201
|
Goblirsch BR, Arachea BT, Councell DJ, Wiener MC. Phosphoramidon inhibits the integral membrane protein zinc metalloprotease ZMPSTE24. Acta Crystallogr D Struct Biol 2018; 74:739-747. [PMID: 30082509 PMCID: PMC6079626 DOI: 10.1107/s2059798318003431] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Accepted: 02/27/2018] [Indexed: 11/10/2022] Open
Abstract
The integral membrane protein zinc metalloprotease ZMPSTE24 possesses a completely novel structure, comprising seven long kinked transmembrane helices that encircle a voluminous 14 000 Å3 cavity within the membrane. Functionally conserved soluble zinc metalloprotease residues are contained within this cavity. As part of an effort to understand the structural and functional relationships between ZMPSTE24 and soluble zinc metalloproteases, the inhibition of ZMPSTE24 by phosphoramidon [N-(α-rhamnopyranosyl-oxyhydroxyphosphinyl)-Leu-Trp], a transition-state analog and competitive inhibitor of multiple soluble zinc metalloproteases, especially gluzincins, has been characterized functionally and structurally. The functional results, the determination of preliminary IC50 values by the use of an intramolecular quenched-fluorescence fluorogenic peptide assay, indicate that phosphoramidon inhibits ZMPSTE24 in a manner consistent with competitive inhibition. The structural results, a 3.85 Å resolution X-ray crystal structure of a ZMPSTE24-phosphoramidon complex, indicate that the overall binding mode observed between phosphoramidon and soluble gluzincins is conserved. Based on the structural data, a significantly lower potency than that observed for soluble gluzincins such as thermolysin and neprilysin is predicted. These results strongly suggest a close relationship between soluble gluzincins and the integral membrane protein zinc metalloprotease ZMPSTE24.
Collapse
Affiliation(s)
- Brandon R. Goblirsch
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA 22908-0886, USA
| | - Buenafe T. Arachea
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA 22908-0886, USA
| | - Daniel J. Councell
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA 22908-0886, USA
| | - Michael C. Wiener
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA 22908-0886, USA
| |
Collapse
|
202
|
Krishnamoorthy V, Khanna R, Parnaik VK. E3 ubiquitin ligase HECW2 targets PCNA and lamin B1. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2018; 1865:1088-1104. [PMID: 29753763 DOI: 10.1016/j.bbamcr.2018.05.008] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Revised: 04/20/2018] [Accepted: 05/07/2018] [Indexed: 12/21/2022]
Abstract
Lamins constitute the major architectural proteins of the nuclear lamina that help in maintaining nuclear organization. Mutations in lamins are associated with diverse degenerative diseases, collectively termed laminopathies. HECW2, a HECT-type E3 ubiquitin ligase, is transcriptionally upregulated in HeLa cells expressing Emery-Dreifuss muscular dystrophy-causing-lamin A mutants. However, the role of HECW2 upregulation in mediating downstream effects in lamin mutant-expressing cells was previously unexplored. Here, we show that HECW2 interacts with two lamin A-binding proteins, proliferating cell nuclear antigen (PCNA), via a canonical PCNA-interacting protein (PIP) motif, and lamin B1. HECW2 mediates their ubiquitination and targets them for proteasomal degradation. Cells expressing lamin A mutants G232E and Q294P, in which HECW2 is upregulated, show increased proteasomal degradation of PCNA and lamin B1 most likely mediated by HECW2. Our findings establish HECW2 as an E3 ubiquitin ligase for PCNA and lamin B1 which regulates their levels in laminopathic cells. We also found that HECW2 interacts with wild-type lamin A and ubiquitinates it and this interaction is reduced in case of lamin mutants G232E and Q294P. Our findings suggest that interplay among HECW2, lamin A, PCNA, and lamin B1 determines their respective homeostatic levels in the cell and dysregulation of these interactions may contribute to the pathogenicity of laminopathies.
Collapse
Affiliation(s)
| | - Richa Khanna
- CSIR-Centre for Cellular and Molecular Biology, Hyderabad, India
| | - Veena K Parnaik
- CSIR-Centre for Cellular and Molecular Biology, Hyderabad, India.
| |
Collapse
|
203
|
Meinke P, Hintze S, Limmer S, Schoser B. Myotonic Dystrophy-A Progeroid Disease? Front Neurol 2018; 9:601. [PMID: 30140252 PMCID: PMC6095001 DOI: 10.3389/fneur.2018.00601] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Accepted: 07/06/2018] [Indexed: 12/27/2022] Open
Abstract
Myotonic dystrophies (DM) are slowly progressing multisystemic disorders caused by repeat expansions in the DMPK or CNBP genes. The multisystemic involvement in DM patients often reflects the appearance of accelerated aging. This is partly due to visible features such as cataracts, muscle weakness, and frontal baldness, but there are also less obvious features like cardiac arrhythmia, diabetes or hypogammaglobulinemia. These aging features suggest the hypothesis that DM could be a segmental progeroid disease. To identify the molecular cause of this characteristic appearance of accelerated aging we compare clinical features of DM to “typical” segmental progeroid disorders caused by mutations in DNA repair or nuclear envelope proteins. Furthermore, we characterize if this premature aging effect is also reflected on the cellular level in DM and investigate overlaps with “classical” progeroid disorders. To investigate the molecular similarities at the cellular level we use primary DM and control cell lines. This analysis reveals many similarities to progeroid syndromes linked to the nuclear envelope. Our comparison on both clinical and molecular levels argues for qualification of DM as a segmental progeroid disorder.
Collapse
Affiliation(s)
- Peter Meinke
- Friedrich-Baur-Institute at the Department of Neurology, University Hospital, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Stefan Hintze
- Friedrich-Baur-Institute at the Department of Neurology, University Hospital, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Sarah Limmer
- Friedrich-Baur-Institute at the Department of Neurology, University Hospital, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Benedikt Schoser
- Friedrich-Baur-Institute at the Department of Neurology, University Hospital, Ludwig-Maximilians-University Munich, Munich, Germany
| |
Collapse
|
204
|
Spear ED, Hsu ET, Nie L, Carpenter EP, Hrycyna CA, Michaelis S. ZMPSTE24 missense mutations that cause progeroid diseases decrease prelamin A cleavage activity and/or protein stability. Dis Model Mech 2018; 11:dmm.033670. [PMID: 29794150 PMCID: PMC6078402 DOI: 10.1242/dmm.033670] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Accepted: 05/16/2018] [Indexed: 12/24/2022] Open
Abstract
The human zinc metalloprotease ZMPSTE24 is an integral membrane protein crucial for the final step in the biogenesis of the nuclear scaffold protein lamin A, encoded by LMNA. After farnesylation and carboxyl methylation of its C-terminal CAAX motif, the lamin A precursor (prelamin A) undergoes proteolytic removal of its modified C-terminal 15 amino acids by ZMPSTE24. Mutations in LMNA or ZMPSTE24 that impede this prelamin A cleavage step cause the premature aging disease Hutchinson-Gilford progeria syndrome (HGPS), and the related progeroid disorders mandibuloacral dysplasia type B (MAD-B) and restrictive dermopathy (RD). Here, we report the development of a ‘humanized yeast system’ to assay ZMPSTE24-dependent cleavage of prelamin A and examine the eight known disease-associated ZMPSTE24 missense mutations. All mutations show diminished prelamin A processing and fall into three classes, with defects in activity, protein stability or both. Notably, some ZMPSTE24 mutants can be rescued by deleting the E3 ubiquitin ligase Doa10, involved in endoplasmic reticulum (ER)-associated degradation of misfolded membrane proteins, or by treatment with the proteasome inhibitor bortezomib. This finding may have important therapeutic implications for some patients. We also show that ZMPSTE24-mediated prelamin A cleavage can be uncoupled from the recently discovered role of ZMPSTE24 in clearance of ER membrane translocon-clogged substrates. Together with the crystal structure of ZMPSTE24, this humanized yeast system can guide structure-function studies to uncover mechanisms of prelamin A cleavage, translocon unclogging, and membrane protein folding and stability. Summary: The zinc metalloprotease ZMPSTE24 performs the final step of prelamin A processing. Here, a yeast-based system shows differences in protein stability and activity for alleles of ZMPSTE24 that cause progeria disease.
Collapse
Affiliation(s)
- Eric D Spear
- Department of Cell Biology, The Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Erh-Ting Hsu
- Department of Chemistry, Purdue University, West Lafayette, IN 47907, USA
| | - Laiyin Nie
- Structural Genomics Consortium, University of Oxford, Oxford OX3 7DQ, UK
| | | | | | - Susan Michaelis
- Department of Cell Biology, The Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
205
|
Danda R, Ganapathy K, Sathe G, Madugundu AK, Krishnan UM, Khetan V, Rishi P, Gowda H, Pandey A, Subramanian K, Prasad TSK, Elchuri SV. Membrane Proteome of Invasive Retinoblastoma: Differential Proteins and Biomarkers. Proteomics Clin Appl 2018; 12:e1700101. [DOI: 10.1002/prca.201700101] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 03/22/2018] [Indexed: 11/09/2022]
Affiliation(s)
- Ravikanth Danda
- Department of Ocular Pathology, Vision Research Foundation; Sankara Nethralaya; Chennai 600006 Tamil Nadu India
- Centre for Nanotechnology and Advanced Biomaterials; SASTRA University; Tanjore 613401 Tamil Nadu India
| | - Kalaivani Ganapathy
- Department of Ocular Pathology, Vision Research Foundation; Sankara Nethralaya; Chennai 600006 Tamil Nadu India
| | - Gajanan Sathe
- Institute of Bioinformatics; International Technology Park; Bangalore 560066 Karnataka India
| | - Anil K. Madugundu
- Institute of Bioinformatics; International Technology Park; Bangalore 560066 Karnataka India
| | - Uma Maheswari Krishnan
- Centre for Nanotechnology and Advanced Biomaterials; SASTRA University; Tanjore 613401 Tamil Nadu India
| | - Vikas Khetan
- Shri Bhagwan Mahavir Vitreoretinal Services and Ocular Oncology Services, Medical Research Foundation; Sankara Nethralaya; Chennai 600006 Tamil Nadu India
| | - Pukhraj Rishi
- Shri Bhagwan Mahavir Vitreoretinal Services and Ocular Oncology Services, Medical Research Foundation; Sankara Nethralaya; Chennai 600006 Tamil Nadu India
| | - Harsha Gowda
- Institute of Bioinformatics; International Technology Park; Bangalore 560066 Karnataka India
- Manipal Academy of Higher Education (MAHE); Manipal 576104 Karnataka India
| | - Akhilesh Pandey
- McKusick-Nathans Institute of Genetic Medicine; Johns Hopkins University School of Medicine; 21205 Baltimore MD USA
- Department of Pathology; Johns Hopkins University School of Medicine; 21205 Baltimore MD USA
- Department of Oncology; Johns Hopkins University School of Medicine; 21205 Baltimore MD USA
- Department of Biological Chemistry; Johns Hopkins University School of Medicine; 21205 Baltimore MD USA
- Manipal Academy of Higher Education (MAHE); Manipal 576104 Karnataka India
| | - Krishnakumar Subramanian
- Department of Ocular Pathology, Vision Research Foundation; Sankara Nethralaya; Chennai 600006 Tamil Nadu India
| | - T. S. Keshava Prasad
- Institute of Bioinformatics; International Technology Park; Bangalore 560066 Karnataka India
| | - Sailaja V. Elchuri
- Department of Nano-Biotechnology, Vision Research Foundation; Sankara Nethralya; Chennai 600006 Tamil Nadu India
| |
Collapse
|
206
|
Bhide S, Trujillo AS, O'Connor MT, Young GH, Cryderman DE, Chandran S, Nikravesh M, Wallrath LL, Melkani GC. Increasing autophagy and blocking Nrf2 suppress laminopathy-induced age-dependent cardiac dysfunction and shortened lifespan. Aging Cell 2018; 17:e12747. [PMID: 29575479 PMCID: PMC5946079 DOI: 10.1111/acel.12747] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/10/2018] [Indexed: 12/16/2022] Open
Abstract
Mutations in the human LMNA gene cause a collection of diseases known as laminopathies. These include myocardial diseases that exhibit age-dependent penetrance of dysrhythmias and heart failure. The LMNA gene encodes A-type lamins, intermediate filaments that support nuclear structure and organize the genome. Mechanisms by which mutant lamins cause age-dependent heart defects are not well understood. To address this issue, we modeled human disease-causing mutations in the Drosophila melanogaster Lamin C gene and expressed mutant Lamin C exclusively in the heart. This resulted in progressive cardiac dysfunction, loss of adipose tissue homeostasis, and a shortened adult lifespan. Within cardiac cells, mutant Lamin C aggregated in the cytoplasm, the CncC(Nrf2)/Keap1 redox sensing pathway was activated, mitochondria exhibited abnormal morphology, and the autophagy cargo receptor Ref2(P)/p62 was upregulated. Genetic analyses demonstrated that simultaneous over-expression of the autophagy kinase Atg1 gene and an RNAi against CncC eliminated the cytoplasmic protein aggregates, restored cardiac function, and lengthened lifespan. These data suggest that simultaneously increasing rates of autophagy and blocking the Nrf2/Keap1 pathway are a potential therapeutic strategy for cardiac laminopathies.
Collapse
Affiliation(s)
- Shruti Bhide
- Department of Biology, Molecular Biology and Heart Institutes; San Diego State University; San Diego CA USA
| | - Adriana S. Trujillo
- Department of Biology, Molecular Biology and Heart Institutes; San Diego State University; San Diego CA USA
| | - Maureen T. O'Connor
- Department of Biochemistry; Carver College of Medicine; University of Iowa; Iowa City IA USA
| | - Grant H. Young
- Department of Biochemistry; Carver College of Medicine; University of Iowa; Iowa City IA USA
| | - Diane E. Cryderman
- Department of Biochemistry; Carver College of Medicine; University of Iowa; Iowa City IA USA
| | - Sahaana Chandran
- Department of Biology, Molecular Biology and Heart Institutes; San Diego State University; San Diego CA USA
| | - Mastaneh Nikravesh
- Department of Biology, Molecular Biology and Heart Institutes; San Diego State University; San Diego CA USA
| | - Lori L. Wallrath
- Department of Biochemistry; Carver College of Medicine; University of Iowa; Iowa City IA USA
| | - Girish C. Melkani
- Department of Biology, Molecular Biology and Heart Institutes; San Diego State University; San Diego CA USA
| |
Collapse
|
207
|
Simon DN, Wriston A, Fan Q, Shabanowitz J, Florwick A, Dharmaraj T, Peterson SB, Gruenbaum Y, Carlson CR, Grønning-Wang LM, Hunt DF, Wilson KL. OGT ( O-GlcNAc Transferase) Selectively Modifies Multiple Residues Unique to Lamin A. Cells 2018; 7:E44. [PMID: 29772801 PMCID: PMC5981268 DOI: 10.3390/cells7050044] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Revised: 05/11/2018] [Accepted: 05/14/2018] [Indexed: 12/31/2022] Open
Abstract
The LMNA gene encodes lamins A and C with key roles in nuclear structure, signaling, gene regulation, and genome integrity. Mutations in LMNA cause over 12 diseases ('laminopathies'). Lamins A and C are identical for their first 566 residues. However, they form separate filaments in vivo, with apparently distinct roles. We report that lamin A is β-O-linked N-acetylglucosamine-(O-GlcNAc)-modified in human hepatoma (Huh7) cells and in mouse liver. In vitro assays with purified O-GlcNAc transferase (OGT) enzyme showed robust O-GlcNAcylation of recombinant mature lamin A tails (residues 385⁻646), with no detectable modification of lamin B1, lamin C, or 'progerin' (Δ50) tails. Using mass spectrometry, we identified 11 O-GlcNAc sites in a 'sweet spot' unique to lamin A, with up to seven sugars per peptide. Most sites were unpredicted by current algorithms. Double-mutant (S612A/T643A) lamin A tails were still robustly O-GlcNAc-modified at seven sites. By contrast, O-GlcNAcylation was undetectable on tails bearing deletion Δ50, which causes Hutchinson⁻Gilford progeria syndrome, and greatly reduced by deletion Δ35. We conclude that residues deleted in progeria are required for substrate recognition and/or modification by OGT in vitro. Interestingly, deletion Δ35, which does not remove the majority of identified O-GlcNAc sites, does remove potential OGT-association motifs (lamin A residues 622⁻625 and 639⁻645) homologous to that in mouse Tet1. These biochemical results are significant because they identify a novel molecular pathway that may profoundly influence lamin A function. The hypothesis that lamin A is selectively regulated by OGT warrants future testing in vivo, along with two predictions: genetic variants may contribute to disease by perturbing OGT-dependent regulation, and nutrient or other stresses might cause OGT to misregulate wildtype lamin A.
Collapse
Affiliation(s)
- Dan N Simon
- Department of Cell Biology, Johns Hopkins University School of Medicine, 725 North Wolfe Street, Baltimore, MD 21205, USA.
| | - Amanda Wriston
- Department of Chemistry, University of Virginia, Charlottesville, VA 22904, USA.
| | - Qiong Fan
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, 0317 Oslo, Norway.
| | - Jeffrey Shabanowitz
- Department of Chemistry, University of Virginia, Charlottesville, VA 22904, USA.
| | - Alyssa Florwick
- Department of Cell Biology, Johns Hopkins University School of Medicine, 725 North Wolfe Street, Baltimore, MD 21205, USA.
| | - Tejas Dharmaraj
- Department of Cell Biology, Johns Hopkins University School of Medicine, 725 North Wolfe Street, Baltimore, MD 21205, USA.
| | - Sherket B Peterson
- Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| | - Yosef Gruenbaum
- Department of Genetics, Institute of Life Sciences, Hebrew University of Jerusalem, Givat Ram Jerusalem 91904, Israel.
| | - Cathrine R Carlson
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, 0450 Oslo, Norway.
| | - Line M Grønning-Wang
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, 0317 Oslo, Norway.
| | - Donald F Hunt
- Department of Chemistry, University of Virginia, Charlottesville, VA 22904, USA.
- Department of Pathology, University of Virginia, Charlottesville, VA 22904, USA.
| | - Katherine L Wilson
- Department of Cell Biology, Johns Hopkins University School of Medicine, 725 North Wolfe Street, Baltimore, MD 21205, USA.
| |
Collapse
|
208
|
DuBose AJ, Lichtenstein ST, Petrash NM, Erdos MR, Gordon LB, Collins FS. Everolimus rescues multiple cellular defects in laminopathy-patient fibroblasts. Proc Natl Acad Sci U S A 2018; 115:4206-4211. [PMID: 29581305 PMCID: PMC5910873 DOI: 10.1073/pnas.1802811115] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
LMNA encodes the A-type lamins that are part of the nuclear scaffold. Mutations in LMNA can cause a variety of disorders called laminopathies, including Hutchinson-Gilford progeria syndrome (HGPS), atypical Werner syndrome, and Emery-Dreifuss muscular dystrophy. Previous work has shown that treatment of HGPS cells with the mTOR inhibitor rapamycin or with the rapamycin analog everolimus corrects several of the phenotypes seen at the cellular level-at least in part by increasing autophagy and reducing the amount of progerin, the toxic form of lamin A that is overproduced in HGPS patients. Since other laminopathies also result in production of abnormal and potentially toxic lamin proteins, we hypothesized that everolimus would also be beneficial in those disorders. To test this, we applied everolimus to fibroblast cell lines from six laminopathy patients, each with a different mutation in LMNA Everolimus treatment increased proliferative ability and delayed senescence in all cell lines. In several cell lines, we observed that with treatment, there is a significant improvement in nuclear blebbing, which is a cellular hallmark of HGPS and other lamin disorders. These preclinical results suggest that everolimus might have clinical benefit for multiple laminopathy syndromes.
Collapse
Affiliation(s)
- Amanda J DuBose
- Genome Technology Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892
| | - Stephen T Lichtenstein
- Genome Technology Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892
| | - Noreen M Petrash
- Genome Technology Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892
| | - Michael R Erdos
- Genome Technology Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892
| | - Leslie B Gordon
- Department of Pediatrics, Hasbro Children's Hospital and Warren Alpert Medical School of Brown University, Providence, RI 02903
- Department of Anesthesia, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115
| | - Francis S Collins
- Genome Technology Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892;
| |
Collapse
|
209
|
Borroni AP, Emanuelli A, Shah PA, Ilić N, Apel-Sarid L, Paolini B, Manikoth Ayyathan D, Koganti P, Levy-Cohen G, Blank M. Smurf2 regulates stability and the autophagic-lysosomal turnover of lamin A and its disease-associated form progerin. Aging Cell 2018; 17. [PMID: 29405587 PMCID: PMC5847874 DOI: 10.1111/acel.12732] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/06/2018] [Indexed: 12/19/2022] Open
Abstract
A‐lamins, encoded by the LMNA gene, are major structural components of the nuclear lamina coordinating essential cellular processes. Mutations in the LMNA gene and/or alterations in its expression levels have been linked to a distinct subset of human disorders, collectively known as laminopathies, and to cancer. Mechanisms regulating A‐lamins are mostly obscure. Here, we identified E3 ubiquitin ligase Smurf2 as a physiological regulator of lamin A and its disease‐associated mutant form progerin (LAΔ50), whose expression underlies the development of Hutchinson‐Gilford progeria syndrome (HGPS), a devastating premature aging syndrome. We show that Smurf2 directly binds, ubiquitinates, and negatively regulates the expression of lamin A and progerin in Smurf2 dose‐ and E3 ligase‐dependent manners. Overexpression of catalytically active Smurf2 promotes the autophagic–lysosomal breakdown of lamin A and progerin, whereas Smurf2 depletion increases lamin A levels. Remarkably, acute overexpression of Smurf2 in progeria fibroblasts was able to significantly reduce the nuclear deformability. Furthermore, we demonstrate that the reciprocal relationship between Smurf2 and A‐lamins is preserved in different types of mouse and human normal and cancer tissues. These findings establish Smurf2 as an essential regulator of lamin A and progerin and lay a foundation for evaluating the efficiency of progerin clearance by Smurf2 in HGPS, and targeting of the Smurf2–lamin A axis in age‐related diseases such as cancer.
Collapse
Affiliation(s)
- Aurora Paola Borroni
- Laboratory of Molecular and Cellular Cancer Biology; Azrieli Faculty of Medicine; Bar-Ilan University; Safed Israel
| | - Andrea Emanuelli
- Laboratory of Molecular and Cellular Cancer Biology; Azrieli Faculty of Medicine; Bar-Ilan University; Safed Israel
| | - Pooja Anil Shah
- Laboratory of Molecular and Cellular Cancer Biology; Azrieli Faculty of Medicine; Bar-Ilan University; Safed Israel
| | - Nataša Ilić
- Laboratory of Molecular and Cellular Cancer Biology; Azrieli Faculty of Medicine; Bar-Ilan University; Safed Israel
| | - Liat Apel-Sarid
- Department of Pathology; The Galilee Medical Center; Nahariya Israel
| | - Biagio Paolini
- Department of Pathology and Laboratory Medicine; IRCCS Fondazione; Istituto Nazionale dei Tumori; Milan Italy
| | - Dhanoop Manikoth Ayyathan
- Laboratory of Molecular and Cellular Cancer Biology; Azrieli Faculty of Medicine; Bar-Ilan University; Safed Israel
| | - Praveen Koganti
- Laboratory of Molecular and Cellular Cancer Biology; Azrieli Faculty of Medicine; Bar-Ilan University; Safed Israel
| | - Gal Levy-Cohen
- Laboratory of Molecular and Cellular Cancer Biology; Azrieli Faculty of Medicine; Bar-Ilan University; Safed Israel
| | - Michael Blank
- Laboratory of Molecular and Cellular Cancer Biology; Azrieli Faculty of Medicine; Bar-Ilan University; Safed Israel
| |
Collapse
|
210
|
Maraldi NM. The lamin code. Biosystems 2018; 164:68-75. [DOI: 10.1016/j.biosystems.2017.07.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Revised: 07/10/2017] [Accepted: 07/14/2017] [Indexed: 12/24/2022]
|
211
|
Guillín-Amarelle C, Fernández-Pombo A, Sánchez-Iglesias S, Araújo-Vilar D. Lipodystrophic laminopathies: Diagnostic clues. Nucleus 2018; 9:249-260. [PMID: 29557732 PMCID: PMC5973260 DOI: 10.1080/19491034.2018.1454167] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Revised: 11/02/2017] [Accepted: 03/15/2018] [Indexed: 01/19/2023] Open
Abstract
The nuclear lamina is a complex reticular structure that covers the inner face of the nucleus membrane in metazoan cells. It is mainly formed by intermediate filaments called lamins, and exerts essential functions to maintain the cellular viability. Lamin A/C provides mechanical steadiness to the nucleus and regulates genetic machinery. Laminopathies are tissue-specific or systemic disorders caused by variants in LMNA gene (primary laminopathies) or in other genes encoding proteins which are playing some role in prelamin A maturation or in lamin A/C function (secondary laminopathies). Those disorders in which adipose tissue is affected are called laminopathic lipodystrophies and include type 2 familial partial lipodystrophy and certain premature aging syndromes. This work summarizes the main clinical features of these syndromes, their associated comorbidities and the clues for the differential diagnosis with other lipodystrophic disorders.
Collapse
Affiliation(s)
- Cristina Guillín-Amarelle
- UETeM-Molecular Pathology Group, Department of Medicine, IDIS-CIMUS, University of Santiago de Compostela, Spain
| | - Antía Fernández-Pombo
- UETeM-Molecular Pathology Group, Department of Medicine, IDIS-CIMUS, University of Santiago de Compostela, Spain
| | - Sofía Sánchez-Iglesias
- UETeM-Molecular Pathology Group, Department of Medicine, IDIS-CIMUS, University of Santiago de Compostela, Spain
| | - David Araújo-Vilar
- UETeM-Molecular Pathology Group, Department of Medicine, IDIS-CIMUS, University of Santiago de Compostela, Spain
| |
Collapse
|
212
|
Izdebska M, Gagat M, Grzanka A. Overexpression of lamin B1 induces mitotic catastrophe in colon cancer LoVo cells and is associated with worse clinical outcomes. Int J Oncol 2018; 52:89-102. [PMID: 29115590 PMCID: PMC5743383 DOI: 10.3892/ijo.2017.4182] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2017] [Accepted: 10/10/2017] [Indexed: 02/07/2023] Open
Abstract
Lamins are the major components of the nuclear lamina and play important roles in many cellular processes. The role of lamins in cancer development and progression is still unclear but it is known that reduced expression of lamin B1 has been observed in colon cancer. Thus, the aim of the present study was to elucidate the influence of LMNB1 upregulation on colon cancer cell line after treatment with 5-FU. The results indicate, that overexpression of LMNB1 induced dose-dependent cell death mainly by mitotic catastrophe pathway. Furthermore, after upregulation of this intermediate protein, lower expression of lamin A/C was observed. Moreover, we observed an increase in fluorescence intensity of nuclear β-catenin and decrease in cell-cell interaction area, that was connected with inhibition of colon cancer cells migration. We present the reorganization of actin filament and β-tubulin, because these cytoskeletal proteins are directly or indirectly linked with lamins, and analyzing publicly available mRNA data we show that patients with overexpression of LMNB1 are characterized by lower survival rates within the first 30 months from diagnosis. Summarizing our results, upregulation of LMNB1 induce mitotic catastrophe and only small percentage of apoptosis. Moreover, we showed inhibition of cell migration and promotion of cell-cell contact as a results of direct and indirect regulation of β-catenin, lamin A/C, actin and tubulin. However, it is possible that mitotic catastrophe cells in patients with colorectal cancer may be a reservoir of the cells responsible for faster disease progression, and further investigations are necessary to confirm this hypothesis.
Collapse
Affiliation(s)
| | | | - Alina Grzanka
- Department of Histology and Embryology, Faculty of Medicine, Nicolaus Copernicus University in Toruń, Collegium Medicum in Bydgoszcz, 85-092 Bydgoszcz, Poland
| |
Collapse
|
213
|
Kolb T, Kraxner J, Skodzek K, Haug M, Crawford D, Maaß KK, Aifantis KE, Whyte G. Optomechanical measurement of the role of lamins in whole cell deformability. JOURNAL OF BIOPHOTONICS 2017; 10:1657-1664. [PMID: 28485113 DOI: 10.1002/jbio.201600198] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Revised: 03/12/2017] [Accepted: 03/13/2017] [Indexed: 06/07/2023]
Abstract
There is mounting evidence that the nuclear envelope, and particularly the lamina, plays a critical role in the mechanical and regulation properties of the cell and changes to the lamina can have implications for the physical properties of the whole cell. In this study we demonstrate that the optical stretcher can measure changes in the time-dependent mechanical properties of living cells with different levels of A-type lamin expression. Results from the optical stretcher shows a decrease in the deformability of cells as the levels of lamin A increases, for cells which grow both adherently and in suspension. Further detail can be probed by combining the optical stretcher with fluorescence microscopy to investigate the nuclear mechanical properties which show a larger decrease in deformability than for the whole cell.
Collapse
Affiliation(s)
- Thorsten Kolb
- Department of Physics, Friedrich-Alexander-Universität Erlangen-Nürnberg, Henkestrasse 91, 91052, Erlangen, Germany
- Division of Molecular Genetics, Deutsches Krebsforschungszentrum, Im Neuenheimer Feld 580, 69120, Heidelberg, Germany
| | - Julia Kraxner
- Department of Physics, Friedrich-Alexander-Universität Erlangen-Nürnberg, Henkestrasse 91, 91052, Erlangen, Germany
| | - Kai Skodzek
- Institute of Biological Chemistry, Biophysics and Bioengineering, Heriot-Watt University, Edinburgh, EH14 4AS, UK
| | - Michael Haug
- Department of Physics, Friedrich-Alexander-Universität Erlangen-Nürnberg, Henkestrasse 91, 91052, Erlangen, Germany
| | - Dean Crawford
- Institute of Biological Chemistry, Biophysics and Bioengineering, Heriot-Watt University, Edinburgh, EH14 4AS, UK
| | - Kendra K Maaß
- Division of Molecular Genetics, Deutsches Krebsforschungszentrum, Im Neuenheimer Feld 580, 69120, Heidelberg, Germany
| | - Katerina E Aifantis
- Lab of Mechanics and Materials, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
- Department of Civil Engineering-Engineering Mechanics, University of Arizona, Tuscon, Arizona, 85721
| | - Graeme Whyte
- Department of Physics, Friedrich-Alexander-Universität Erlangen-Nürnberg, Henkestrasse 91, 91052, Erlangen, Germany
- Institute of Biological Chemistry, Biophysics and Bioengineering, Heriot-Watt University, Edinburgh, EH14 4AS, UK
| |
Collapse
|
214
|
Bhattacharjee P, Dasgupta D, Sengupta K. DCM associated LMNA mutations cause distortions in lamina structure and assembly. Biochim Biophys Acta Gen Subj 2017; 1861:2598-2608. [PMID: 28844980 DOI: 10.1016/j.bbagen.2017.08.016] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Revised: 08/01/2017] [Accepted: 08/11/2017] [Indexed: 11/22/2022]
Abstract
BACKGROUND A and B-type lamins are integral scaffolding components of the nuclear lamina which impart rigidity and shape to all metazoan nuclei. Over 450 mutations in A-type lamins are associated with 16 human diseases including dilated cardiomyopathy (DCM). Here, we show that DCM mutants perturb the self-association of lamin A (LA) and it's binding with lamin B1 (LB1). METHODS We used confocal and superresolution microscopy (NSIM) to study the effect of LA mutants on the nuclear lamina. We further used circular dichroism, fluorescence spectroscopy and isothermal titration calorimetry (ITC) to probe the structural modulations, self-association and heteropolymeric association of mutant LA. RESULTS Transfection of mutants in cultured cell lines result in the formation of nuclear aggregates of varied size and distribution. Endogenous LB1 is sequestered into these aggregates. This is consistent with the ten-fold increase in association constant of the mutant proteins compared to the wild type. These mutants exhibit differential heterotypic interaction with LB1, along with significant secondary and tertiary structural alterations of the interacting proteins. Thermodynamic studies demonstrate that the mutants bind to LB1 with different stoichiometry, affinity and energetics. CONCLUSIONS In this report we show that increased self-association propensity of mutant LA modulates the LA-LB1 interaction and precludes the formation of an otherwise uniform laminar network. GENERAL SIGNIFICANCE Our results might highlight the role of homotypic and heterotypic interactions of LA in the pathogenesis of DCM and hence laminopathies in the broader sense.
Collapse
Affiliation(s)
- Pritha Bhattacharjee
- Biophysics and Structural Genomics Division, Saha Institute of Nuclear Physics, 1/AF Bidhannagar, Kolkata 700064, India
| | - Dipak Dasgupta
- Biophysics and Structural Genomics Division, Saha Institute of Nuclear Physics, 1/AF Bidhannagar, Kolkata 700064, India.
| | - Kaushik Sengupta
- Biophysics and Structural Genomics Division, Saha Institute of Nuclear Physics, 1/AF Bidhannagar, Kolkata 700064, India.
| |
Collapse
|
215
|
Serebryannyy L, Misteli T. Protein sequestration at the nuclear periphery as a potential regulatory mechanism in premature aging. J Cell Biol 2017; 217:21-37. [PMID: 29051264 PMCID: PMC5748986 DOI: 10.1083/jcb.201706061] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Revised: 08/10/2017] [Accepted: 08/17/2017] [Indexed: 12/19/2022] Open
Abstract
Serebryannyy and Misteli provide a perspective on how protein sequestration at the inner nuclear membrane and nuclear lamina might influence aging. Despite the extensive description of numerous molecular changes associated with aging, insights into the driver mechanisms of this fundamental biological process are limited. Based on observations in the premature aging syndrome Hutchinson–Gilford progeria, we explore the possibility that protein regulation at the inner nuclear membrane and the nuclear lamina contributes to the aging process. In support, sequestration of nucleoplasmic proteins to the periphery impacts cell stemness, the response to cytotoxicity, proliferation, changes in chromatin state, and telomere stability. These observations point to the nuclear periphery as a central regulator of the aging phenotype.
Collapse
Affiliation(s)
| | - Tom Misteli
- National Cancer Institute, National Institutes of Health, Bethesda, MD
| |
Collapse
|
216
|
Pecorari I, Borin D, Sbaizero O. A Perspective on the Experimental Techniques for Studying Lamins. Cells 2017; 6:E33. [PMID: 28994747 PMCID: PMC5755493 DOI: 10.3390/cells6040033] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Revised: 10/01/2017] [Accepted: 10/05/2017] [Indexed: 01/29/2023] Open
Abstract
Lamins are type V intermediate filaments that collectively form a meshwork underneath the inner nuclear membrane, called nuclear lamina. Furthermore, they are also present in the nucleoplasm. Lamins are experiencing a growing interest, since a wide range of diseases are induced by mutations in the gene coding for A-type lamins, globally known as laminopathies. Moreover, it has been demonstrated that lamins are involved in other pathological conditions, like cancer. The role of lamins has been studied from several perspectives, exploiting different techniques and procedures. This multidisciplinary approach has contributed to resolving the unique features of lamins and has provided a thorough insight in their role in living organisms. Yet, there are still many unanswered questions, which constantly generate research in the field. The present work is aimed to review some interesting experimental techniques performed so far to study lamins. Scientists can take advantage of this collection for their novel investigations, being aware of the already pursued and consolidated methodologies. Hopefully, advances in these research directions will provide insights to achieve better diagnostic procedures and effective therapeutic options.
Collapse
Affiliation(s)
- Ilaria Pecorari
- Department of Engineering and Architecture, University of Trieste, Via Valerio 10, 34127 Trieste, Italy.
| | - Daniele Borin
- Department of Engineering and Architecture, University of Trieste, Via Valerio 10, 34127 Trieste, Italy.
| | - Orfeo Sbaizero
- Department of Engineering and Architecture, University of Trieste, Via Valerio 10, 34127 Trieste, Italy.
| |
Collapse
|
217
|
Discher DE, Smith L, Cho S, Colasurdo M, García AJ, Safran S. Matrix Mechanosensing: From Scaling Concepts in 'Omics Data to Mechanisms in the Nucleus, Regeneration, and Cancer. Annu Rev Biophys 2017; 46:295-315. [PMID: 28532215 DOI: 10.1146/annurev-biophys-062215-011206] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Many of the most important molecules of life are polymers. In animals, the most abundant of the proteinaceous polymers are the collagens, which constitute the fibrous matrix outside cells and which can also self-assemble into gels. The physically measurable stiffness of gels, as well as tissues, increases with the amount of collagen, and cells seem to sense this stiffness. An understanding of this mechanosensing process in complex tissues, including fibrotic disease states with high collagen, is now utilizing 'omics data sets and is revealing polymer physics-type, nonlinear scaling relationships between concentrations of seemingly unrelated biopolymers. The nuclear structure protein lamin A provides one example, with protein and transcript levels increasing with collagen 1 and tissue stiffness, and with mechanisms rooted in protein stabilization induced by cytoskeletal stress. Physics-based models of fibrous matrix, cytoskeletal force dipoles, and the lamin A gene circuit illustrate the wide range of testable predictions emerging for tissues, cell cultures, and even stem cell-based tissue regeneration. Beyond the epigenetics of mechanosensing, the scaling in cancer of chromosome copy number variations and other mutations with tissue stiffness suggests that genomic changes are occurring by mechanogenomic processes that now require elucidation.
Collapse
Affiliation(s)
- Dennis E Discher
- Molecular and Cell Biophysics Lab, University of Pennsylvania, Philadelphia, Pennsylvania 19104;
| | - Lucas Smith
- Molecular and Cell Biophysics Lab, University of Pennsylvania, Philadelphia, Pennsylvania 19104;
| | - Sangkyun Cho
- Molecular and Cell Biophysics Lab, University of Pennsylvania, Philadelphia, Pennsylvania 19104;
| | - Mark Colasurdo
- Parker H. Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, Georgia 30332
| | - Andrés J García
- Parker H. Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, Georgia 30332
| | - Sam Safran
- Department of Materials and Interfaces, Weizmann Institute of Science, Rehovet 76100, Israel
| |
Collapse
|
218
|
Prior R, Van Helleputte L, Benoy V, Van Den Bosch L. Defective axonal transport: A common pathological mechanism in inherited and acquired peripheral neuropathies. Neurobiol Dis 2017; 105:300-320. [DOI: 10.1016/j.nbd.2017.02.009] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Revised: 01/29/2017] [Accepted: 02/20/2017] [Indexed: 12/29/2022] Open
|
219
|
Norton HK, Phillips-Cremins JE. Crossed wires: 3D genome misfolding in human disease. J Cell Biol 2017; 216:3441-3452. [PMID: 28855250 PMCID: PMC5674879 DOI: 10.1083/jcb.201611001] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Revised: 06/09/2017] [Accepted: 08/16/2017] [Indexed: 12/13/2022] Open
Abstract
Norton and Phillips-Cremins review the 3D architecture of the genome and discuss links between chromatin misfolding and human diseases. Mammalian genomes are folded into unique topological structures that undergo precise spatiotemporal restructuring during healthy development. Here, we highlight recent advances in our understanding of how the genome folds inside the 3D nucleus and how these folding patterns are miswired during the onset and progression of mammalian disease states. We discuss potential mechanisms underlying the link among genome misfolding, genome dysregulation, and aberrant cellular phenotypes. We also discuss cases in which the endogenous 3D genome configurations in healthy cells might be particularly susceptible to mutation or translocation. Together, these data support an emerging model in which genome folding and misfolding is critically linked to the onset and progression of a broad range of human diseases.
Collapse
Affiliation(s)
- Heidi K Norton
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA
| | - Jennifer E Phillips-Cremins
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA .,Epigenetics Program, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
220
|
Ma Y, Fei L, Zhang M, Zhang W, Liu X, Wang C, Luo Y, Zhang H, Han Y. Lamin B2 binding to minichromosome maintenance complex component 7 promotes non-small cell lung carcinogenesis. Oncotarget 2017; 8:104813-104830. [PMID: 29285216 PMCID: PMC5739603 DOI: 10.18632/oncotarget.20338] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Accepted: 07/17/2017] [Indexed: 12/13/2022] Open
Abstract
We investigated the role of lamin B2 in non-small cell lung cancer (NSCLC). We detected higher lamin B2 expression in 20 NSCLC tumor tissues obtained from The Cancer Genome Atlas than in adjacent normal lung tissues. LMNB2-RNAi knockdown in A549 and H1299 NSCLC cells inhibited colony formation, cell proliferation and G1-S cell cycle progression while increasing apoptosis. LMNB2 overexpression had the opposite effects. Tumor xenograft experiments showed diminished tumor growth with LMNB2 knockdown H1299 cells than with controls. Yeast two-hybrid studies revealed minichromosome maintenance complex component 7 (MCM7) to be a binding partner of lamin B2, which was confirmed by co-immunoprecipitation and co-localization studies. Lamin B2 binding enhanced DNA binding and helicase activities of MCM7. Deletion analysis with MCM7-N, MCM7-M or MCM7-C mutant proteins showed that lamin B2 binds to the C-terminus of MCM7, and competes with the binding of the tumor suppressor retinoblastoma (RB) protein. Immunohistochemical analysis of 150 NSCLC patient samples revealed that both lamin B2 and MCM7 levels positively correlated with histological grade and tumor TNM stage. Moreover, high lamin B2 and MCM7 levels correlated with shorter overall survival of NSCLC patients. In sum, these results show that lamin B2 interaction with MCM7 promotes NSCLC progression.
Collapse
Affiliation(s)
- Yinan Ma
- Departments of Pathology, School of Basic Medical Sciences, China Medical University, Liaoning, China
| | - Liangru Fei
- Departments of Pathology, School of Basic Medical Sciences, China Medical University, Liaoning, China
| | - Meiyu Zhang
- Departments of Pathology, School of Basic Medical Sciences, China Medical University, Liaoning, China
| | - Wenzhu Zhang
- Departments of Pathology, School of Basic Medical Sciences, China Medical University, Liaoning, China
| | - Xiaofang Liu
- Department of Pathology, The First Affiliated Hospital of China Medical University, Liaoning, China
| | - Congcong Wang
- Department of Pathology, The First Affiliated Hospital of China Medical University, Liaoning, China
| | - Yuan Luo
- Departments of Pathology, School of Basic Medical Sciences, China Medical University, Liaoning, China
| | - Haiyan Zhang
- Department of Pathology, The First People's Hospital of Jining, Shandong, China
| | - Yuchen Han
- Departments of Pathology, School of Basic Medical Sciences, China Medical University, Liaoning, China.,Department of Pathology, The First Affiliated Hospital of China Medical University, Liaoning, China.,Department of Pathology, Shanghai Chest Hospital, Shanghai, China
| |
Collapse
|
221
|
Kubben N, Misteli T. Shared molecular and cellular mechanisms of premature ageing and ageing-associated diseases. Nat Rev Mol Cell Biol 2017; 18:595-609. [PMID: 28792007 DOI: 10.1038/nrm.2017.68] [Citation(s) in RCA: 178] [Impact Index Per Article: 25.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Ageing is the predominant risk factor for many common diseases. Human premature ageing diseases are powerful model systems to identify and characterize cellular mechanisms that underpin physiological ageing. Their study also leads to a better understanding of the causes, drivers and potential therapeutic strategies of common diseases associated with ageing, including neurological disorders, diabetes, cardiovascular diseases and cancer. Using the rare premature ageing disorder Hutchinson-Gilford progeria syndrome as a paradigm, we discuss here the shared mechanisms between premature ageing and ageing-associated diseases, including defects in genetic, epigenetic and metabolic pathways; mitochondrial and protein homeostasis; cell cycle; and stem cell-regenerative capacity.
Collapse
Affiliation(s)
- Nard Kubben
- National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Tom Misteli
- National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, USA
| |
Collapse
|
222
|
Fal K, Asnacios A, Chabouté ME, Hamant O. Nuclear envelope: a new frontier in plant mechanosensing? Biophys Rev 2017; 9:389-403. [PMID: 28801801 PMCID: PMC5578935 DOI: 10.1007/s12551-017-0302-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Accepted: 07/28/2017] [Indexed: 02/07/2023] Open
Abstract
In animals, it is now well established that forces applied at the cell surface are propagated through the cytoskeleton to the nucleus, leading to deformations of the nuclear structure and, potentially, to modification of gene expression. Consistently, altered nuclear mechanics has been related to many genetic disorders, such as muscular dystrophy, cardiomyopathy and progeria. In plants, the integration of mechanical signals in cell and developmental biology has also made great progress. Yet, while the link between cell wall stresses and cytoskeleton is consolidated, such cortical mechanical cues have not been integrated with the nucleoskeleton. Here, we propose to take inspiration from studies on animal nuclei to identify relevant methods amenable to probing nucleus mechanics and deformation in plant cells, with a focus on microrheology. To identify potential molecular targets, we also compare the players at the nuclear envelope, namely lamina and LINC complex, in both plant and animal nuclei. Understanding how mechanical signals are transduced to the nucleus across kingdoms will likely have essential implications in development (e.g. how mechanical cues add robustness to gene expression patterns), in the nucleoskeleton-cytoskeleton nexus (e.g. how stress is propagated in turgid/walled cells), as well as in transcriptional control, chromatin biology and epigenetics.
Collapse
Affiliation(s)
- Kateryna Fal
- Laboratoire Reproduction et Développement des Plantes, Université de Lyon, ENS de Lyon, UCB Lyon 1, CNRS, INRA, 69342, Lyon, France
| | - Atef Asnacios
- Laboratoire Matières et Systèmes Complexes, Université Paris-Diderot and CNRS, UMR 7057, Sorbonne Paris Cité, Paris, France
| | - Marie-Edith Chabouté
- Institut de Biologie Moléculaire des Plantes, CNRS, Université de Strasbourg, 67000, Strasbourg, France
| | - Olivier Hamant
- Laboratoire Reproduction et Développement des Plantes, Université de Lyon, ENS de Lyon, UCB Lyon 1, CNRS, INRA, 69342, Lyon, France.
| |
Collapse
|
223
|
Patel S, Rauf A, Meher BR. In silico analysis of ChtBD3 domain to find its role in bacterial pathogenesis and beyond. Microb Pathog 2017; 110:519-526. [PMID: 28760454 DOI: 10.1016/j.micpath.2017.07.047] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2016] [Revised: 07/24/2017] [Accepted: 07/27/2017] [Indexed: 12/12/2022]
Abstract
Chitin binding domain 3, known by the acronym ChtBD3, is a domain in the enzymes and proteins of several pathogenic virus, bacteria and fungi. As this domain is evolutionarily-conserved in virulence factors of these infectious agents, its detailed investigation is of clinical interest. In this regard, the current in silico study analyzed ChtBD3 domain distribution in bacterial proteins present in publicly-available SMART (simple modular architecture research tool) database. Also, the co-occurring domains of ChtBD3 in the studied proteins were mapped to understand positional rearrangement of the domain and consequent functional diversity. Custom-made scripts were used to interpret the data and to derive patterns. As expected, interesting results were obtained. ChtBD3 domain co-occurred with other critical domains like peptidase, glycol_hydrolase, kinase, hemagglutinin-acting, collagen-binding, among others. The findings are expected to be of clinical relevance.
Collapse
Affiliation(s)
- Seema Patel
- Bioinformatics and Medical Informatics Research Center, San Diego State University, San Diego, 92182, USA.
| | - Abdur Rauf
- Department of Chemistry, University of Swabi, Anbar, 23561, Khyber Pakhtunkhwa, Pakistan.
| | - Biswa Ranjan Meher
- Centre for Life Sciences, Central University of Jharkhand, Brambe, Ranchi, 835205, Jharkhand, India
| |
Collapse
|
224
|
Abstract
The nuclear lamina is involved in fundamental nuclear functions and provides mechanical stability to the nucleus. Lamin filaments form a meshwork closely apposed to the inner nuclear membrane and a small fraction of lamins exist in the nuclear interior. Mutations in lamin genes cause severe hereditary diseases, the laminopathies. During vertebrate evolution the lamin protein family has expanded. While most vertebrate genomes contain 4 lamin genes, encoding the lamins A, B1, B2, and LIII, the majority of non-vertebrate genomes harbor only a single lamin gene. We have collected lamin gene and cDNA sequence information for representatives of the major vertebrate lineages. With the help of RNA-seq data we have determined relative lamin expression levels for representative tissues for species of 9 different gnathostome lineages. Here we report that the level of lamin A expression is low in cartilaginous fishes and ancient fishes and increases toward the mammals. Lamin B1 expression shows an inverse tendency to that of lamin A. Possible implications for the change in the lamin A to B ratio is discussed in the light of its role in nuclear mechanics.
Collapse
Affiliation(s)
- Reimer Stick
- a FB2 Biology/Chemistry, University of Bremen , Bremen , Germany
| | - Annette Peter
- a FB2 Biology/Chemistry, University of Bremen , Bremen , Germany
| |
Collapse
|
225
|
Wang X, Zabell A, Koh W, Tang WHW. Lamin A/C Cardiomyopathies: Current Understanding and Novel Treatment Strategies. CURRENT TREATMENT OPTIONS IN CARDIOVASCULAR MEDICINE 2017; 19:21. [PMID: 28299614 DOI: 10.1007/s11936-017-0520-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
OPINION STATEMENT Dilated cardiomyopathy (DCM) is the third leading cause of heart failure in the USA. A major gene associated with DCM with cardiac conduction system disease is lamin A/C (LMNA) gene. Lamins are type V filaments that serve a variety of roles, including nuclear structure support, DNA repair, cell signaling pathway mediation, and chromatin organization. In 1999, LMNA was found responsible for Emery-Dreifuss muscular dystrophy (EDMD) and, since then, has been found in association with a wide spectrum of diseases termed laminopathies, including LMNA cardiomyopathy. Patients with LMNA mutations have a poor prognosis and a higher risk for sudden cardiac death, along with other cardiac effects like dysrhythmias, development of congestive heart failure, and potential need of a pacemaker or ICD. As of now, there is no specific treatment for laminopathies, including LMNA cardiomyopathy, because the mechanism of LMNA mutations in humans is still unclear. This review discusses LMNA mutations and how they relate to DCM, the necessity for further investigation to better understand LMNA mutations, and potential treatment options ranging from clinical and therapeutic to cellular and molecular techniques.
Collapse
Affiliation(s)
- Xi Wang
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland, OH, USA
| | - Allyson Zabell
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland, OH, USA
| | - Wonshill Koh
- Children's Hospital of Pittsburgh, Children's Hospital of Pittsburgh, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - W H Wilson Tang
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland, OH, USA. .,Department of Cardiovascular Medicine, Heart and Vascular Institute, Cleveland Clinic, Cleveland, OH, USA. .,Center for Clinical Genomics, Cleveland Clinic, Cleveland, OH, USA.
| |
Collapse
|
226
|
Rout MP, Obado SO, Schenkman S, Field MC. Specialising the parasite nucleus: Pores, lamins, chromatin, and diversity. PLoS Pathog 2017; 13:e1006170. [PMID: 28253370 PMCID: PMC5333908 DOI: 10.1371/journal.ppat.1006170] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Affiliation(s)
- Michael P. Rout
- The Rockefeller University, New York, New York, United States of America
| | - Samson O. Obado
- The Rockefeller University, New York, New York, United States of America
| | | | - Mark C. Field
- Wellcome Centre for Anti-Infectives Research, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| |
Collapse
|
227
|
Turgay Y, Eibauer M, Goldman AE, Shimi T, Khayat M, Ben-Harush K, Dubrovsky-Gaupp A, Sapra KT, Goldman RD, Medalia O. The molecular architecture of lamins in somatic cells. Nature 2017; 543:261-264. [PMID: 28241138 DOI: 10.1038/nature21382] [Citation(s) in RCA: 276] [Impact Index Per Article: 39.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Accepted: 01/11/2017] [Indexed: 01/13/2023]
Abstract
The nuclear lamina is a fundamental constituent of metazoan nuclei. It is composed mainly of lamins, which are intermediate filament proteins that assemble into a filamentous meshwork, bridging the nuclear envelope and chromatin. Besides providing structural stability to the nucleus, the lamina is involved in many nuclear activities, including chromatin organization, transcription and replication. However, the structural organization of the nuclear lamina is poorly understood. Here we use cryo-electron tomography to obtain a detailed view of the organization of the lamin meshwork within the lamina. Data analysis of individual lamin filaments resolves a globular-decorated fibre appearance and shows that A- and B-type lamins assemble into tetrameric filaments of 3.5 nm thickness. Thus, lamins exhibit a structure that is remarkably different from the other canonical cytoskeletal elements. Our findings define the architecture of the nuclear lamin meshworks at molecular resolution, providing insights into their role in scaffolding the nuclear lamina.
Collapse
Affiliation(s)
- Yagmur Turgay
- Department of Biochemistry, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Matthias Eibauer
- Department of Biochemistry, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Anne E Goldman
- Department of Cell and Molecular Biology, Northwestern University Feinberg School of Medicine, 303 E Chicago Avenue, Chicago, Illinois 60611, USA
| | - Takeshi Shimi
- Department of Cell and Molecular Biology, Northwestern University Feinberg School of Medicine, 303 E Chicago Avenue, Chicago, Illinois 60611, USA
| | - Maayan Khayat
- Department of Life Sciences and the National Institute for Biotechnology in the Negev, Ben-Gurion University, 84105 Beer-Sheva, Israel
| | - Kfir Ben-Harush
- Department of Chemical Engineering, Shamoon College of Engineering, Jabotinsky 84, 77245 Ashdod, Israel
| | - Anna Dubrovsky-Gaupp
- Department of Biochemistry, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - K Tanuj Sapra
- Department of Biochemistry, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Robert D Goldman
- Department of Cell and Molecular Biology, Northwestern University Feinberg School of Medicine, 303 E Chicago Avenue, Chicago, Illinois 60611, USA
| | - Ohad Medalia
- Department of Biochemistry, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland.,Department of Life Sciences and the National Institute for Biotechnology in the Negev, Ben-Gurion University, 84105 Beer-Sheva, Israel
| |
Collapse
|
228
|
Collins CM, Ellis JA, Holaska JM. MAPK signaling pathways and HDAC3 activity are disrupted during differentiation of emerin-null myogenic progenitor cells. Dis Model Mech 2017; 10:385-397. [PMID: 28188262 PMCID: PMC5399572 DOI: 10.1242/dmm.028787] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Accepted: 02/01/2017] [Indexed: 01/28/2023] Open
Abstract
Mutations in the gene encoding emerin cause Emery–Dreifuss muscular dystrophy (EDMD). Emerin is an integral inner nuclear membrane protein and a component of the nuclear lamina. EDMD is characterized by skeletal muscle wasting, cardiac conduction defects and tendon contractures. The failure to regenerate skeletal muscle is predicted to contribute to the skeletal muscle pathology of EDMD. We hypothesize that muscle regeneration defects are caused by impaired muscle stem cell differentiation. Myogenic progenitors derived from emerin-null mice were used to confirm their impaired differentiation and analyze selected myogenic molecular pathways. Emerin-null progenitors were delayed in their cell cycle exit, had decreased myosin heavy chain (MyHC) expression and formed fewer myotubes. Emerin binds to and activates histone deacetylase 3 (HDAC3). Here, we show that theophylline, an HDAC3-specific activator, improved myotube formation in emerin-null cells. Addition of the HDAC3-specific inhibitor RGFP966 blocked myotube formation and MyHC expression in wild-type and emerin-null myogenic progenitors, but did not affect cell cycle exit. Downregulation of emerin was previously shown to affect the p38 MAPK and ERK/MAPK pathways in C2C12 myoblast differentiation. Using a pure population of myogenic progenitors completely lacking emerin expression, we show that these pathways are also disrupted. ERK inhibition improved MyHC expression in emerin-null cells, but failed to rescue myotube formation or cell cycle exit. Inhibition of p38 MAPK prevented differentiation in both wild-type and emerin-null progenitors. These results show that each of these molecular pathways specifically regulates a particular stage of myogenic differentiation in an emerin-dependent manner. Thus, pharmacological targeting of multiple pathways acting at specific differentiation stages may be a better therapeutic approach in the future to rescue muscle regeneration in vivo. Editors' choice: HDAC3, p38 MAPK and ERK signaling are altered during differentiation of myogenic progenitors lacking emerin; pharmacological activation or inhibition of these signaling proteins rescues specific stages of myogenic differentiation.
Collapse
Affiliation(s)
- Carol M Collins
- University of the Sciences, Department of Pharmaceutical Sciences, 600 S. 43rd St, Philadelphia, PA 19104, USA
| | - Joseph A Ellis
- University of the Sciences, Department of Pharmaceutical Sciences, 600 S. 43rd St, Philadelphia, PA 19104, USA
| | - James M Holaska
- University of the Sciences, Department of Pharmaceutical Sciences, 600 S. 43rd St, Philadelphia, PA 19104, USA
| |
Collapse
|
229
|
Cho S, Irianto J, Discher DE. Mechanosensing by the nucleus: From pathways to scaling relationships. J Cell Biol 2017; 216:305-315. [PMID: 28043971 PMCID: PMC5294790 DOI: 10.1083/jcb.201610042] [Citation(s) in RCA: 238] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Revised: 12/05/2016] [Accepted: 12/14/2016] [Indexed: 01/01/2023] Open
Abstract
The nucleus is linked mechanically to the extracellular matrix via multiple polymers that transmit forces to the nuclear envelope and into the nuclear interior. Here, we review some of the emerging mechanisms of nuclear mechanosensing, which range from changes in protein conformation and transcription factor localization to chromosome reorganization and membrane dilation up to rupture. Nuclear mechanosensing encompasses biophysically complex pathways that often converge on the main structural proteins of the nucleus, the lamins. We also perform meta-analyses of public transcriptomics and proteomics data, which indicate that some of the mechanosensing pathways relaying signals from the collagen matrix to the nucleus apply to a broad range of species, tissues, and diseases.
Collapse
Affiliation(s)
- Sangkyun Cho
- Molecular and Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA 19104
| | - Jerome Irianto
- Molecular and Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA 19104
| | - Dennis E Discher
- Molecular and Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA 19104
| |
Collapse
|
230
|
Sewitz SA, Fahmi Z, Lipkow K. Higher order assembly: folding the chromosome. Curr Opin Struct Biol 2017; 42:162-168. [DOI: 10.1016/j.sbi.2017.02.004] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Accepted: 02/13/2017] [Indexed: 11/28/2022]
|
231
|
A Novel Lamin A Mutant Responsible for Congenital Muscular Dystrophy Causes Distinct Abnormalities of the Cell Nucleus. PLoS One 2017; 12:e0169189. [PMID: 28125586 PMCID: PMC5268432 DOI: 10.1371/journal.pone.0169189] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Accepted: 12/13/2016] [Indexed: 11/26/2022] Open
Abstract
A-type lamins, the intermediate filament proteins participating in nuclear structure and function, are encoded by LMNA. LMNA mutations can lead to laminopathies such as lipodystrophies, premature aging syndromes (progeria) and muscular dystrophies. Here, we identified a novel heterozygous LMNA p.R388P de novo mutation in a patient with a non-previously described severe phenotype comprising congenital muscular dystrophy (L-CMD) and lipodystrophy. In culture, the patient’s skin fibroblasts entered prematurely into senescence, and some nuclei showed a lamina honeycomb pattern. C2C12 myoblasts were transfected with a construct carrying the patient’s mutation; R388P-lamin A (LA) predominantly accumulated within the nucleoplasm and was depleted at the nuclear periphery, altering the anchorage of the inner nuclear membrane protein emerin and the nucleoplasmic protein LAP2-alpha. The mutant LA triggered a frequent and severe nuclear dysmorphy that occurred independently of prelamin A processing, as well as increased histone H3K9 acetylation. Nuclear dysmorphy was not significantly improved when transfected cells were treated with drugs disrupting microtubules or actin filaments or modifying the global histone acetylation pattern. Therefore, releasing any force exerted at the nuclear envelope by the cytoskeleton or chromatin did not rescue nuclear shape, in contrast to what was previously shown in Hutchinson-Gilford progeria due to other LMNA mutations. Our results point to the specific cytotoxic effect of the R388P-lamin A mutant, which is clinically related to a rare and severe multisystemic laminopathy phenotype.
Collapse
|
232
|
Patel S, Goyal A. Chitin and chitinase: Role in pathogenicity, allergenicity and health. Int J Biol Macromol 2017; 97:331-338. [PMID: 28093332 DOI: 10.1016/j.ijbiomac.2017.01.042] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Revised: 01/05/2017] [Accepted: 01/10/2017] [Indexed: 01/09/2023]
Abstract
Chitin, a polysaccharide with particular abundance in fungi, nematodes and arthropods is immunogenic. It acts as a threat to other organisms, to tackle which they have been endowed with chitinase enzyme. Even if this enzyme is not present in all organisms, they possess proteins having chitin-binding domain(s) (ChtBD). Many lethal viruses like Ebola, and HCV (Hepatitis C virus) have these domains to manipulate their carriers and target organisms. In keeping with the basic rule of survival, the self-origin (own body component) chitins and chitinases are protective, but that of non-self origin (from other organisms) are detrimental to health. The exogenous chitins and chitinases provoke human innate immunity to generate a deluge of inflammatory cytokines, which injure organs (leading to asthma, atopic dermatitis etc.), and in persistent situations lead to death (multiple sclerosis, systemic lupus erythromatosus (SLE), cancer, etc.). Unfortunately, chitin-chitinase-stimulated hypersensitivity is a common cause of occupational allergy. On the other hand, chitin, and its deacetylated derivative chitosan are increasingly proving useful in pharmaceutical, agriculture, and biocontrol applications. This critical review discusses the complex nexus of chitin and chitinase and assesses both their pathogenic as well as utilitarian aspects.
Collapse
Affiliation(s)
- Seema Patel
- Bioinformatics and Medical Informatics Research Center, San Diego State University, 5500 Campanile Dr, San Diego, CA 92182, USA.
| | - Arun Goyal
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati 781039, Assam, India.
| |
Collapse
|
233
|
Dorado B, Andrés V. A-type lamins and cardiovascular disease in premature aging syndromes. Curr Opin Cell Biol 2017; 46:17-25. [PMID: 28086161 DOI: 10.1016/j.ceb.2016.12.005] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Revised: 12/14/2016] [Accepted: 12/21/2016] [Indexed: 01/17/2023]
Abstract
Lamin A is a nuclear intermediate filament protein with important structural and regulatory roles in most differentiated mammalian cells. Excessive accumulation of its precursor prelamin A or the mutant form called 'progerin' causes premature aging syndromes. Progeroid 'laminopathies' are characterized by severe cardiovascular problems (cardiac electrical defects, vascular calcification and stiffening, atherosclerosis, myocardial infarction, and stroke) and premature death. Here, we review studies in cell and mouse models and patients that are unraveling how abnormal prelamin A and progerin accumulation accelerates cardiovascular disease and aging. This knowledge is essential for developing effective therapies to treat progeria and may help identify new mechanisms underlying normal aging.
Collapse
Affiliation(s)
- Beatriz Dorado
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), CIBER de Enfermedades Cardiovasculares, Madrid, Spain
| | - Vicente Andrés
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), CIBER de Enfermedades Cardiovasculares, Madrid, Spain.
| |
Collapse
|
234
|
Cohen-Fix O, Askjaer P. Cell Biology of the Caenorhabditis elegans Nucleus. Genetics 2017; 205:25-59. [PMID: 28049702 PMCID: PMC5216270 DOI: 10.1534/genetics.116.197160] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Accepted: 11/09/2016] [Indexed: 12/25/2022] Open
Abstract
Studies on the Caenorhabditis elegans nucleus have provided fascinating insight to the organization and activities of eukaryotic cells. Being the organelle that holds the genetic blueprint of the cell, the nucleus is critical for basically every aspect of cell biology. The stereotypical development of C. elegans from a one cell-stage embryo to a fertile hermaphrodite with 959 somatic nuclei has allowed the identification of mutants with specific alterations in gene expression programs, nuclear morphology, or nuclear positioning. Moreover, the early C. elegans embryo is an excellent model to dissect the mitotic processes of nuclear disassembly and reformation with high spatiotemporal resolution. We review here several features of the C. elegans nucleus, including its composition, structure, and dynamics. We also discuss the spatial organization of chromatin and regulation of gene expression and how this depends on tight control of nucleocytoplasmic transport. Finally, the extensive connections of the nucleus with the cytoskeleton and their implications during development are described. Most processes of the C. elegans nucleus are evolutionarily conserved, highlighting the relevance of this powerful and versatile model organism to human biology.
Collapse
Affiliation(s)
- Orna Cohen-Fix
- Laboratory of Molecular and Cellular Biology, National Institute of Diabetes, Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892
| | - Peter Askjaer
- Andalusian Center for Developmental Biology, Consejo Superior de Investigaciones Científicas/Junta de Andalucia/Universidad Pablo de Olavide, 41013 Seville, Spain
| |
Collapse
|
235
|
Abstract
Intermediate filaments (IFs), together with microtubules and actin microfilaments, are the three main cytoskeletal components in metazoan cells. IFs are formed by a distinct protein family, which is made up of 70 members in humans. Most IF proteins are tissue- or organelle-specific, which includes lamins, the IF proteins of the nucleus. The building block of IFs is an elongated dimer, which consists of a central α-helical 'rod' domain flanked by flexible N- and C-terminal domains. The conserved rod domain is the 'signature feature' of the IF family. Bioinformatics analysis reveals that the rod domain of all IF proteins contains three α-helical segments of largely conserved length, interconnected by linkers. Moreover, there is a conserved pattern of hydrophobic repeats within each segment, which includes heptads and hendecads. This defines the presence of both left-handed and almost parallel coiled-coil regions along the rod length. Using X-ray crystallography on multiple overlapping fragments of IF proteins, the atomic structure of the nearly complete rod domain has been determined. Here, we discuss some specific challenges of this procedure, such as crystallization and diffraction data phasing by molecular replacement. Further insights into the structure of the coiled coil and the terminal domains have been obtained using electron paramagnetic resonance measurements on the full-length protein, with spin labels attached at specific positions. This atomic resolution information, as well as further interesting findings, such as the variation of the coiled-coil stability along the rod length, provide clues towards interpreting the data on IF assembly, collected by a range of methods. However, a full description of this process at the molecular level is not yet at hand.
Collapse
Affiliation(s)
- Dmytro Guzenko
- Laboratory for Biocrystallography, Department of Pharmaceutical and Pharmacological Sciences, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Anastasia A Chernyatina
- Laboratory for Biocrystallography, Department of Pharmaceutical and Pharmacological Sciences, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Sergei V Strelkov
- Laboratory for Biocrystallography, Department of Pharmaceutical and Pharmacological Sciences, Katholieke Universiteit Leuven, Leuven, Belgium.
| |
Collapse
|
236
|
Abstract
For years intermediate filaments (IF), belonging to the third class of filamentous cytoskeletal proteins alongside microtubules and actin filaments, were thought to be exclusive to metazoan cells. Structurally these eukaryote IFs are very well defined, consisting of globular head and tail domains, which flank the central rod-domain. This central domain is dominated by an α-helical secondary structure predisposed to form the characteristic coiled-coil, parallel homo-dimer. These elementary dimers can further associate, both laterally and longitudinally, generating a variety of filament-networks built from filaments in the range of 10 nm in diameter. The general role of these filaments with their characteristic mechano-elastic properties both in the cytoplasm and in the nucleus of eukaryote cells is to provide mechanical strength and a scaffold supporting diverse shapes and cellular functions.Since 2003, after the first bacterial IF-like protein, crescentin was identified, it has been evident that bacteria also employ filamentous networks, other than those built from bacterial tubulin or actin homologues, in order to support their cell shape, growth and, in some cases, division. Intriguingly, compared to their eukaryote counterparts, the group of bacterial IF-like proteins shows much wider structural diversity. The sizes of both the head and tail domains are markedly reduced and there is great variation in the length of the central rod-domain. Furthermore, bacterial rod-domains often lack the sub-domain organisation of eukaryote IFs that is the defining feature of the IF-family. However, the fascinating display of filamentous assemblies, including rope, striated cables and hexagonal laces together with the conditions required for their formation both in vitro and in vivo strongly resemble that of eukaryote IFs suggesting that these bacterial proteins are deservedly classified as part of the IF-family and that the current definition should be relaxed slightly to allow their inclusion. The lack of extensive head and tail domains may well make the bacterial proteins more amenable for structural characterisation, which will be essential for establishing the mechanism for their association into filaments. What is more, the well-developed tools for bacterial manipulations provide an excellent opportunity of studying the bacterial systems with the prospect of making significant progress in our understanding of the general underlying principles of intermediate filament assemblies.
Collapse
Affiliation(s)
- Gabriella H Kelemen
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, UK.
| |
Collapse
|
237
|
Zhang W, Zhang C, Chen P, Yang C, Gan X, Hussain M, Xun Y, Tian Y, Du H. Circulation autoantibody against Lamin A/C in patients with Sjögren's syndrome. Oncotarget 2016; 7:80252-80261. [PMID: 27835913 PMCID: PMC5348317 DOI: 10.18632/oncotarget.13256] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Accepted: 10/25/2016] [Indexed: 01/17/2023] Open
Abstract
Lamin A/C proteins are major components of nuclear laminae and were encoded by the LMNA gene. Recent studies have found that in addition to provides nuclear-membrane strength; it also regulates the gene expression. Lamin A/C has been confirmed as an autoantigen in RA, SLE and vasculitis. Anti-Lamin A/C antibodies also have been found by indirect immunofluorescence method. In this study, we used various research methods to confirm Lamin A/C is an autoantigen in Han Chinese patients with confirmed Sjögren's syndrome (SS). To further investigate the relationship between the autoimmune disease antigens, we compared the amino acid sequence of Lamin A/C epitope and several common antigens' antigenic determinant. As a result, we found that Lamin A/C has similar epitopes with U1RNP. It means that the potential relationship exist between Lamin A/C and U1RNP. Clinical data we collected also showed that anti-Lamin A/C and anti-U1RNP antibodies always appear in same serum sample. Therefore, we speculated that cross-reaction may take place between antigen and potential antigen, which have similar epitope. Then, by epitope spreading, the potential antigen can be a new autoantigen. Our study provided a new thinking for further research about the relationship between autoantigens and their development mechanism in autoimmune diseases.
Collapse
Affiliation(s)
- Wen Zhang
- 112 Lab, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing, China
| | - Chunyan Zhang
- Department of Clinical Biochemistry, Chinese PLA General Hospital, Beijing, China
| | - Peng Chen
- 112 Lab, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing, China
| | - Chunhe Yang
- 112 Lab, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing, China
| | - Xianfeng Gan
- 112 Lab, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing, China
| | - Muhammad Hussain
- 112 Lab, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing, China
| | - Yiping Xun
- 112 Lab, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing, China
| | - Yaping Tian
- Department of Clinical Biochemistry, Chinese PLA General Hospital, Beijing, China
| | - Hongwu Du
- 112 Lab, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing, China
| |
Collapse
|
238
|
Cobb AM, Larrieu D, Warren DT, Liu Y, Srivastava S, Smith AJO, Bowater RP, Jackson SP, Shanahan CM. Prelamin A impairs 53BP1 nuclear entry by mislocalizing NUP153 and disrupting the Ran gradient. Aging Cell 2016; 15:1039-1050. [PMID: 27464478 PMCID: PMC5114580 DOI: 10.1111/acel.12506] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/28/2016] [Indexed: 01/29/2023] Open
Abstract
The nuclear lamina is essential for the proper structure and organization of the nucleus. Deregulation of A-type lamins can compromise genomic stability, alter chromatin organization and cause premature vascular aging. Here, we show that accumulation of the lamin A precursor, prelamin A, inhibits 53BP1 recruitment to sites of DNA damage and increases basal levels of DNA damage in aged vascular smooth muscle cells. We identify that this genome instability arises through defective nuclear import of 53BP1 as a consequence of abnormal topological arrangement of nucleoporin NUP153. We show for the first time that this nucleoporin is important for the nuclear localization of Ran and that the deregulated Ran gradient is likely to be compromising the nuclear import of 53BP1. Importantly, many of the defects associated with prelamin A expression were significantly reduced upon treatment with Remodelin, a small molecule recently reported to reverse deficiencies associated with abnormal nuclear lamina.
Collapse
Affiliation(s)
- Andrew M. Cobb
- The James Black CentreKing's College London125 Coldharbour LaneLondonSE5 9NUUK
| | - Delphine Larrieu
- Wellcome Trust/Cancer Research UK Gurdon InstituteThe Henry Wellcome Building of Cancer and Developmental BiologyUniversity of CambridgeTennis Court RoadCambridgeCB2 1QNUK
| | - Derek T. Warren
- The James Black CentreKing's College London125 Coldharbour LaneLondonSE5 9NUUK
| | - Yiwen Liu
- The James Black CentreKing's College London125 Coldharbour LaneLondonSE5 9NUUK
| | - Sonal Srivastava
- The James Black CentreKing's College London125 Coldharbour LaneLondonSE5 9NUUK
| | | | | | - Stephen P. Jackson
- Wellcome Trust/Cancer Research UK Gurdon InstituteThe Henry Wellcome Building of Cancer and Developmental BiologyUniversity of CambridgeTennis Court RoadCambridgeCB2 1QNUK
| | | |
Collapse
|
239
|
Zhang X, Hu J, Chen Y. Betulinic acid and the pharmacological effects of tumor suppression (Review). Mol Med Rep 2016; 14:4489-4495. [PMID: 27748864 DOI: 10.3892/mmr.2016.5792] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2015] [Accepted: 09/21/2016] [Indexed: 11/06/2022] Open
Abstract
Betulinic acid (BA), a lupane-type pentacyclic triterpenoid saponin from tree bark, has the potential to induce the apoptosis of cancer cells without toxicity towards normal cells in vitro and in vivo. The antitumor pharmacological effects of BA consist of triggering apoptosis via the mitochondrial pathway, regulating the cell cycle and the angiogenic pathway via factors, including specificity protein transcription factors, cyclin D1 and epidermal growth factor receptor, inhibiting the signal transducer and activator of transcription 3 and nuclear factor‑κB signaling pathways, preventing the invasion and metastasis of tumor cells, and affecting the expression of topoisomerase I, p53 and lamin B1. In previous years, several studies have shown its antitumor effect, initially applied to malignant melanoma, however, it also has broad efficacies against most solid types of tumor from different regions of the body. There have been few investigations in hematological malignancies, however, this direction may offer potential in such a novel field of research. In this review, the primary pharmacological effects of BA in tumors, particularly in hematological malignancies are discussed.
Collapse
Affiliation(s)
- Xia Zhang
- Department of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Jingyu Hu
- Department of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Yan Chen
- Department of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| |
Collapse
|
240
|
The assembly of C. elegans lamins into macroscopic fibers. J Mech Behav Biomed Mater 2016; 63:35-43. [DOI: 10.1016/j.jmbbm.2016.05.037] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Revised: 04/23/2016] [Accepted: 05/28/2016] [Indexed: 11/18/2022]
|
241
|
Apte K, Stick R, Radmacher M. Mechanics in human fibroblasts and progeria: Lamin A mutation E145K results in stiffening of nuclei. J Mol Recognit 2016; 30. [PMID: 27677907 DOI: 10.1002/jmr.2580] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Revised: 08/18/2016] [Accepted: 08/19/2016] [Indexed: 12/12/2022]
Abstract
The lamina is a filamentous meshwork beneath the inner nuclear membrane that confers mechanical stability to nuclei. The E145K mutation in lamin A causes Hutchinson-Gilford progeria syndrome (HGPS). It affects lamin filament assembly and induces profound changes in the nuclear architecture. Expression of wild-type and E145K lamin A in Xenopus oocytes followed by atomic force microscopy (AFM) probing of isolated oocyte nuclei has shown significant changes in the mechanical properties of the lamina. Nuclei of oocytes expressing E145K lamin A are stiffer than those expressing wild-type lamin A. Here we present mechanical measurements by AFM on dermal fibroblasts obtained from a 4-year-old progeria patient bearing the E145K lamin A mutation and compared it to fibroblasts obtained from 2 healthy donors of 10 and 61 years of age, respectively. The abnormal shape of nuclei expressing E145K lamin A was analyzed by fluorescence microscopy. Lamina thickness was measured using electron micrographs. Fluorescence microscopy showed alterations in the actin network of progeria cells. AFM probing of whole dermal fibroblasts did not demonstrate significant differences in the elastic moduli of nuclear and cytoplasmic cell regions. In contrast, AFM measurements of isolated nuclei showed that nuclei of progeria and old person's cells are significantly stiffer than those of the young person, indicating that the process of aging, be it natural or abnormal, increases nuclear stiffness. Our results corroborate AFM data obtained using Xenopus oocyte nuclei and prove that the presence of E145K lamin A abnormally increases nuclear stiffness.
Collapse
Affiliation(s)
- Ketaki Apte
- Department for Cell Biology, University of Bremen, 28359, Bremen, Germany
| | - Reimer Stick
- Department for Cell Biology, University of Bremen, 28359, Bremen, Germany
| | | |
Collapse
|
242
|
Zuela N, Dorfman J, Gruenbaum Y. Global transcriptional changes caused by an EDMD mutation correlate to tissue specific disease phenotypes in C. elegans. Nucleus 2016; 8:60-69. [PMID: 27673727 DOI: 10.1080/19491034.2016.1238999] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
There are numerous heritable diseases associated with mutations in the LMNA gene. Most of these laminopathic diseases, including several muscular dystrophies, are autosomal dominant and have tissue-specific phenotypes. Our previous studies have shown that the globally expressed Emery-Dreifuss muscular dystrophy (EDMD)-linked lamin mutation, L535P, disrupts nuclear mechanical response specifically in muscle nuclei of C. elegans leading to atrophy of the body muscle cells and to reduced motility. Here we used RNA sequencing to analyze the global changes in gene expression caused by the L535P EDMD lamin mutation in order to gain better understanding of disease mechanisms and the correlation between transcription and phenotype. Our results show changes in key genes and biological pathways that can help explain the muscle specific phenotypes. In addition, the differential gene expression between wild-type and L535P mutant animals suggests that the pharynx function in the L535P mutant animals is affected by this lamin mutation. Moreover, these transcriptional changes were then correlated with reduced pharynx activity and abnormal pharynx muscle structure. Understanding disease mechanisms will potentially lead to new therapeutic approaches toward curing EDMD.
Collapse
Affiliation(s)
- Noam Zuela
- a Department of Genetics, Institute of Life Sciences, Hebrew University of Jerusalem , Jerusalem , Israel
| | | | - Yosef Gruenbaum
- a Department of Genetics, Institute of Life Sciences, Hebrew University of Jerusalem , Jerusalem , Israel
| |
Collapse
|
243
|
Abstract
The nucleus is separated from the cytosol by the nuclear envelope, which is a double lipid bilayer composed of the outer nuclear membrane and the inner nuclear membrane. The intermediate filament proteins lamin A, lamin B, and lamin C form a network underlying the inner nuclear membrane. This proteinaceous network provides the nucleus with its strength, rigidity, and elasticity. Positioned within the inner nuclear membrane are more than 150 inner nuclear membrane proteins, many of which interact directly with lamins and require lamins for their inner nuclear membrane localization. Inner nuclear membrane proteins and the nuclear lamins define the nuclear lamina. These inner nuclear membrane proteins have tissue-specific expression and diverse functions including regulating cytoskeletal organization, nuclear architecture, cell cycle dynamics, and genomic organization. Loss or mutations in lamins and inner nuclear membrane proteins cause a wide spectrum of diseases. Here, I will review the functions of the well-studied nuclear lamina proteins and the diseases associated with loss or mutations in these proteins. © 2016 American Physiological Society. Compr Physiol 6:1655-1674, 2016.
Collapse
Affiliation(s)
- James M. Holaska
- Department of Pharmaceutical Sciences, University of the Sciences, Philadelphia, Pennsylvania, USA
| |
Collapse
|
244
|
Maishman L, Obado SO, Alsford S, Bart JM, Chen WM, Ratushny AV, Navarro M, Horn D, Aitchison JD, Chait BT, Rout MP, Field MC. Co-dependence between trypanosome nuclear lamina components in nuclear stability and control of gene expression. Nucleic Acids Res 2016; 44:10554-10570. [PMID: 27625397 PMCID: PMC5159534 DOI: 10.1093/nar/gkw751] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Revised: 08/02/2016] [Accepted: 08/20/2016] [Indexed: 12/17/2022] Open
Abstract
The nuclear lamina is a filamentous structure subtending the nuclear envelope and required for chromatin organization, transcriptional regulation and maintaining nuclear structure. The trypanosomatid coiled-coil NUP-1 protein is a lamina component functionally analogous to lamins, the major lamina proteins of metazoa. There is little evidence for shared ancestry, suggesting the presence of a distinct lamina system in trypanosomes. To find additional trypanosomatid lamina components we identified NUP-1 interacting proteins by affinity capture and mass-spectrometry. Multiple components of the nuclear pore complex (NPC) and a second coiled-coil protein, which we termed NUP-2, were found. NUP-2 has a punctate distribution at the nuclear periphery throughout the cell cycle and is in close proximity to NUP-1, the NPCs and telomeric chromosomal regions. RNAi-mediated silencing of NUP-2 leads to severe proliferation defects, gross alterations to nuclear structure, chromosomal organization and nuclear envelope architecture. Further, transcription is altered at telomere-proximal variant surface glycoprotein (VSG) expression sites (ESs), suggesting a role in controlling ES expression, although NUP-2 silencing does not increase VSG switching. Transcriptome analysis suggests specific alterations to Pol I-dependent transcription. NUP-1 is mislocalized in NUP-2 knockdown cells and vice versa, implying that NUP-1 and NUP-2 form a co-dependent network and identifying NUP-2 as a second trypanosomatid nuclear lamina component.
Collapse
Affiliation(s)
- Luke Maishman
- School of Life Sciences, University of Dundee, Dundee, Scotland, DD1 5EH, UK
| | - Samson O Obado
- The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA
| | - Sam Alsford
- London School of Hygiene and Tropical Medicine, Keppel Street, London, WC1E 7HT, UK
| | - Jean-Mathieu Bart
- Instituto de Parasitología y Biomedicina López-Neyra, Consejo Superior de Investigaciones Cientificas, 18100 Grenada, Spain
| | - Wei-Ming Chen
- Center for Infectious Disease Research (formerly Seattle Biomedical Research Institute), Seattle, WA 98109, USA
| | - Alexander V Ratushny
- Center for Infectious Disease Research (formerly Seattle Biomedical Research Institute), Seattle, WA 98109, USA.,Institute for Systems Biology, Seattle, WA 98109, USA
| | - Miguel Navarro
- Instituto de Parasitología y Biomedicina López-Neyra, Consejo Superior de Investigaciones Cientificas, 18100 Grenada, Spain
| | - David Horn
- School of Life Sciences, University of Dundee, Dundee, Scotland, DD1 5EH, UK
| | - John D Aitchison
- Center for Infectious Disease Research (formerly Seattle Biomedical Research Institute), Seattle, WA 98109, USA.,Institute for Systems Biology, Seattle, WA 98109, USA
| | - Brian T Chait
- The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA
| | - Michael P Rout
- The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA
| | - Mark C Field
- School of Life Sciences, University of Dundee, Dundee, Scotland, DD1 5EH, UK
| |
Collapse
|
245
|
Kokusho R, Koh Y, Fujimoto M, Shimada T, Katsuma S. Bombyx mori nucleopolyhedrovirus BM5 protein regulates progeny virus production and viral gene expression. Virology 2016; 498:240-249. [PMID: 27614700 DOI: 10.1016/j.virol.2016.08.032] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Revised: 08/27/2016] [Accepted: 08/30/2016] [Indexed: 10/21/2022]
Abstract
Bombyx mori nucleopolyhedrovirus (BmNPV) orf5 (Bm5) is a core gene of lepidopteran baculoviruses and encodes the protein with the conserved amino acid residues (DUF3627) in its C-terminus. Here, we found that Bm5 disruption resulted in lower titers of budded viruses and fewer numbers of occlusion bodies (OBs) in B. mori cultured cells and larvae, although viral genome replication was not affected. Bm5 disruption also caused aberrant expression of various viral genes at the very late stage of infection. Immunocytochemical analysis revealed that BM5 localized to the nuclear membrane. We also found that DUF3627 is important for OB production, transcriptional regulation of viral genes, and subcellular localization of BM5. Compared with wild-type BmNPV infection, larval death was delayed when B. mori larvae were infected with Bm5 mutants. These results suggest that BM5 is involved in progeny virus production and regulation of viral gene expression at the very late stage of infection.
Collapse
Affiliation(s)
- Ryuhei Kokusho
- Department of Agricultural and Environmental Biology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-Ku, Tokyo 113-8657, Japan.
| | - Yoshikazu Koh
- Department of Agricultural and Environmental Biology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-Ku, Tokyo 113-8657, Japan
| | - Masaru Fujimoto
- Department of Agricultural and Environmental Biology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-Ku, Tokyo 113-8657, Japan
| | - Toru Shimada
- Department of Agricultural and Environmental Biology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-Ku, Tokyo 113-8657, Japan
| | - Susumu Katsuma
- Department of Agricultural and Environmental Biology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-Ku, Tokyo 113-8657, Japan.
| |
Collapse
|
246
|
Impaired nuclear functions in micronuclei results in genome instability and chromothripsis. Arch Toxicol 2016; 90:2657-2667. [PMID: 27542123 DOI: 10.1007/s00204-016-1818-4] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Accepted: 08/04/2016] [Indexed: 12/25/2022]
Abstract
Micronuclei (MN) have generally been considered a consequence of DNA damage and, as such, have been used as markers of exposure to genotoxic agents. However, advances in DNA sequencing methods and the development of high-resolution microscopy with which to analyse chromosome dynamics in live cells have been fundamental in building a more refined view of the existing links between DNA damage and micronuclei. Here, we review recent progress indicating that defects of micronuclei affect basic nuclear functions, such as DNA repair and replication, generating massive damage in the chromatin of the MN. In addition, the physical isolation of chromosomes within MN offers an attractive mechanistic explanation for chromothripsis, a massive local DNA fragmentation that produces complex rearrangements restricted to only one or a few chromosomes. When micronuclear chromatin is reincorporated in the daughter cell nuclei, the under-replicated, damaged or rearranged micronuclear chromatin might contribute to genome instability. The traditional conception of micronuclei has been overturned, as they have evolved from passive indicators of DNA damage to active players in the formation of DNA lesions, thus unravelling previously unforeseen roles of micronuclei in the origins of chromosome instability.
Collapse
|
247
|
Machowska M, Piekarowicz K, Rzepecki R. Regulation of lamin properties and functions: does phosphorylation do it all? Open Biol 2016; 5:rsob.150094. [PMID: 26581574 PMCID: PMC4680568 DOI: 10.1098/rsob.150094] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
The main functions of lamins are their mechanical and structural roles as major building blocks of the karyoskeleton. They are also involved in chromatin structure regulation, gene expression, intracellular signalling pathway modulation and development. All essential lamin functions seem to depend on their capacity for assembly or disassembly after the receipt of specific signals, and after specific, selective and precisely regulated interactions through their various domains. Reversible phosphorylation of lamins is crucial for their functions, so it is important to understand how lamin polymerization and interactions are modulated, and which sequences may undergo such modifications. This review combines experimental data with results of our in silico analyses focused on lamin phosphorylation in model organisms to show the presence of evolutionarily conserved sequences and to indicate specific in vivo phosphorylations that affect particular functions.
Collapse
Affiliation(s)
- Magdalena Machowska
- Laboratory of Nuclear Proteins, Faculty of Biotechnology, University of Wrocław, ul. Fryderyka Joliot-Curie 14a, Wrocław 50-383, Poland
| | - Katarzyna Piekarowicz
- Laboratory of Nuclear Proteins, Faculty of Biotechnology, University of Wrocław, ul. Fryderyka Joliot-Curie 14a, Wrocław 50-383, Poland
| | - Ryszard Rzepecki
- Laboratory of Nuclear Proteins, Faculty of Biotechnology, University of Wrocław, ul. Fryderyka Joliot-Curie 14a, Wrocław 50-383, Poland
| |
Collapse
|
248
|
Hu Z, Yang A, Su G, Zhao Y, Wang Y, Chai X, Tu P. Huaier restrains proliferative and invasive potential of human hepatoma SKHEP-1 cells partially through decreased Lamin B1 and elevated NOV. Sci Rep 2016; 6:31298. [PMID: 27503760 PMCID: PMC4977525 DOI: 10.1038/srep31298] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Accepted: 07/15/2016] [Indexed: 02/06/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most common cause of malignancy-related mortality worldwide. It is urgently needed to develop potential drugs with good efficacy and low toxicity for HCC treatment. The anti-tumor effect of Traditional Chinese Medicine (TCM) has received increasing attention worldwide. Trametes robiniophila Murr. (Huaier) has been used in TCM for approximately 1,600 years. Clinically, Huaier has satisfactory therapeutic effects in cancer treatment, especially in HCC. However, the mechanisms underlying the anti-cancer effect of Huaier remain ill defined. Herein we have demonstrated that Huaier dramatically inhibited cell proliferation and induced apoptosis in human hepatoma cell line SKHEP-1. Importantly, Huaier restrained the metastatic capability of SKHEP-1 cells. Mechanistically, down-regulation of Lamin B1 and up-regulation of Nephroblastoma overexpressed (NOV) were at least partially responsible for the inhibitory effect of Huaier on the proliferative and invasive capacity of SKHEP-1 cells. Our finding provided new insights into mechanisms of anti-HCC effect of Huaier and suggested a new scientific basis for clinical medication.
Collapse
Affiliation(s)
- Zhongdong Hu
- Modern Research Center for Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Ailin Yang
- Modern Research Center for Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100102, China
| | - Guozhu Su
- Modern Research Center for Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100102, China
| | - Yunfang Zhao
- Modern Research Center for Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Ying Wang
- Department of Molecular Orthopaedics, Beijing Institute of Traumatology and Orthopaedics, Beijing Jishuitan Hospital, Beijing 100035, China
| | - Xingyun Chai
- Modern Research Center for Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Pengfei Tu
- Modern Research Center for Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| |
Collapse
|
249
|
Meaburn KJ. Spatial Genome Organization and Its Emerging Role as a Potential Diagnosis Tool. Front Genet 2016; 7:134. [PMID: 27507988 PMCID: PMC4961005 DOI: 10.3389/fgene.2016.00134] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Accepted: 07/13/2016] [Indexed: 12/12/2022] Open
Abstract
In eukaryotic cells the genome is highly spatially organized. Functional relevance of higher order genome organization is implied by the fact that specific genes, and even whole chromosomes, alter spatial position in concert with functional changes within the nucleus, for example with modifications to chromatin or transcription. The exact molecular pathways that regulate spatial genome organization and the full implication to the cell of such an organization remain to be determined. However, there is a growing realization that the spatial organization of the genome can be used as a marker of disease. While global genome organization patterns remain largely conserved in disease, some genes and chromosomes occupy distinct nuclear positions in diseased cells compared to their normal counterparts, with the patterns of reorganization differing between diseases. Importantly, mapping the spatial positioning patterns of specific genomic loci can distinguish cancerous tissue from benign with high accuracy. Genome positioning is an attractive novel biomarker since additional quantitative biomarkers are urgently required in many cancer types. Current diagnostic techniques are often subjective and generally lack the ability to identify aggressive cancer from indolent, which can lead to over- or under-treatment of patients. Proof-of-principle for the use of genome positioning as a diagnostic tool has been provided based on small scale retrospective studies. Future large-scale studies are required to assess the feasibility of bringing spatial genome organization-based diagnostics to the clinical setting and to determine if the positioning patterns of specific loci can be useful biomarkers for cancer prognosis. Since spatial reorganization of the genome has been identified in multiple human diseases, it is likely that spatial genome positioning patterns as a diagnostic biomarker may be applied to many diseases.
Collapse
Affiliation(s)
- Karen J. Meaburn
- Cell Biology of Genomes Group, National Cancer Institute, National Institutes of HealthBethesda, MD, USA
| |
Collapse
|
250
|
Nothisen M, Bagilet J, Behr JP, Remy JS, Kotera M. Structure Tuning of Cationic Oligospermine-siRNA Conjugates for Carrier-Free Gene Silencing. Mol Pharm 2016; 13:2718-28. [PMID: 27398779 DOI: 10.1021/acs.molpharmaceut.6b00309] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Oligospermine-siRNA conjugates are able to induce efficient luciferase gene silencing upon carrier-free transfection. These conjugates are readily accessible by a versatile automated chemistry that we developed using a DMT-spermine phosphoramidite reagent. In this article, we used this chemistry to study a wide range of structural modifications of the oligospermine-siRNA conjugates, i.e., variation of conjugate positions and introduction of chemical modifications to increase nuclease resistance. At first we examined gene silencing activity of a series of siRNA-tris(spermine) conjugates with and without chemical modifications in standard carrier assisted conditions. The three spermine units attached at one of the two ends of the sense strand or at the 3'-end of the antisense strand are compatible with gene silencing activity whereas attachment of spermine units at the 5'-end of the antisense strand abolished the activity. 2'-O-Methylated nucleotides introduced in the sense strand are compatible while not in the antisense strand. Thiophosphate links could be used without activity loss at the 3'-end of both strands and at the 5'-end of the sense strand to conjugate oligospermine. Consequently a series of oligospermine-siRNA conjugates containing 15 to 45 spermines units in various configurations were chosen, prepared, and examined in carrier-free conditions. Attachment of 30 spermine units singly at the 5'-end of the sense strand provides the most potent carrier-free siRNA. Longevity of luciferase gene silencing was studied using oligospermine-siRNA conjugates. Five day long efficiency with more than 80% gene expression knockdown was observed upon transfection without vector. Oligospermine-siRNA conjugates targeting cell-constitutive natural lamin A/C gene were prepared. Efficient gene silencing was observed upon carrier-free transfection of siRNA conjugates containing 20 or 30 spermine residues grafted at the 5'-end of the sense strand.
Collapse
Affiliation(s)
- Marc Nothisen
- Laboratoire V-SAT, CAMB, UMR 7199, Université de Strasbourg and CNRS, Faculté de Pharmacie , F-67401 Illkirch, France
| | - Jérémy Bagilet
- Laboratoire V-SAT, CAMB, UMR 7199, Université de Strasbourg and CNRS, Faculté de Pharmacie , F-67401 Illkirch, France
| | - Jean-Paul Behr
- Laboratoire V-SAT, CAMB, UMR 7199, Université de Strasbourg and CNRS, Faculté de Pharmacie , F-67401 Illkirch, France
| | - Jean-Serge Remy
- Laboratoire V-SAT, CAMB, UMR 7199, Université de Strasbourg and CNRS, Faculté de Pharmacie , F-67401 Illkirch, France
| | - Mitsuharu Kotera
- Laboratoire V-SAT, CAMB, UMR 7199, Université de Strasbourg and CNRS, Faculté de Pharmacie , F-67401 Illkirch, France
| |
Collapse
|