29/2023] Open
Abstract
The persistence of A-type nuclear lamin in somatic cell nuclear transfer (SCNT) embryos has been proposed as a marker for incomplete nuclear reprogramming. Using monoclonal antibodies to A/C- (A/C-346 and A/C-131C3) and B-type lamin, we compared distribution during early development of bovine IVF, parthenogenetic and SCNT embryos. A/C-346 staining was observed in the pronuclei of IVF embryos and in nuclei at the two-cell stage, but was not detected in subsequent cleavage stages up to and including hatched blastocysts. In contrast, A/C-131C3 and anti-lamin B2 stained all preimplantation stage embryos. Parthenogenetic and SCNT embryos had similar staining patterns to IVF embryos for all three antibodies, demonstrating correct nuclear architecture reprogramming. Inhibiting protein synthesis with cycloheximide (CHX) in parthenogenetic and SCNT embryos did not affect lamin A/C localisation, suggesting that lamin A/C is maternal in origin. However, activation with CHX delayed lamin A/C incorporation compared with 6-dimethylaminopurine activation. In SCNT embryos, staining for both A/C- and B-type lamin was delayed compared with parthenotes, although lamin B2 incorporation preceded lamin A/C in both. In conclusion, the lamin A/C distribution in SCNT bovine embryos paralleled that of IVF and parthenogenetic controls and therefore is not a marker of incomplete reprogramming.
Collapse
Affiliation(s)
- Richard D W Kelly
- Animal Development and Biotechnology Group, School of Biosciences, University of Nottingham, Sutton Bonington Campus, Loughborough, Leicestershire, UK
| | | | | |
Collapse
|
119
|
Odgren PR, Pratt CH, MacKay CA, Mason-Savas A, Curtain M, Shopland L, Ichicki T, Sundberg JP, Donahue LR. Disheveled hair and ear (Dhe), a spontaneous mouse Lmna mutation modeling human laminopathies. PLoS One 2010; 5:e9959. [PMID: 20376364 PMCID: PMC2848607 DOI: 10.1371/journal.pone.0009959] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2009] [Accepted: 03/11/2010] [Indexed: 12/23/2022] Open
Abstract
Background Investigations of naturally-occurring mutations in animal models provide important insights and valuable disease models. Lamins A and C, along with lamin B, are type V intermediate filament proteins which constitute the proteinaceous boundary of the nucleus. LMNA mutations in humans cause a wide range of phenotypes, collectively termed laminopathies. To identify the mutation and investigate the phenotype of a spontaneous, semi-dominant mutation that we have named Disheveled hair and ear (Dhe), which causes a sparse coat and small external ears in heterozygotes and lethality in homozygotes by postnatal day 10. Findings Genetic mapping identified a point mutation in the Lmna gene, causing a single amino acid change, L52R, in the coiled coil rod domain of lamin A and C proteins. Cranial sutures in Dhe/+ mice failed to close. Gene expression for collagen types I and III in sutures was deficient. Skulls were small and disproportionate. Skeletons of Dhe/+ mice were hypomineralized and total body fat was deficient in males. In homozygotes, skin and oral mucosae were dysplastic and ulcerated. Nuclear morphometry of cultured cells revealed gene dose-dependent blebbing and wrinkling. Conclusion Dhe mice should provide a useful new model for investigations of the pathogenesis of laminopathies.
Collapse
Affiliation(s)
- Paul R. Odgren
- Department of Cell Biology, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Craig H. Pratt
- Institute for Molecular Biophysics, Bar Harbor, Maine, United States of America
- The Jackson Laboratory, Bar Harbor, Maine, United States of America
| | - Carole A. MacKay
- Department of Cell Biology, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - April Mason-Savas
- Department of Cell Biology, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Michelle Curtain
- Genetic Resource Science, Bar Harbor, Maine, United States of America
- The Jackson Laboratory, Bar Harbor, Maine, United States of America
| | - Lindsay Shopland
- Institute for Molecular Biophysics, Bar Harbor, Maine, United States of America
- The Jackson Laboratory, Bar Harbor, Maine, United States of America
| | - Tsutomu Ichicki
- The Jackson Laboratory, Bar Harbor, Maine, United States of America
| | - John P. Sundberg
- The Jackson Laboratory, Bar Harbor, Maine, United States of America
| | - Leah Rae Donahue
- Genetic Resource Science, Bar Harbor, Maine, United States of America
- The Jackson Laboratory, Bar Harbor, Maine, United States of America
- * E-mail:
| |
Collapse
|
120
|
Jahn D, Schramm S, Benavente R, Alsheimer M. Dynamic properties of meiosis-specific lamin C2 and its impact on nuclear envelope integrity. Nucleus 2010; 1:273-83. [PMID: 21327075 DOI: 10.4161/nucl.1.3.11800] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2010] [Revised: 03/12/2010] [Accepted: 03/15/2010] [Indexed: 11/19/2022] Open
Abstract
A hallmark of meiosis is the precise pairing and the stable physical connection (synapsis) of the homologous chromosomes. These processes are essential prerequisite for their proper segregation. Pairing of the homologs during meiotic prophase I critically depends on characteristic movements of chromosomes. These movements, in turn, require attachment of meiotic telomeres to the nuclear envelope and their subsequent dynamic repositioning. Dynamic repositioning of meiotic telomeres goes along with profound structural reorganization of the nuclear envelope. The short A-type lamin C2 is thought to play a critical role in this process due to its specific expression during meiotic prophase I and the unique localization surrounding telomere attachments. Consistent with this notion, here we provide compelling evidence that meiosis-specific lamin C2 features a significantly increased mobility compared to somatic lamins as revealed by photobleaching techniques. We show that this property can be clearly ascribed to the lack of the N-terminal head and the significantly shorter α-helical coil domain. Moreover, expression of lamin C2 in somatic cells induces nuclear deformations and alters the distribution of the endogenous nuclear envelope proteins lamin B1, LAP2, SUN1 and SUN2. Together, our data define lamin C2 as a "natural lamin deletion mutant" that confers unique properties to the nuclear envelope which would be essential for dynamic telomere repositioning during meiotic prophase I.
Collapse
Affiliation(s)
- Daniel Jahn
- Department of Cell and Developmental Biology, Biocenter, University of Würzburg, Germany
| | | | | | | |
Collapse
|
121
|
Lombardi ML, Lammerding J. Altered Mechanical Properties of the Nucleus in Disease. Methods Cell Biol 2010; 98:121-41. [DOI: 10.1016/s0091-679x(10)98006-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
|
122
|
Dauer WT, Worman HJ. The nuclear envelope as a signaling node in development and disease. Dev Cell 2009; 17:626-38. [PMID: 19922868 DOI: 10.1016/j.devcel.2009.10.016] [Citation(s) in RCA: 185] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The development of a membrane-bound structure separating DNA from other cellular components was the epochal evolutionary event that gave rise to eukaryotes, possibly occurring up to 2 billion years ago. Yet, this view of the nuclear envelope as a physical barrier greatly underestimates its fundamental impact on cellular organization and complexity, much of which is only beginning to be understood. Indeed, alterations of nuclear envelope structure and protein composition are essential to many aspects of metazoan development and cellular differentiation. Mutations in genes encoding nuclear envelope proteins cause a fascinating array of diseases referred to as "nuclear envelopathies" or "laminopathies" that affect different tissues and organ systems. We review recent work on the nuclear envelope, including insights derived from the study of nuclear envelopathies. These studies are uncovering new functions for nuclear envelope proteins and underlie an emerging view of the nuclear envelope as a critical signaling node in development and disease.
Collapse
Affiliation(s)
- William T Dauer
- Department of Neurology, University of Michigan Medical School, Ann Arbor, MI 48109 USA.
| | | |
Collapse
|
123
|
Epigenetic regulatory mechanisms during preimplantation development. ACTA ACUST UNITED AC 2009; 87:297-313. [DOI: 10.1002/bdrc.20165] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
124
|
Peter A, Stick R. Ectopic expression of prelamin A in early Xenopus embryos induces apoptosis. Eur J Cell Biol 2009; 87:879-91. [PMID: 18675490 DOI: 10.1016/j.ejcb.2008.06.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2008] [Revised: 05/23/2008] [Accepted: 06/11/2008] [Indexed: 01/24/2023] Open
Abstract
Lamin proteins are components of metazoan cell nuclei. During evolution, two classes of lamin proteins evolved, A- and B-type lamins. B-type lamins are expressed in nearly all cell types and in all developmental stages and are thought to be indispensable for cellular survival. In contrast, A-type lamins have a more restricted expression pattern. They are expressed in differentiated cells and appear late in embryogenesis. In the earliest steps of mammalian development, A-type lamins are present in oocytes, pronuclei and during the first cleavage stages of the developing embryo. But latest after the 16-cell stage, A-type lamin proteins are not any longer detectable in embryonic cells. Amphibian oocytes and early embryos do not express lamin A. Moreover, extracts of Xenopus oocytes and eggs have the ability to selectively remove A-type lamins from somatic nuclei. This observation and the restricted expression pattern suggest that the presence of lamin A might interfere with developmental processes in the early phase of embryogenesis. To test this, we ectopically expressed lamin A during early embryonic development of Xenopus laevis by microinjection of synthetic mRNA. Here, we show that introducing mature lamin A does not interfere with normal development. However, expression of prelamin A or lamin A variants that cannot be fully processed cause severe disturbances and lead to apoptosis during gastrulation. The toxic effect is due to lack of the conversion of prenylated prelamin A to its mature form. Remarkably, even a cytoplasmic prelamin A variant that is excluded from the nucleus drives embryos into apoptosis.
Collapse
Affiliation(s)
- Annette Peter
- Department of Cell Biology, University of Bremen, P.O. Box 33 04 40, D-28334 Bremen, Germany
| | | |
Collapse
|
125
|
Worman HJ, Fong LG, Muchir A, Young SG. Laminopathies and the long strange trip from basic cell biology to therapy. J Clin Invest 2009; 119:1825-36. [PMID: 19587457 PMCID: PMC2701866 DOI: 10.1172/jci37679] [Citation(s) in RCA: 208] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The main function of the nuclear lamina, an intermediate filament meshwork lying primarily beneath the inner nuclear membrane, is to provide structural scaffolding for the cell nucleus. However, the lamina also serves other functions, such as having a role in chromatin organization, connecting the nucleus to the cytoplasm, gene transcription, and mitosis. In somatic cells, the main protein constituents of the nuclear lamina are lamins A, C, B1, and B2. Interest in the nuclear lamins increased dramatically in recent years with the realization that mutations in LMNA, the gene encoding lamins A and C, cause a panoply of human diseases ("laminopathies"), including muscular dystrophy, cardiomyopathy, partial lipodystrophy, and progeroid syndromes. Here, we review the laminopathies and the long strange trip from basic cell biology to therapeutic approaches for these diseases.
Collapse
Affiliation(s)
- Howard J. Worman
- Department of Medicine and
Department of Pathology and Cell Biology, Columbia University College of Physicians and Surgeons, New York, New York, USA.
Department of Medicine and
Department of Human Genetics, UCLA David Geffen School of Medicine, Los Angeles, California, USA
| | - Loren G. Fong
- Department of Medicine and
Department of Pathology and Cell Biology, Columbia University College of Physicians and Surgeons, New York, New York, USA.
Department of Medicine and
Department of Human Genetics, UCLA David Geffen School of Medicine, Los Angeles, California, USA
| | - Antoine Muchir
- Department of Medicine and
Department of Pathology and Cell Biology, Columbia University College of Physicians and Surgeons, New York, New York, USA.
Department of Medicine and
Department of Human Genetics, UCLA David Geffen School of Medicine, Los Angeles, California, USA
| | - Stephen G. Young
- Department of Medicine and
Department of Pathology and Cell Biology, Columbia University College of Physicians and Surgeons, New York, New York, USA.
Department of Medicine and
Department of Human Genetics, UCLA David Geffen School of Medicine, Los Angeles, California, USA
| |
Collapse
|
126
|
Martin C, Chen S, Maya-Mendoza A, Lovric J, Sims PFG, Jackson DA. Lamin B1 maintains the functional plasticity of nucleoli. J Cell Sci 2009; 122:1551-62. [PMID: 19383719 PMCID: PMC2722682 DOI: 10.1242/jcs.046284] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/26/2009] [Indexed: 01/14/2023] Open
Abstract
The dynamic ability of genomes to interact with discrete nuclear compartments appears to be essential for chromatin function. However, the extent to which structural nuclear proteins contribute to this level of organization is largely unresolved. To test the links between structure and function, we evaluated how nuclear lamins contribute to the organization of a major functional compartment, the nucleolus. HeLa cells with compromised expression of the genes encoding lamins were analyzed using high-resolution imaging and pull-down assays. When lamin B1 expression was depleted, inhibition of RNA synthesis correlated with complex structural changes within the nucleolar active centers until, eventually, the nucleoli were dispersed completely. With normal lamin expression, the nucleoli were highly plastic, with dramatic and freely reversible structural changes correlating with the demand for ribosome biogenesis. Preservation of the nucleolar compartment throughout these structural transitions is shown to be linked to lamin B1 expression, with the lamin B1 protein interacting with the major nucleolar protein nucleophosmin/B23.
Collapse
Affiliation(s)
- Catherine Martin
- Faculty of Life Sciences, University of Manchester, MIB, 131 Princess Street, Manchester M1 7DN, UK
| | | | | | | | | | | |
Collapse
|
127
|
Lee DC, Welton KL, Smith ED, Kennedy BK. A-type nuclear lamins act as transcriptional repressors when targeted to promoters. Exp Cell Res 2009; 315:996-1007. [PMID: 19272320 PMCID: PMC2746445 DOI: 10.1016/j.yexcr.2009.01.003] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2008] [Revised: 11/27/2008] [Accepted: 01/07/2009] [Indexed: 11/17/2022]
Abstract
Regions of heterochromatin are often found at the periphery of the mammalian nucleus, juxtaposed to the nuclear lamina. Genes in these regions are likely maintained in a transcriptionally silent state, although other locations at the nuclear periphery associated with nuclear pores are sites of active transcription. As primary components of the nuclear lamina, A- and B-type nuclear lamins are intermediate filament proteins that interact with DNA, histones and known transcriptional repressors, leading to speculation that they may promote establishment of repressive domains. However, no direct evidence of a role for nuclear lamins in transcriptional repression has been reported. Here we find that human lamin A, when expressed in yeast and cultured human cells as a fusion protein to the Gal4 DNA-binding domain (DBD), can mediate robust transcriptional repression of promoters with Gal4 binding sites. Full repression by lamin A requires both the coiled-coil rod domain and the C-terminal tail domain. In human cells, other intermediate filament proteins such as lamin B and vimentin are unable to confer robust repression as Gal4-DBD fusions, indicating that this property is specific to A-type nuclear lamins. These findings indicate that A-type lamins can promote transcriptional repression when in proximity of a promoter.
Collapse
Affiliation(s)
- Damian C Lee
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | | | | | | |
Collapse
|
128
|
Abstract
Recent findings that some 24 inherited diseases and anomalies are caused by defects in proteins of the NE (nuclear envelope) and lamina have resulted in a fundamental reassessment of the functions of the NE and underlying lamina. Instead of just regarding the NE and lamina as a molecular filtering device, regulating the transfer of macromolecules between the cytoplasm and nucleus, we now envisage the NE/lamina functioning as a key cellular 'hub' in integrating critical functions that include chromatin organization, transcriptional regulation, mechanical integrity of the cell and signalling pathways, as well as acting as a key component in the organization and function of the cytoskeleton.
Collapse
|
129
|
Wu Z, Wu L, Weng D, Xu D, Geng J, Zhao F. Reduced expression of lamin A/C correlates with poor histological differentiation and prognosis in primary gastric carcinoma. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2009; 28:8. [PMID: 19144202 PMCID: PMC2632624 DOI: 10.1186/1756-9966-28-8] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/29/2008] [Accepted: 01/15/2009] [Indexed: 01/03/2023]
Abstract
Background Lamin A/C is very important in DNA replication, RNA dependent transcription and nuclear stabilization. Reduced or absent lamin A/C expression has been found to be a common feature of a variety of different cancers. To investigate the role of lamin A/C in gastric carcinoma (GC) pathogenesis, we analyzed the correlations between the lamin A/C expression level and clinicopathological factors and studied its prognostic role in primary GC. Methods The expression of lamin A/C at mRNA level was detected by the reverse transcription-polymerase chain reaction (RT-PCR) and real time RT-PCR, and western blot was used to examine the protein expression. Lamin A/C expression and its prognostic significance were investigated by performing immunohistochemical analysis on a total of 126 GC clinical tissue samples. Results Both lamin A/C mRNA and protein expression were downregulated in the majority of tumours compared with corresponding normal gastric tissues (p = 0.011 and p = 0.036, respectively). Real time RT-PCR further validated that downregulation of lamin A/C is associated with poor histological differentiation (r = 0.438, p = 0.025). The immunohistochemical staining showed an evident decrease of lamin A/C expression in 55.6% (70/126) GC cases. Importantly, the negative lamin A/C expression correlated strongly with histological classification (r = 0.361, p = 0.034). Survival analysis revealed that patients with lamin A/C downregulation have a poorer prognosis (p = 0.034). In addition, lamin A/C expression was found to be an independent prognostic factor by multivariate analysis. Conclusion Data of this study suggest that lamin A/C is involved in the pathogenesis of GC, and it may serve as a valuable biomarker for assessing the prognosis for primary GC.
Collapse
Affiliation(s)
- Zhengrong Wu
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, PR China.
| | | | | | | | | | | |
Collapse
|
130
|
Hanif M, Rosengardten Y, Sagelius H, Rozell B, Eriksson M. Differential expression of A-type and B-type lamins during hair cycling. PLoS One 2009; 4:e4114. [PMID: 19122810 PMCID: PMC2606029 DOI: 10.1371/journal.pone.0004114] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2008] [Accepted: 12/02/2008] [Indexed: 11/18/2022] Open
Abstract
Multiple genetic disorders caused by mutations that affect the proteins lamin A and C show strong skin phenotypes. These disorders include the premature aging disorders Hutchinson-Gilford progeria syndrome and mandibuloacral dysplasia, as well as restrictive dermopathy. Prior studies have shown that the lamin A/C and B proteins are expressed in skin, but little is known about their normal expression in the different skin cell-types and during the hair cycle. Our immunohistochemical staining for lamins A/C and B in wild-type mice revealed strong expression in the basal cell layer of the epidermis, the outer root sheath, and the dermal papilla during all stages of the hair cycle. Lower expression of both lamins A/C and B was seen in suprabasal cells of the epidermis, in the hypodermis, and in the bulb of catagen follicles. In addition, we have utilized a previously described mouse model of Hutchinson-Gilford progeria syndrome and show here that the expression of progerin does not result in pronounced effects on hair cycling or the expression of lamin B.
Collapse
Affiliation(s)
- Mubashir Hanif
- The Folkhälsan Institute of Genetics, Biomedicum Helsinki, University of Helsinki, Helsinki, Finland
| | - Ylva Rosengardten
- Department of Biosciences and Nutrition, Karolinska Institutet, Karolinska University Hospital, Huddinge, Novum, Stockholm, Sweden
| | - Hanna Sagelius
- Department of Biosciences and Nutrition, Karolinska Institutet, Karolinska University Hospital, Huddinge, Novum, Stockholm, Sweden
| | - Björn Rozell
- Clinical Research Center, Department of Laboratory Medicine, Karolinska Institutet, Karolinska University Hospital, Huddinge, Stockholm, Sweden
| | - Maria Eriksson
- Department of Biosciences and Nutrition, Karolinska Institutet, Karolinska University Hospital, Huddinge, Novum, Stockholm, Sweden
- * E-mail:
| |
Collapse
|
131
|
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
|
132
|
Willis ND, Wilson RG, Hutchison CJ. Lamin A: a putative colonic epithelial stem cell biomarker which identifies colorectal tumours with a more aggressive phenotype. Biochem Soc Trans 2008; 36:1350-3. [PMID: 19021554 DOI: 10.1042/bst0361350] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/31/2025]
Abstract
Abnormalities in the expression, distribution and structural organization of A-type lamins are most commonly associated with a spectrum of inherited disorders which predominantly affect mesenchymal lineages, collectively known as laminopathies. However, a new role for lamin A has been discovered in the progression of a common epithelial cancer. CRC (colorectal cancer) patients expressing lamin A/C in their tumour tissue were found to have a 2-fold greater risk of CRC-related mortality compared with patients with lamin A/C-negative tumours. Consequently, lamin A/C is a prognostic biomarker in CRC. In vitro studies suggest that lamin A is an upstream regulator of a pathway linking actin dynamics to loss of cell adhesion, leading to enhanced cell motility and consequently increased invasive potential within a tumour. The finding that lamin A is a putative colonic epithelial stem cell biomarker suggests that the poor outcome associated with lamin A/C-positive tumours may be reflective of a more stem-cell-like phenotype. The present review discusses the link between lamin A expression and tumour progression in one of the commonest causes of cancer-related death in the Western world.
Collapse
Affiliation(s)
- Naomi D Willis
- School of Biological and Biomedical Sciences, Durham University, Durham, UK
| | | | | |
Collapse
|
133
|
Dysfunctional connections between the nucleus and the actin and microtubule networks in laminopathic models. Biophys J 2008; 95:5462-75. [PMID: 18790843 DOI: 10.1529/biophysj.108.139428] [Citation(s) in RCA: 165] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Laminopathies encompass a wide array of human diseases associated to scattered mutations along LMNA, a single gene encoding A-type lamins. How such genetic alterations translate to cellular defects and generate such diverse disease phenotypes remains enigmatic. Recent work has identified nuclear envelope proteins--emerin and the linker of the nucleoskeleton and cytoskeleton (LINC) complex--which connect the nuclear lamina to the cytoskeleton. Here we quantitatively examine the composition of the nuclear envelope, as well as the architecture and functions of the cytoskeleton in cells derived from two laminopathic mouse models, including Hutchinson-Gilford progeria syndrome (Lmna(L530P/L530P)) and Emery-Dreifuss muscular dystrophy (Lmna(-/-)). Cells derived from the overtly aphenotypical model of X-linked Emery-Dreifuss muscular dystrophy (Emd(-/y)) were also included. We find that the centrosome is detached from the nucleus, preventing centrosome polarization in cells under flow--defects that are mediated by the loss of emerin from the nuclear envelope. Moreover, while basal actin and focal adhesion structure are mildly affected, RhoA activation, cell-substratum adhesion, and cytoplasmic elasticity are greatly lowered, exclusively in laminopathic models in which the LINC complex is disrupted. These results indicate a new function for emerin in cell polarization and suggest that laminopathies are not directly associated with cells' inability to polarize, but rather with cytoplasmic softening and weakened adhesion mediated by the disruption of the LINC complex across the nuclear envelope.
Collapse
|
134
|
Abstract
The human genome is contained within the nucleus and is separated from the cytoplasm by the nuclear envelope. Mutations in the nuclear envelope proteins emerin and lamin A cause a number of diseases including premature aging syndromes, muscular dystrophy, and cardiomyopathy. Emerin and lamin A are implicated in regulating muscle- and heart-specific gene expression and nuclear architecture. For example, lamin A regulates the expression and localization of gap junction and intercalated disc components. Additionally, emerin and lamin A are also required to maintain nuclear envelope integrity. Demonstrating the importance of maintaining nuclear integrity in heart disease, atrioventricular node cells lacking lamin A exhibit increased nuclear deformation and apoptosis. This review highlights the present understanding of lamin A and emerin function in regulating nuclear architecture, gene expression, and cell signaling and discusses putative mechanisms for how specific mutations in lamin A and emerin cause cardiac- or muscle-specific disease.
Collapse
Affiliation(s)
- James M. Holaska
- From the Department of Medicine, Section of Cardiology, University of Chicago, Ill
| |
Collapse
|
135
|
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
|
136
|
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
|
137
|
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
|
138
|
Cohen TV, Stewart CL. Fraying at the edge mouse models of diseases resulting from defects at the nuclear periphery. Curr Top Dev Biol 2008; 84:351-84. [PMID: 19186248 DOI: 10.1016/s0070-2153(08)00607-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Eukaryotic cells compartmentalize their genetic material within the nucleus. The boundary separating the genetic material from the cytoplasm is the nuclear envelope (NE) and lamina. Historically, the NE was perceived as functioning primarily as a barrier regulating the entry and exit of macromolecules between the nucleus and cytoplasm via the nuclear pore complexes (NPCs) that traverse the nuclear membranes. However, recent findings have caused a fundamental reassessment with regard to NE and lamina functions. Evidence now points to the NE and lamina functioning as a "hub" in regulating and perhaps integrating critical cellular functions that include chromatin organization, transcriptional regulation, mechanical integrity of the cell, signaling pathways, as well as acting as a key component of the cytoskeleton. Such an integral role for the nuclear boundary has emerged from increased interest into the functions of the NE/lamina, which has been largely stimulated by the discovery that some 24 different diseases and anomalies are caused by defects in proteins of the NE and lamina.
Collapse
Affiliation(s)
- Tatiana V Cohen
- Center for Genetic Medicine, Children's National Medical Center, N.W. Washington, DC 20010, USA
| | | |
Collapse
|
139
|
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]
|
140
|
Stewart CL, Kozlov S, Fong LG, Young SG. Mouse models of the laminopathies. Exp Cell Res 2007; 313:2144-56. [PMID: 17493612 PMCID: PMC1949387 DOI: 10.1016/j.yexcr.2007.03.026] [Citation(s) in RCA: 87] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2007] [Revised: 03/05/2007] [Accepted: 03/12/2007] [Indexed: 12/25/2022]
Abstract
The A and B type lamins are nuclear intermediate filament proteins that comprise the bulk of the nuclear lamina, a thin proteinaceous structure underlying the inner nuclear membrane. The A type lamins are encoded by the lamin A gene (LMNA). Mutations in this gene have been linked to at least nine diseases, including the progeroid diseases Hutchinson-Gilford progeria and atypical Werner's syndromes, striated muscle diseases including muscular dystrophies and dilated cardiomyopathies, lipodystrophies affecting adipose tissue deposition, diseases affecting skeletal development, and a peripheral neuropathy. To understand how different diseases arise from different mutations in the same gene, mouse lines carrying some of the same mutations found in the human diseases have been established. We, and others have generated mice with different mutations that result in progeria, muscular dystrophy, and dilated cardiomyopathy. To further our understanding of the functions of the lamins, we also created mice lacking lamin B1, as well as mice expressing only one of the A type lamins. These mouse lines are providing insights into the functions of the lamina and how changes to the lamina affect the mechanical integrity of the nucleus as well as signaling pathways that, when disrupted, may contribute to the disease.
Collapse
Affiliation(s)
- Colin L Stewart
- Laboratory of Cancer and Developmental Biology, National Cancer Institute, Frederick, Maryland 21702, USA.
| | | | | | | |
Collapse
|
141
|
Worman HJ, Bonne G. "Laminopathies": a wide spectrum of human diseases. Exp Cell Res 2007; 313:2121-33. [PMID: 17467691 PMCID: PMC2964355 DOI: 10.1016/j.yexcr.2007.03.028] [Citation(s) in RCA: 496] [Impact Index Per Article: 27.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2007] [Revised: 03/08/2007] [Accepted: 03/12/2007] [Indexed: 12/23/2022]
Abstract
Mutations in genes encoding the intermediate filament nuclear lamins and associated proteins cause a wide spectrum of diseases sometimes called "laminopathies." Diseases caused by mutations in LMNA encoding A-type lamins include autosomal dominant Emery-Dreifuss muscular dystrophy and related myopathies, Dunnigan-type familial partial lipodystrophy, Charcot-Marie-Tooth disease type 2B1 and developmental and accelerated aging disorders. Duplication in LMNB1 encoding lamin B1 causes autosomal dominant leukodystrophy and mutations in LMNB2 encoding lamin B2 are associated with acquired partial lipodystrophy. Disorders caused by mutations in genes encoding lamin-associated integral inner nuclear membrane proteins include X-linked Emery-Dreifuss muscular dystrophy, sclerosing bone dysplasias, HEM/Greenberg skeletal dysplasia and Pelger-Huet anomaly. While mutations and clinical phenotypes of "laminopathies" have been carefully described, data explaining pathogenic mechanisms are only emerging. Future investigations will likely identify new "laminopathies" and a combination of basic and clinical research will lead to a better understanding of pathophysiology and the development of therapies.
Collapse
Affiliation(s)
- Howard J Worman
- Department of Medicine, College of Physicians and Surgeons, Columbia University, 630 West 168th Street, New York, NY 10032, USA.
| | | |
Collapse
|
142
|
Takamori Y, Tamura Y, Kataoka Y, Cui Y, Seo S, Kanazawa T, Kurokawa K, Yamada H. Differential expression of nuclear lamin, the major component of nuclear lamina, during neurogenesis in two germinal regions of adult rat brain. Eur J Neurosci 2007; 25:1653-62. [PMID: 17432957 DOI: 10.1111/j.1460-9568.2007.05450.x] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Lamins are major structural proteins of the nuclear envelope. Three lamin subtypes, A/C, B1 and B2, predominate in mammalian somatic cells. While the expression levels of lamins in several tissues are known to change during cell differentiation, lamin expression is poorly understood in the nervous system. To investigate the expression of lamins during neuronal differentiation in the mammalian adult brain, we performed immunohistochemical studies on lamins A/C, B1 and B2 in two neurogenic regions of rat brain: the subgranular zone of the dentate gyrus and the subventricular zone of the lateral ventricle. In particular, three types of cells were analysed using confocal microscopy: GFAP-positive cells as primary progenitor (stem) cells, PSA-NCAM-positive cells as subsequent neuronal progenitor cells, and NeuN-positive mature neurons. GFAP-positive cells possesed lamin A/C (++), B1 (++) and B2 (++), PSA-NCAM-positive cells possessed lamin A/C (-), B1 (+++) and B2 (+), and mature neurons possessed lamin A/C (++), B1 (+) and B2 (+++), in both neurogenic regions. These observations showed that the compositions of expressing lamin subtypes are distinct in particular differentiation stages during neurogenesis in the adult rat brain. Our results suggest that the alteration of nuclear lamina structure is coupled with the progression of neuronal differentiation.
Collapse
Affiliation(s)
- Yasuharu Takamori
- Department of Anatomy and Cell Science, Kansai Medical University, Moriguchi, Osaka, Japan
| | | | | | | | | | | | | | | |
Collapse
|
143
|
Roux KJ, Burke B. Nuclear envelope defects in muscular dystrophy. Biochim Biophys Acta Mol Basis Dis 2007; 1772:118-27. [PMID: 16904876 DOI: 10.1016/j.bbadis.2006.06.001] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2006] [Accepted: 06/03/2006] [Indexed: 11/29/2022]
Abstract
Muscular dystrophies are a heterogeneous group of disorders linked to defects in 20-30 different genes. Mutations in the genes encoding a pair of nuclear envelope proteins, emerin and lamin A/C, have been shown to cause the X-linked and autosomal forms respectively of Emery-Dreifuss muscular dystrophy. A third form of muscular dystrophy, limb girdle muscular dystrophy 1b, has also been linked to mutations in the lamin A/C gene. Given that these two genes are ubiquitously expressed, a major goal is to determine how they can be associated with tissue specific diseases. Recent results suggest that lamin A/C and emerin contribute to the maintenance of nuclear envelope structure and at the same time may modulate the expression patterns of certain mechanosensitive and stress induced genes. Both emerin and lamin A/C may play an important role in the response of cells to mechanical stress and in this way may help to maintain muscle cell integrity.
Collapse
Affiliation(s)
- Kyle J Roux
- Department of Anatomy and Cell Biology, The University of Florida College of Medicine, 1600 SW Archer Road, Gainesville, FL 32606, USA
| | | |
Collapse
|
144
|
Muchir A, Massart C, van Engelen BG, Lammens M, Bonne G, Worman HJ. Proteasome-mediated degradation of integral inner nuclear membrane protein emerin in fibroblasts lacking A-type lamins. Biochem Biophys Res Commun 2006; 351:1011-7. [PMID: 17097067 PMCID: PMC1771114 DOI: 10.1016/j.bbrc.2006.10.147] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2006] [Accepted: 10/26/2006] [Indexed: 11/18/2022]
Abstract
We previously identified and characterized a homozygous LMNA nonsense mutation leading to the absence of A-type lamins in a premature neonate who died at birth. We show here that the absence of A-type lamins is due to degradation of the aberrant mRNA transcript with a premature termination codon. In cultured fibroblasts from the subject with the homozygous LMNA nonsense mutation, there was a decreased steady-state expression of the integral inner nuclear membrane proteins emerin and nesprin-1alpha associated with their mislocalization to the bulk endoplasmic reticulum and a hyperphosphorylation of emerin. To determine if decreased emerin expression occurred post-translationally, we treated cells with a selective proteasome inhibitor and observed an increase in expression. Our results show that mislocalization of integral inner nuclear membrane proteins to the endoplasmic reticulum in human cells lacking A-type lamins leads to their degradation and provides the first evidence that their degradation is mediated by the proteasome.
Collapse
Affiliation(s)
- Antoine Muchir
- Departments of Medicine and Anatomy and Cell Biology, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | | | | | | | | | | |
Collapse
|
145
|
Lammerding J, Fong LG, Ji JY, Reue K, Stewart CL, Young SG, Lee RT. Lamins A and C but not lamin B1 regulate nuclear mechanics. J Biol Chem 2006; 281:25768-80. [PMID: 16825190 DOI: 10.1074/jbc.m513511200] [Citation(s) in RCA: 519] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Mutations in the nuclear envelope proteins lamins A and C cause a broad variety of human diseases, including Emery-Dreifuss muscular dystrophy, dilated cardiomyopathy, and Hutchinson-Gilford progeria syndrome. Cells lacking lamins A and C have reduced nuclear stiffness and increased nuclear fragility, leading to increased cell death under mechanical strain and suggesting a potential mechanism for disease. Here, we investigated the contribution of major lamin subtypes (lamins A, C, and B1) to nuclear mechanics by analyzing nuclear shape, nuclear dynamics over time, nuclear deformations under strain, and cell viability under prolonged mechanical stimulation in cells lacking both lamins A and C, cells lacking only lamin A (i.e. "lamin C-only" cells), cells lacking wild-type lamin B1, and wild-type cells. Lamin A/C-deficient cells exhibited increased numbers of misshapen nuclei and had severely reduced nuclear stiffness and decreased cell viability under strain. Lamin C-only cells had slightly abnormal nuclear shape and mildly reduced nuclear stiffness but no decrease in cell viability under strain. Interestingly, lamin B1-deficient cells exhibited normal nuclear mechanics despite having a significantly increased frequency of nuclear blebs. Our study indicates that lamins A and C are important contributors to the mechanical stiffness of nuclei, whereas lamin B1 contributes to nuclear integrity but not stiffness.
Collapse
Affiliation(s)
- Jan Lammerding
- Cardiovascular Division, Department of Medicine, and Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02139, USA.
| | | | | | | | | | | | | |
Collapse
|
146
|
Worman HJ. Inner nuclear membrane and regulation of Smad-mediated signaling. Biochim Biophys Acta Mol Cell Biol Lipids 2006; 1761:626-31. [PMID: 16574476 DOI: 10.1016/j.bbalip.2006.02.010] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2005] [Revised: 02/17/2006] [Accepted: 02/17/2006] [Indexed: 11/30/2022]
Abstract
Smads mediate signal transduction by cytokines of the transforming growth factor-beta family. Recent data show that intrinsic and extrinsic proteins of the inner nuclear membrane affect the activities of Smads. MAN1, an integral protein of the inner nuclear membrane, binds to receptor-regulated Smads and antagonizes signaling by transforming growth factor-beta, activin and bone morphogenic protein. Lamins A and C, extrinsic intermediate filament proteins of the inner nuclear membrane that are mutated in several human diseases, appear to regulate phosphorylation of Smads. These data demonstrate that proteins within and associated with the inner nuclear membrane lipid bilayer regulate signal transduction pathways involved in numerous developmental, physiological and pathophysiological processes.
Collapse
Affiliation(s)
- Howard J Worman
- Department of Medicine, College of Physicians and Surgeons, Columbia University, 630 West 168th Street, 10th Floor, Room 508, New York, NY 10032, USA.
| |
Collapse
|
147
|
Delbarre E, Tramier M, Coppey-Moisan M, Gaillard C, Courvalin JC, Buendia B. The truncated prelamin A in Hutchinson–Gilford progeria syndrome alters segregation of A-type and B-type lamin homopolymers. Hum Mol Genet 2006; 15:1113-22. [PMID: 16481358 DOI: 10.1093/hmg/ddl026] [Citation(s) in RCA: 96] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Hutchinson-Gilford progeria syndrome (HGPS) is a dominant autosomal premature aging syndrome caused by the expression of a truncated prelamin A designated progerin (Pgn). A-type and B-type lamins are intermediate filament proteins that polymerize to form the nuclear lamina network apposed to the inner nuclear membrane of vertebrate somatic cells. It is not known if in vivo both type of lamins assemble independently or co-assemble. The blebbing and disorganization of the nuclear envelope and adjacent heterochromatin in cells from patients with HGPS is a hallmark of the disease, and the ex vivo reversal of this phenotype is considered important for the development of therapeutic strategies. Here, we investigated the alterations in the lamina structure that may underlie the disorganization caused in nuclei by Pgn expression. We studied the polymerization of enhanced green fluorescent protein- and red fluorescent protein-tagged wild-type and mutated lamins in the nuclear envelope of living cells by measuring fluorescence resonance energy transfer (FRET) that occurs between the two fluorophores when tagged lamins interact. Using time domain fluorescence lifetime imaging microscopy that allows a quantitative analysis of FRET signals, we show that wild-type lamins A and B1 polymerize in distinct homopolymers that further interact in the lamina. In contrast, expressed Pgn co-assembles with lamin B1 and lamin A to form a mixed heteropolymer in which A-type and B-type lamin segregation is lost. We propose that such structural lamina alterations may be part of the primary mechanisms leading to HGPS, possibly by impairing functions specific for each lamin type such as nuclear membrane biogenesis, signal transduction, nuclear compartmentalization and gene regulation.
Collapse
Affiliation(s)
- Erwan Delbarre
- Département de Biologie Cellulaire, Institut Jacques Monod, CNRS, Université Paris 6 and 7, 2 Place Jussieu Tour 43, 75251 Paris Cedex 05, France
| | | | | | | | | | | |
Collapse
|
148
|
Crisp M, Liu Q, Roux K, Rattner JB, Shanahan C, Burke B, Stahl PD, Hodzic D. Coupling of the nucleus and cytoplasm: role of the LINC complex. ACTA ACUST UNITED AC 2005; 172:41-53. [PMID: 16380439 PMCID: PMC2063530 DOI: 10.1083/jcb.200509124] [Citation(s) in RCA: 1032] [Impact Index Per Article: 51.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2022]
Abstract
The nuclear envelope defines the barrier between the nucleus and cytoplasm and features inner and outer membranes separated by a perinuclear space (PNS). The inner nuclear membrane contains specific integral proteins that include Sun1 and Sun2. Although the outer nuclear membrane (ONM) is continuous with the endoplasmic reticulum, it is nevertheless enriched in several integral membrane proteins, including nesprin 2 Giant (nesp2G), an 800-kD protein featuring an NH2-terminal actin-binding domain. A recent study (Padmakumar, V.C., T. Libotte, W. Lu, H. Zaim, S. Abraham, A.A. Noegel, J. Gotzmann, R. Foisner, and I. Karakesisoglou. 2005. J. Cell Sci. 118:3419–3430) has shown that localization of nesp2G to the ONM is dependent upon an interaction with Sun1. In this study, we confirm and extend these results by demonstrating that both Sun1 and Sun2 contribute to nesp2G localization. Codepletion of both of these proteins in HeLa cells leads to the loss of ONM-associated nesp2G, as does overexpression of the Sun1 lumenal domain. Both treatments result in the expansion of the PNS. These data, together with those of Padmakumar et al. (2005), support a model in which Sun proteins tether nesprins in the ONM via interactions spanning the PNS. In this way, Sun proteins and nesprins form a complex that links the nucleoskeleton and cytoskeleton (the LINC complex).
Collapse
Affiliation(s)
- Melissa Crisp
- Department of Anatomy and Cell Biology, University of Florida, Gainesville, FL 32610
| | | | | | | | | | | | | | | |
Collapse
|
149
|
Constantinescu D, Gray HL, Sammak PJ, Schatten GP, Csoka AB. Lamin A/C expression is a marker of mouse and human embryonic stem cell differentiation. Stem Cells 2005; 24:177-85. [PMID: 16179429 DOI: 10.1634/stemcells.2004-0159] [Citation(s) in RCA: 271] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Nuclear lamins comprise the nuclear lamina, a scaffold-like structure that lines the inner nuclear membrane. B-type lamins are present in almost all cell types, but A-type lamins are expressed predominantly in differentiated cells, suggesting a role in maintenance of the differentiated state. Previous studies have shown that lamin A/C is not expressed during mouse development before day 9, nor in undifferentiated mouse embryonic carcinoma cells. To further investigate the role of lamins in cell phenotype maintenance and differentiation, we examined lamin expression in undifferentiated mouse and human embryonic stem (ES) cells. Wide-field and confocal immunofluorescence microscopy and semiquantitative reverse transcription-polymerase chain reaction analysis revealed that undifferentiated mouse and human ES cells express lamins B1 and B2 but not lamin A/C. Mouse ES cells display high levels of lamins B1 and B2 localized both at the nuclear periphery and throughout the nucleoplasm, but in human ES cells, B1 and B2 expression is dimmer and localized primarily at the nuclear periphery. Lamin A/C expression is activated during human ES cell differentiation before downregulation of the pluripotency marker Oct-3/4 but not before the downregulation of the pluripotency markers Tra-1-60, Tra-1-81, and SSEA-4. Our results identify the absence of A-type lamin expression as a novel marker for undifferentiated ES cells and further support a role for nuclear lamins in cell maintenance and differentiation.
Collapse
Affiliation(s)
- Dan Constantinescu
- Department of Cell Biology and Physiology, University of Pittsburgh School of Medicine, Pittsburgh Development Center of Magee-Womens Research Institute, 204 Craft Ave, Pittsburgh, Pennsylvania 15213, USA
| | | | | | | | | |
Collapse
|
150
|
Gotzmann J, Foisner R. A-type lamin complexes and regenerative potential: a step towards understanding laminopathic diseases? Histochem Cell Biol 2005; 125:33-41. [PMID: 16142451 DOI: 10.1007/s00418-005-0050-8] [Citation(s) in RCA: 84] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/19/2005] [Indexed: 01/09/2023]
Abstract
The lamins are nuclear intermediate filament-type proteins forming the nuclear lamina meshwork at the inner nuclear membrane as well as complexes in the nucleoplasm. The recent discoveries that mutated A-type lamins and lamin-binding nuclear membrane proteins can be linked to numerous rare human diseases (laminopathies) affecting a multitude of tissues has changed the cell biologist's view of lamins as mere structural nuclear scaffold proteins. It is still unclear how mutations in these ubiquitously expressed proteins give rise to tissue-restricted pathological phenotypes. Potential disease models include mutation-caused defects in lamin structure and stability, the deregulation of gene expression, and impaired cell cycle control. This review brings together various previously proposed ideas and suggests a novel, more general, disease model based on an impairment of adult stem cell function and thus compromised tissue regeneration in laminopathic diseases.
Collapse
Affiliation(s)
- Josef Gotzmann
- Max F. Perutz Laboratories, University Departments at the Vienna Biocenter, Department of Medical Biochemistry, Medical University of Vienna, Dr. Bohrgasse 9/3, 1030, Vienna, Austria
| | | |
Collapse
|