51
|
Mattioli E, Andrenacci D, Garofalo C, Prencipe S, Scotlandi K, Remondini D, Gentilini D, Di Blasio AM, Valente S, Scarano E, Cicchilitti L, Piaggio G, Mai A, Lattanzi G. Altered modulation of lamin A/C-HDAC2 interaction and p21 expression during oxidative stress response in HGPS. Aging Cell 2018; 17:e12824. [PMID: 30109767 PMCID: PMC6156291 DOI: 10.1111/acel.12824] [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] [Received: 12/28/2017] [Revised: 05/22/2018] [Accepted: 06/26/2018] [Indexed: 02/01/2023] Open
Abstract
Defects in stress response are main determinants of cellular senescence and organism aging. In fibroblasts from patients affected by Hutchinson-Gilford progeria, a severe LMNA-linked syndrome associated with bone resorption, cardiovascular disorders, and premature aging, we found altered modulation of CDKN1A, encoding p21, upon oxidative stress induction, and accumulation of senescence markers during stress recovery. In this context, we unraveled a dynamic interaction of lamin A/C with HDAC2, an histone deacetylase that regulates CDKN1A expression. In control skin fibroblasts, lamin A/C is part of a protein complex including HDAC2 and its histone substrates; protein interaction is reduced at the onset of DNA damage response and recovered after completion of DNA repair. This interplay parallels modulation of p21 expression and global histone acetylation, and it is disrupted by LMNAmutations leading to progeroid phenotypes. In fact, HGPS cells show impaired lamin A/C-HDAC2 interplay and accumulation of p21 upon stress recovery. Collectively, these results link altered physical interaction between lamin A/C and HDAC2 to cellular and organism aging. The lamin A/C-HDAC2 complex may be a novel therapeutic target to slow down progression of progeria symptoms.
Collapse
Affiliation(s)
- Elisabetta Mattioli
- CNR Institute of Molecular Genetics, Unit of Bologna; Bologna Italy
- Rizzoli Orthopedic Institute; IRCCS; Bologna Italy
| | - Davide Andrenacci
- CNR Institute of Molecular Genetics, Unit of Bologna; Bologna Italy
- Rizzoli Orthopedic Institute; IRCCS; Bologna Italy
| | - Cecilia Garofalo
- Rizzoli Orthopedic Institute; IRCCS; Bologna Italy
- CRS Development of Biomolecular Therapies, Experimental Oncology Lab; Rizzoli Institute; Bologna Italy
| | - Sabino Prencipe
- CNR Institute of Molecular Genetics, Unit of Bologna; Bologna Italy
- Rizzoli Orthopedic Institute; IRCCS; Bologna Italy
| | - Katia Scotlandi
- Rizzoli Orthopedic Institute; IRCCS; Bologna Italy
- CRS Development of Biomolecular Therapies, Experimental Oncology Lab; Rizzoli Institute; Bologna Italy
| | - Daniel Remondini
- Department of Physics and Astronomy; University of Bologna; Bologna Italy
| | - Davide Gentilini
- Centre for Biomedical Research and Technologies; Italian Auxologic Institute, IRCCS; Milan Italy
| | - Anna Maria Di Blasio
- Centre for Biomedical Research and Technologies; Italian Auxologic Institute, IRCCS; Milan Italy
| | - Sergio Valente
- Department of Drug Chemistry and Technologies; Pasteur Institute Italy; Cenci-Bolognetti Foundation; Sapienza University of Rome; Rome Italy
| | - Emanuela Scarano
- Pediatric Endocrinology and Rare Diseases Unit; University of Bologna; Bologna Italy
| | - Lucia Cicchilitti
- UOSD SAFU, Department of Research, Diagnosis and Innovative Technologies; IRCCS - Regina Elena National Cancer Institute; Rome Italy
| | - Giulia Piaggio
- UOSD SAFU, Department of Research, Diagnosis and Innovative Technologies; IRCCS - Regina Elena National Cancer Institute; Rome Italy
| | - Antonello Mai
- Department of Drug Chemistry and Technologies; Pasteur Institute Italy; Cenci-Bolognetti Foundation; Sapienza University of Rome; Rome Italy
| | - Giovanna Lattanzi
- CNR Institute of Molecular Genetics, Unit of Bologna; Bologna Italy
- Rizzoli Orthopedic Institute; IRCCS; Bologna Italy
| |
Collapse
|
52
|
Affiliation(s)
- Saverio Cinti
- Professor of Human Anatomy, Director, Center of Obesity, University of Ancona (Politecnica delle Marche), Ancona, Italy
| |
Collapse
|
53
|
Abstract
Pancreatic cancer is an aggressive and intractable malignancy with high mortality. This is due in part to a high resistance to chemotherapeutics and radiation treatment conferred by diverse regulatory mechanisms. Among these, constituents of the nuclear envelope play a significant role in regulating oncogenesis and pancreatic tumor biology, and this review focuses on three specific components and their roles in cancer. The LINC complex is a nuclear envelope component formed by proteins with SUN and KASH domains that interact in the periplasmic space of the nuclear envelope. These interactions functionally and structurally couple the cytoskeleton to chromatin and facilitates gene regulation informed by cytoplasmic activity. Furthermore, cancer cell invasiveness is impacted by LINC complex biology. The nuclear lamina is adjacent to the inner nuclear membrane of the nuclear envelope and can actively regulate chromatin in addition to providing structural integrity to the nucleus. A disrupted lamina can impart biophysical compromise to nuclear structure and function, as well as form dysfunctional micronuclei that may lead to genomic instability and chromothripsis. In close relationship to the nuclear lamina is the nuclear pore complex, a large megadalton structure that spans both outer and inner membranes of the nuclear envelope. The nuclear pore complex mediates bidirectional nucleocytoplasmic transport and is comprised of specialized proteins called nucleoporins that are overexpressed in many cancers and are diagnostic markers for oncogenesis. Furthermore, recent demonstration of gene regulatory functions for discrete nucleoporins independent of their nuclear trafficking function suggests that these proteins may contribute more to malignant phenotypes beyond serving as biomarkers. The nuclear envelope is thus a complex, intricate regulator of cell signaling, with roles in pancreatic tumorigenesis and general oncogenic transformation.
Collapse
Affiliation(s)
| | - Randolph S. Faustino
- Genetics and Genomics, Sanford Research, Sioux Falls, SD 57104, USA
- Department of Pediatrics, Sanford School of Medicine of the University of South Dakota, Sioux Falls, SD 57105, USA
| |
Collapse
|
54
|
Martino F, Perestrelo AR, Vinarský V, Pagliari S, Forte G. Cellular Mechanotransduction: From Tension to Function. Front Physiol 2018; 9:824. [PMID: 30026699 PMCID: PMC6041413 DOI: 10.3389/fphys.2018.00824] [Citation(s) in RCA: 540] [Impact Index Per Article: 90.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2018] [Accepted: 06/12/2018] [Indexed: 12/15/2022] Open
Abstract
Living cells are constantly exposed to mechanical stimuli arising from the surrounding extracellular matrix (ECM) or from neighboring cells. The intracellular molecular processes through which such physical cues are transformed into a biological response are collectively dubbed as mechanotransduction and are of fundamental importance to help the cell timely adapt to the continuous dynamic modifications of the microenvironment. Local changes in ECM composition and mechanics are driven by a feed forward interplay between the cell and the matrix itself, with the first depositing ECM proteins that in turn will impact on the surrounding cells. As such, these changes occur regularly during tissue development and are a hallmark of the pathologies of aging. Only lately, though, the importance of mechanical cues in controlling cell function (e.g., proliferation, differentiation, migration) has been acknowledged. Here we provide a critical review of the recent insights into the molecular basis of cellular mechanotransduction, by analyzing how mechanical stimuli get transformed into a given biological response through the activation of a peculiar genetic program. Specifically, by recapitulating the processes involved in the interpretation of ECM remodeling by Focal Adhesions at cell-matrix interphase, we revise the role of cytoskeleton tension as the second messenger of the mechanotransduction process and the action of mechano-responsive shuttling proteins converging on stage and cell-specific transcription factors. Finally, we give few paradigmatic examples highlighting the emerging role of malfunctions in cell mechanosensing apparatus in the onset and progression of pathologies.
Collapse
Affiliation(s)
- Fabiana Martino
- Center for Translational Medicine, International Clinical Research Center, St. Anne’s University Hospital, Brno, Czechia
- Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czechia
- Competence Center for Mechanobiology in Regenerative Medicine, INTERREG ATCZ133, Brno, Czechia
| | - Ana R. Perestrelo
- Center for Translational Medicine, International Clinical Research Center, St. Anne’s University Hospital, Brno, Czechia
| | - Vladimír Vinarský
- Center for Translational Medicine, International Clinical Research Center, St. Anne’s University Hospital, Brno, Czechia
- Competence Center for Mechanobiology in Regenerative Medicine, INTERREG ATCZ133, Brno, Czechia
| | - Stefania Pagliari
- Center for Translational Medicine, International Clinical Research Center, St. Anne’s University Hospital, Brno, Czechia
| | - Giancarlo Forte
- Center for Translational Medicine, International Clinical Research Center, St. Anne’s University Hospital, Brno, Czechia
- Competence Center for Mechanobiology in Regenerative Medicine, INTERREG ATCZ133, Brno, Czechia
- Department of Biomaterials Science, Institute of Dentistry, University of Turku, Turku, Finland
| |
Collapse
|
55
|
SUN2 Modulates HIV-1 Infection and Latency through Association with Lamin A/C To Maintain the Repressive Chromatin. mBio 2018; 9:mBio.02408-17. [PMID: 29717016 PMCID: PMC5930302 DOI: 10.1128/mbio.02408-17] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
The postintegrational latency of HIV-1 is characterized by reversible silencing of long terminal repeat (LTR)-driven transcription of the HIV genome. It is known that the formation of repressive chromatin at the 5′-LTR of HIV-1 proviral DNA impedes viral transcription by blocking the recruitment of positive transcription factors. How the repressive chromatin is formed and modulated during HIV-1 infection remains elusive. Elucidation of which chromatin reassembly factor mediates the reorganization of chromatin is likely to facilitate the understanding of the host’s modulation of HIV-1 transcription and latency. Here we revealed that “Sad1 and UNC84 domain containing 2” (SUN2), an inner nuclear membrane protein, maintained the repressive chromatin and inhibited HIV LTR-driven transcription of proviral DNA through an association with lamin A/C. Specifically, lamin A/C tethered SUN2 to the nucleosomes 1 and 2 of the HIV-1 5′-LTR to block the initiation and elongation of HIV-1 transcription. SUN2 knockdown converted chromatin to an active form and thus enhanced the phosphorylation of RNA polymerase II and its recruitment to the 5′-LTR HIV-1 proviral DNA, leading to reactivation of HIV-1 from latency. Conversely, the exogenous factors such as tumor necrosis factor alpha (TNF-α) induced reactivation, and the replication of HIV-1 led to the disassociation between SUN2 and lamin A/C, suggesting that disruption of the association between SUN2 and lamin A/C to convert the repressive chromatin to the active form might be a prerequisite for the initiation of HIV-1 transcription and replication. Together, our findings indicate that SUN2 is a novel chromatin reassembly factor that helps to maintain chromatin in a repressive state and consequently inhibits HIV-1 transcription. Despite the successful use of scores of antiretroviral drugs, HIV latency poses a major impediment to virus eradication. Elucidation of the mechanism of latency facilitates the discovery of new therapeutic strategies. It has been known that the formation of repressive chromatin at the 5′-LTR of HIV-1 proviral DNA impedes viral transcription and maintains viral latency, but how the repressive chromatin is formed and modulated during HIV-1 infection remains elusive. In this study, we performed in-depth virological and cell biological studies and discovered that an inner nuclear membrane protein, SUN2, is a novel chromatin reassembly factor that maintains repressive chromatin and thus modulates HIV-1 transcription and latency: therefore, targeting SUN2 may lead to new strategies for HIV cure.
Collapse
|
56
|
Gargiuli C, Schena E, Mattioli E, Columbaro M, D'Apice MR, Novelli G, Greggi T, Lattanzi G. Lamins and bone disorders: current understanding and perspectives. Oncotarget 2018; 9:22817-22831. [PMID: 29854317 PMCID: PMC5978267 DOI: 10.18632/oncotarget.25071] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Accepted: 03/22/2018] [Indexed: 12/31/2022] Open
Abstract
Lamin A/C is a major constituent of the nuclear lamina implicated in a number of genetic diseases, collectively known as laminopathies. The most severe forms of laminopathies feature, among other symptoms, congenital scoliosis, osteoporosis, osteolysis or delayed cranial ossification. Importantly, specific bone districts are typically affected in laminopathies. Spine is severely affected in LMNA-linked congenital muscular dystrophy. Mandible, terminal phalanges and clavicles undergo osteolytic processes in progeroid laminopathies and Restrictive Dermopathy, a lethal developmental laminopathy. This specificity suggests that lamin A/C regulates fine mechanisms of bone turnover, as supported by data showing that lamin A/C mutations activate non-canonical pathways of osteoclastogenesis, as the one dependent on TGF beta 2. Here, we review current knowledge on laminopathies affecting bone and LMNA involvement in bone turnover and highlight lamin-dependent mechanisms causing bone disorders. This knowledge can be exploited to identify new therapeutic approaches not only for laminopathies, but also for other rare diseases featuring bone abnormalities.
Collapse
Affiliation(s)
- Chiara Gargiuli
- CNR Institute of Molecular Genetics, Unit of Bologna, Bologna, Italy
| | - Elisa Schena
- CNR Institute of Molecular Genetics, Unit of Bologna, Bologna, Italy.,Rizzoli Orthopaedic Institute, Laboratory of Cell Biology, Bologna, Italy
| | - Elisabetta Mattioli
- CNR Institute of Molecular Genetics, Unit of Bologna, Bologna, Italy.,Rizzoli Orthopaedic Institute, Laboratory of Cell Biology, Bologna, Italy
| | - Marta Columbaro
- Rizzoli Orthopaedic Institute, Laboratory of Cell Biology, Bologna, Italy
| | | | - Giuseppe Novelli
- Medical Genetics Unit, Policlinico Tor Vergata University Hospital, Rome, Italy
| | - Tiziana Greggi
- Rizzoli Orthopaedic Institute, Spine Deformity Department, Bologna, Italy
| | - Giovanna Lattanzi
- CNR Institute of Molecular Genetics, Unit of Bologna, Bologna, Italy.,Rizzoli Orthopaedic Institute, Laboratory of Cell Biology, Bologna, Italy
| |
Collapse
|
57
|
Cicchillitti L, Manni I, Mancone C, Regazzo G, Spagnuolo M, Alonzi T, Carlomosti F, Dell'Anna ML, Dell'Omo G, Picardo M, Ciana P, Capogrossi MC, Tripodi M, Magenta A, Rizzo MG, Gurtner A, Piaggio G. The laminA/NF-Y protein complex reveals an unknown transcriptional mechanism on cell proliferation. Oncotarget 2018; 8:2628-2646. [PMID: 27793050 PMCID: PMC5356829 DOI: 10.18632/oncotarget.12914] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Accepted: 10/10/2016] [Indexed: 12/02/2022] Open
Abstract
Lamin A is a component of the nuclear matrix that also controls proliferation by largely unknown mechanisms. NF-Y is a ubiquitous protein involved in cell proliferation composed of three subunits (-YA -YB -YC) all required for the DNA binding and transactivation activity. To get clues on new NF-Y partner(s) we performed a mass spectrometry screening of proteins that co-precipitate with the regulatory subunit of the complex, NF-YA. By this screening we identified lamin A as a novel putative NF-Y interactor. Co-immunoprecipitation experiments and confocal analysis confirmed the interaction between the two endogenous proteins. Interestingly, this association occurs on euchromatin regions, too. ChIP experiments demonstrate lamin A enrichment in several promoter regions of cell cycle related genes in a NF-Y dependent manner. Gain and loss of function experiments reveal that lamin A counteracts NF-Y transcriptional activity. Taking advantage of a recently generated transgenic reporter mouse, called MITO-Luc, in which an NF-Y–dependent promoter controls luciferase expression, we demonstrate that lamin A counteracts NF-Y transcriptional activity not only in culture cells but also in living animals. Altogether, our data demonstrate the occurrence of lamin A/NF-Y interaction and suggest a possible role of this protein complex in regulation of NF-Y function in cell proliferation.
Collapse
Affiliation(s)
- Lucia Cicchillitti
- Department of Research, Advanced Diagnostics and Technological Innovation, SAFU Unit, Translational Research Area, Regina Elena National Cancer Institute, 00144 Rome, Italy
| | - Isabella Manni
- Department of Research, Advanced Diagnostics and Technological Innovation, SAFU Unit, Translational Research Area, Regina Elena National Cancer Institute, 00144 Rome, Italy
| | - Carmine Mancone
- National Institute for Infectious Diseases L. Spallanzani, IRCCS, Department of Epidemiology and Preclinical Research, 00149 Rome, Italy.,Department of Cellular Biotechnologies and Haematology, Istituto Pasteur Italia, Fondazione Cenci Bolognetti, Sapienza University of Rome, 00161 Rome, Italy
| | - Giulia Regazzo
- Department of Research, Advanced Diagnostics and Technological Innovation, Genomic and Epigenetic Unit, Translational Research Area, Regina Elena National Cancer Institute, Rome, Italy
| | - Manuela Spagnuolo
- Department of Research, Advanced Diagnostics and Technological Innovation, Genomic and Epigenetic Unit, Translational Research Area, Regina Elena National Cancer Institute, Rome, Italy
| | - Tonino Alonzi
- National Institute for Infectious Diseases L. Spallanzani, IRCCS, Department of Epidemiology and Preclinical Research, 00149 Rome, Italy
| | - Fabrizio Carlomosti
- Fondazione Luigi Maria Monti, Istituto Dermopatico dell'Immacolata-IRCCS, Laboratorio di Patologia Vascolare, 00167 Rome, Italy
| | - Maria Lucia Dell'Anna
- Cutaneous Physiopathology and Integrated Center of Metabolomics Research, San Gallicano Dermatologic Institute, IRCCS, 00144 Rome, Italy
| | - Giulia Dell'Omo
- Department of Oncology and Hemato-Oncology and Department of Pharmacological and Biomolecular Sciences, University of Milan, 20133 Milan, Italy
| | - Mauro Picardo
- Cutaneous Physiopathology and Integrated Center of Metabolomics Research, San Gallicano Dermatologic Institute, IRCCS, Via Elio Chianesi 53, 00144, Rome, Italy
| | - Paolo Ciana
- Center of Excellence on Neurodegenerative Diseases, Department of Oncology and Hemato-Oncology, University of Milan, 20133 Milan, Italy
| | - Maurizio C Capogrossi
- Fondazione Luigi Maria Monti, Istituto Dermopatico dell'Immacolata-IRCCS, Laboratorio di Patologia Vascolare, Via dei Monti di Creta 104, Rome 00167, Italy Rome, Italy
| | - Marco Tripodi
- National Institute for Infectious Diseases L. Spallanzani, IRCCS, Department of Epidemiology and Preclinical Research, 00149 Rome, Italy.,Department of Cellular Biotechnologies and Haematology, Istituto Pasteur Italia, Fondazione Cenci Bolognetti, Sapienza University of Rome, 00161 Rome, Italy
| | - Alessandra Magenta
- Fondazione Luigi Maria Monti, Istituto Dermopatico dell'Immacolata-IRCCS, Laboratorio di Patologia Vascolare, Via dei Monti di Creta 104, Rome 00167, Italy Rome, Italy
| | - Maria Giulia Rizzo
- Department of Research, Advanced Diagnostics and Technological Innovation, Genomic and Epigenetic Unit, Translational Research Area, Regina Elena National Cancer Institute, Rome, Italy
| | - Aymone Gurtner
- Department of Research, Advanced Diagnostics and Technological Innovation, SAFU Unit, Translational Research Area, Regina Elena National Cancer Institute, 00144 Rome, Italy
| | - Giulia Piaggio
- Department of Research, Advanced Diagnostics and Technological Innovation, SAFU Unit, Translational Research Area, Regina Elena National Cancer Institute, 00144 Rome, Italy
| |
Collapse
|
58
|
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]
|
59
|
Stephens AD, Liu PZ, Banigan EJ, Almassalha LM, Backman V, Adam SA, Goldman RD, Marko JF. Chromatin histone modifications and rigidity affect nuclear morphology independent of lamins. Mol Biol Cell 2018; 29:220-233. [PMID: 29142071 PMCID: PMC5909933 DOI: 10.1091/mbc.e17-06-0410] [Citation(s) in RCA: 211] [Impact Index Per Article: 35.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Revised: 10/17/2017] [Accepted: 11/08/2017] [Indexed: 01/29/2023] Open
Abstract
Nuclear shape and architecture influence gene localization, mechanotransduction, transcription, and cell function. Abnormal nuclear morphology and protrusions termed "blebs" are diagnostic markers for many human afflictions including heart disease, aging, progeria, and cancer. Nuclear blebs are associated with both lamin and chromatin alterations. A number of prior studies suggest that lamins dictate nuclear morphology, but the contributions of altered chromatin compaction remain unclear. We show that chromatin histone modification state dictates nuclear rigidity, and modulating it is sufficient to both induce and suppress nuclear blebs. Treatment of mammalian cells with histone deacetylase inhibitors to increase euchromatin or histone methyltransferase inhibitors to decrease heterochromatin results in a softer nucleus and nuclear blebbing, without perturbing lamins. Conversely, treatment with histone demethylase inhibitors increases heterochromatin and chromatin nuclear rigidity, which results in reduced nuclear blebbing in lamin B1 null nuclei. Notably, increased heterochromatin also rescues nuclear morphology in a model cell line for the accelerated aging disease Hutchinson-Gilford progeria syndrome caused by mutant lamin A, as well as cells from patients with the disease. Thus, chromatin histone modification state is a major determinant of nuclear blebbing and morphology via its contribution to nuclear rigidity.
Collapse
Affiliation(s)
- Andrew D Stephens
- Department of Molecular Biosciences, Northwestern University, Evanston, IL 60208
| | - Patrick Z Liu
- Department of Molecular Biosciences, Northwestern University, Evanston, IL 60208
| | - Edward J Banigan
- Department of Physics and Astronomy, Northwestern University, Evanston, IL 60208.,Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Luay M Almassalha
- Department of Biomedical Engineering, Northwestern University, Evanston, IL 60208
| | - Vadim Backman
- Department of Biomedical Engineering, Northwestern University, Evanston, IL 60208
| | - Stephen A Adam
- Department of Cell and Molecular Biology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| | - Robert D Goldman
- Department of Cell and Molecular Biology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| | - John F Marko
- Department of Molecular Biosciences, Northwestern University, Evanston, IL 60208.,Department of Physics and Astronomy, Northwestern University, Evanston, IL 60208
| |
Collapse
|
60
|
Tschumperlin DJ, Ligresti G, Hilscher MB, Shah VH. Mechanosensing and fibrosis. J Clin Invest 2018; 128:74-84. [PMID: 29293092 DOI: 10.1172/jci93561] [Citation(s) in RCA: 179] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Tissue injury disrupts the mechanical homeostasis that underlies normal tissue architecture and function. The failure to resolve injury and restore homeostasis gives rise to progressive fibrosis that is accompanied by persistent alterations in the mechanical environment as a consequence of pathological matrix deposition and stiffening. This Review focuses on our rapidly growing understanding of the molecular mechanisms linking the altered mechanical environment in injury, repair, and fibrosis to cellular activation. In particular, our focus is on the mechanisms by which cells transduce mechanical signals, leading to transcriptional and epigenetic responses that underlie both transient and persistent alterations in cell state that contribute to fibrosis. Translation of these mechanobiological insights may enable new approaches to promote tissue repair and arrest or reverse fibrotic tissue remodeling.
Collapse
Affiliation(s)
| | | | - Moira B Hilscher
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, USA
| | - Vijay H Shah
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
61
|
Barrier-to-autointegration factor (BAF) involvement in prelamin A-related chromatin organization changes. Oncotarget 2017; 7:15662-77. [PMID: 26701887 PMCID: PMC4941268 DOI: 10.18632/oncotarget.6697] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Accepted: 11/21/2015] [Indexed: 11/25/2022] Open
Abstract
Chromatin disorganization is one of the major alterations linked to prelamin A processing impairment. In this study we demonstrate that BAF is necessary to modulate prelamin A effects on chromatin structure. We show that when prelamin A and BAF cannot properly interact no prelamin A-dependent effects on chromatin occur; similar to what is observed in human Nestor Guillermo Progeria Syndrome cells harboring a BAF mutation, in HEK293 cells expressing a BAF mutant unable to bind prelamin A, or in siRNA mediated BAF-depleted HEK293 cells expressing prelamin A. BAF is necessary to induce histone trimethyl-H3K9 as well as HP1-alpha and LAP2-alpha nuclear relocalization in response to prelamin A accumulation. These findings are enforced by electron microscopy evaluations showing how the prelamin A-BAF interaction governs overall chromatin organization. Finally, we demonstrate that the LAP2-alpha nuclear localization defect observed in HGPS cells involves the progerin-BAF interaction, thus establishing a functional link between BAF and prelamin A pathological forms.
Collapse
|
62
|
Perovanovic J, Dell'Orso S, Gnochi VF, Jaiswal JK, Sartorelli V, Vigouroux C, Mamchaoui K, Mouly V, Bonne G, Hoffman EP. Laminopathies disrupt epigenomic developmental programs and cell fate. Sci Transl Med 2017; 8:335ra58. [PMID: 27099177 DOI: 10.1126/scitranslmed.aad4991] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Accepted: 02/28/2016] [Indexed: 12/12/2022]
Abstract
The nuclear envelope protein lamin A is encoded by thelamin A/C(LMNA) gene, which can contain missense mutations that cause Emery-Dreifuss muscular dystrophy (EDMD) (p.R453W). We fused mutated forms of the lamin A protein to bacterial DNA adenine methyltransferase (Dam) to define euchromatic-heterochromatin (epigenomic) transitions at the nuclear envelope during myogenesis (using DamID-seq). Lamin A missense mutations disrupted appropriate formation of lamin A-associated heterochromatin domains in an allele-specific manner-findings that were confirmed by chromatin immunoprecipitation-DNA sequencing (ChIP-seq) in murine H2K cells and DNA methylation studies in fibroblasts from muscular dystrophy patient who carried a distinctLMNAmutation (p.H222P). Observed perturbations of the epigenomic transitions included exit from pluripotency and cell cycle programs [euchromatin (open, transcribed) to heterochromatin (closed, silent)], as well as induction of myogenic loci (heterochromatin to euchromatin). In muscle biopsies from patients with either a gain- or change-of-functionLMNAgene mutation or a loss-of-function mutation in theemeringene, both of which cause EDMD, we observed inappropriate loss of heterochromatin formation at theSox2pluripotency locus, which was associated with persistent mRNA expression ofSox2 Overexpression ofSox2inhibited myogenic differentiation in human immortalized myoblasts. Our findings suggest that nuclear envelopathies are disorders of developmental epigenetic programming that result from altered formation of lamina-associated domains.
Collapse
Affiliation(s)
- Jelena Perovanovic
- Center for Genetic Medicine Research, Children's National Medical Center, Washington, DC 20010, USA. Department of Integrative Systems Biology, The George Washington University School of Medicine and Health Sciences, Washington, DC 20010, USA
| | - Stefania Dell'Orso
- Laboratory of Muscle Stem Cells and Gene Regulation, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD 20852, USA
| | - Viola F Gnochi
- Center for Genetic Medicine Research, Children's National Medical Center, Washington, DC 20010, USA
| | - Jyoti K Jaiswal
- Center for Genetic Medicine Research, Children's National Medical Center, Washington, DC 20010, USA. Department of Integrative Systems Biology, The George Washington University School of Medicine and Health Sciences, Washington, DC 20010, USA
| | - Vittorio Sartorelli
- Laboratory of Muscle Stem Cells and Gene Regulation, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD 20852, USA
| | - Corinne Vigouroux
- Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Saint-Antoine, Laboratoire Commun de Biologie et Génétique Moléculaires, F-75012 Paris, France. INSERM UMR_S938, Centre de Recherche Saint-Antoine, F-75012 Paris, France. Sorbonne Universités, UPMC (Université Pierre et Marie Curie) Univ Paris 06, UMR_S938, F-75005 Paris, France. ICAN (Institute of Cardiometabolism and Nutrition), F-75013 Paris, France
| | - Kamel Mamchaoui
- Sorbonne Universités, UPMC Univ Paris 06, INSERM UMRS974, CNRS FRE3617, Center for Research in Myology, F-75013 Paris, France
| | - Vincent Mouly
- Sorbonne Universités, UPMC Univ Paris 06, INSERM UMRS974, CNRS FRE3617, Center for Research in Myology, F-75013 Paris, France
| | - Gisèle Bonne
- Sorbonne Universités, UPMC Univ Paris 06, INSERM UMRS974, CNRS FRE3617, Center for Research in Myology, F-75013 Paris, France
| | - Eric P Hoffman
- Center for Genetic Medicine Research, Children's National Medical Center, Washington, DC 20010, USA. Department of Integrative Systems Biology, The George Washington University School of Medicine and Health Sciences, Washington, DC 20010, USA.
| |
Collapse
|
63
|
Carrero D, Soria-Valles C, López-Otín C. Hallmarks of progeroid syndromes: lessons from mice and reprogrammed cells. Dis Model Mech 2017; 9:719-35. [PMID: 27482812 PMCID: PMC4958309 DOI: 10.1242/dmm.024711] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Ageing is a process that inevitably affects most living organisms and involves the accumulation of macromolecular damage, genomic instability and loss of heterochromatin. Together, these alterations lead to a decline in stem cell function and to a reduced capability to regenerate tissue. In recent years, several genetic pathways and biochemical mechanisms that contribute to physiological ageing have been described, but further research is needed to better characterize this complex biological process. Because premature ageing (progeroid) syndromes, including progeria, mimic many of the characteristics of human ageing, research into these conditions has proven to be very useful not only to identify the underlying causal mechanisms and identify treatments for these pathologies, but also for the study of physiological ageing. In this Review, we summarize the main cellular and animal models used in progeria research, with an emphasis on patient-derived induced pluripotent stem cell models, and define a series of molecular and cellular hallmarks that characterize progeroid syndromes and parallel physiological ageing. Finally, we describe the therapeutic strategies being investigated for the treatment of progeroid syndromes, and their main limitations. Summary: This Review defines the molecular and cellular hallmarks of progeroid syndromes according to the main cellular and animal models, and discusses the therapeutic strategies developed to date.
Collapse
Affiliation(s)
- Dido Carrero
- Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Instituto Universitario de Oncología (IUOPA), Universidad de Oviedo, Oviedo 33006, Spain
| | - Clara Soria-Valles
- Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Instituto Universitario de Oncología (IUOPA), Universidad de Oviedo, Oviedo 33006, Spain
| | - Carlos López-Otín
- Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Instituto Universitario de Oncología (IUOPA), Universidad de Oviedo, Oviedo 33006, Spain
| |
Collapse
|
64
|
Jafferali MH, Figueroa RA, Hasan M, Hallberg E. Spindle associated membrane protein 1 (Samp1) is required for the differentiation of muscle cells. Sci Rep 2017; 7:16655. [PMID: 29192166 PMCID: PMC5709512 DOI: 10.1038/s41598-017-16746-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Accepted: 10/20/2017] [Indexed: 01/15/2023] Open
Abstract
Muscles are developed and regenerated in a differentiation process called myogenesis, which involves components of the nuclear envelope. We have investigated Samp1 (Spindle Associated Membrane Protein 1), a transmembrane nuclear envelope protein, which interacts with emerin and lamin A, both of which are linked to Emery-Dreifuss muscular dystrophy (EDMD). We found that the levels of Samp1 increased seven-fold during differentiation of mouse C2C12 muscle progenitor cells. To test if Samp1 could have a role in myogenesis we developed stable C2C12 knockdown cell lines expressing short hairpin RNA targeting Samp1 expression. The Samp1 depleted C2C12 cells displayed normal mobility and normal distribution of emerin and lamin A. However, Samp1 depletion increased ERK signaling and completely blocked differentiation of C2C12 cells, which failed to express myogenic marker proteins and failed to form myotubes. The block in myogenesis in Samp1 depleted cells was completely rescued by ectopic expression of RNAi resistant human Samp1, showing that Samp1 is required for muscle differentiation.
Collapse
Affiliation(s)
- Mohammed Hakim Jafferali
- Department of Neurochemistry, Stockholm University, Svante Arrhenius väg 16B, SE-106 91, Stockholm, Sweden
| | - Ricardo A Figueroa
- Department of Neurochemistry, Stockholm University, Svante Arrhenius väg 16B, SE-106 91, Stockholm, Sweden
| | - Mehedi Hasan
- Department of Neurochemistry, Stockholm University, Svante Arrhenius väg 16B, SE-106 91, Stockholm, Sweden
| | - Einar Hallberg
- Department of Neurochemistry, Stockholm University, Svante Arrhenius väg 16B, SE-106 91, Stockholm, Sweden.
| |
Collapse
|
65
|
Davies SJ, Ryan J, O'Connor PBF, Kenny E, Morris D, Baranov PV, O'Connor R, McCarthy TV. Itm2a silencing rescues lamin A mediated inhibition of 3T3-L1 adipocyte differentiation. Adipocyte 2017; 6:259-276. [PMID: 28872940 DOI: 10.1080/21623945.2017.1362510] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Dysregulation of adipose tissue metabolism is associated with multiple metabolic disorders. One such disease, known as Dunnigan-type familial partial lipodystrophy (FPLD2) is characterized by defective fat metabolism and storage. FPLD2 is caused by a specific subset of mutations in the LMNA gene. The mechanisms by which LMNA mutations lead to the adipose specific FPLD2 phenotype have yet to be determined in detail. We used RNA-Seq analysis to assess the effects of wild-type (WT) and mutant (R482W) lamin A on the expression profile of differentiating 3T3-L1 mouse preadipocytes and identified Itm2a as a gene that was upregulated at 36 h post differentiation induction in these cells. In this study we identify Itm2a as a novel modulator of adipogenesis and show that endogenous Itm2a expression is transiently downregulated during induction of 3T3-L1 differentiation. Itm2a overexpression was seen to moderately inhibit differentiation of 3T3-L1 preadipocytes while shRNA mediated knockdown of Itm2a significantly enhanced 3T3-L1 differentiation. Investigation of PPARγ levels indicate that this enhanced adipogenesis is mediated through the stabilization of the PPARγ protein at specific time points during differentiation. Finally, we demonstrate that Itm2a knockdown is sufficient to rescue the inhibitory effects of lamin A WT and R482W mutant overexpression on 3T3-L1 differentiation. This suggests that targeting of Itm2a or its related pathways, including autophagy, may have potential as a therapy for FPLD2.
Collapse
Affiliation(s)
- Stephanie J. Davies
- School of Biochemistry and Cell Biology, University College Cork, Co. Cork, Ireland
| | - James Ryan
- Mater Private Hospital, Citygate, Mahon, Cork, Ireland
| | | | - Elaine Kenny
- Neuropsychiatric Genetics Research Group, Department of Psychiatry and Institute of Molecular Medicine, Trinity College Dublin, Dublin, Ireland
| | - Derek Morris
- Department of Biochemistry, National University of Ireland Galway, Galway, Ireland
| | - Pavel V. Baranov
- School of Biochemistry and Cell Biology, University College Cork, Co. Cork, Ireland
| | - Rosemary O'Connor
- School of Biochemistry and Cell Biology, University College Cork, Co. Cork, Ireland
| | - Tommie V. McCarthy
- School of Biochemistry and Cell Biology, University College Cork, Co. Cork, Ireland
| |
Collapse
|
66
|
Bou Saada Y, Zakharova V, Chernyak B, Dib C, Carnac G, Dokudovskaya S, Vassetzky YS. Control of DNA integrity in skeletal muscle under physiological and pathological conditions. Cell Mol Life Sci 2017; 74:3439-3449. [PMID: 28444416 PMCID: PMC11107590 DOI: 10.1007/s00018-017-2530-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Revised: 04/03/2017] [Accepted: 04/19/2017] [Indexed: 02/07/2023]
Abstract
Skeletal muscle is a highly oxygen-consuming tissue that ensures body support and movement, as well as nutrient and temperature regulation. DNA damage induced by reactive oxygen species is present in muscles and tends to accumulate with age. Here, we present a summary of data obtained on DNA damage and its implication in muscle homeostasis, myogenic differentiation and neuromuscular disorders. Controlled and transient DNA damage appears to be essential for muscular homeostasis and differentiation while uncontrolled and chronic DNA damage negatively affects muscle health.
Collapse
Affiliation(s)
- Yara Bou Saada
- UMR 8126, CNRS, Univ. Paris-Sud, Université Paris Saclay, Institut de Cancérologie Gustave-Roussy, 94805, Villejuif, France
| | - Vlada Zakharova
- UMR 8126, CNRS, Univ. Paris-Sud, Université Paris Saclay, Institut de Cancérologie Gustave-Roussy, 94805, Villejuif, France
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, 117334, Russia
| | - Boris Chernyak
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, 117334, Russia
| | - Carla Dib
- UMR 8126, CNRS, Univ. Paris-Sud, Université Paris Saclay, Institut de Cancérologie Gustave-Roussy, 94805, Villejuif, France
| | - Gilles Carnac
- PhyMedExp, INSERM U1046, CNRS UMR 9214, University of Montpellier, 34295, Montpellier Cedex 5, France
| | - Svetlana Dokudovskaya
- UMR 8126, CNRS, Univ. Paris-Sud, Université Paris Saclay, Institut de Cancérologie Gustave-Roussy, 94805, Villejuif, France
| | - Yegor S Vassetzky
- UMR 8126, CNRS, Univ. Paris-Sud, Université Paris Saclay, Institut de Cancérologie Gustave-Roussy, 94805, Villejuif, France.
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, 117334, Russia.
- Koltzov Institute of Developmental Biology, Moscow, 117334, Russia.
| |
Collapse
|
67
|
Chen Z, Chang WY, Etheridge A, Strickfaden H, Jin Z, Palidwor G, Cho JH, Wang K, Kwon SY, Doré C, Raymond A, Hotta A, Ellis J, Kandel RA, Dilworth FJ, Perkins TJ, Hendzel MJ, Galas DJ, Stanford WL. Reprogramming progeria fibroblasts re-establishes a normal epigenetic landscape. Aging Cell 2017; 16:870-887. [PMID: 28597562 PMCID: PMC5506428 DOI: 10.1111/acel.12621] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/21/2017] [Indexed: 12/14/2022] Open
Abstract
Ideally, disease modeling using patient‐derived induced pluripotent stem cells (iPSCs) enables analysis of disease initiation and progression. This requires any pathological features of the patient cells used for reprogramming to be eliminated during iPSC generation. Hutchinson–Gilford progeria syndrome (HGPS) is a segmental premature aging disorder caused by the accumulation of the truncated form of Lamin A known as Progerin within the nuclear lamina. Cellular hallmarks of HGPS include nuclear blebbing, loss of peripheral heterochromatin, defective epigenetic inheritance, altered gene expression, and senescence. To model HGPS using iPSCs, detailed genome‐wide and structural analysis of the epigenetic landscape is required to assess the initiation and progression of the disease. We generated a library of iPSC lines from fibroblasts of patients with HGPS and controls, including one family trio. HGPS patient‐derived iPSCs are nearly indistinguishable from controls in terms of pluripotency, nuclear membrane integrity, as well as transcriptional and epigenetic profiles, and can differentiate into affected cell lineages recapitulating disease progression, despite the nuclear aberrations, altered gene expression, and epigenetic landscape inherent to the donor fibroblasts. These analyses demonstrate the power of iPSC reprogramming to reset the epigenetic landscape to a revitalized pluripotent state in the face of widespread epigenetic defects, validating their use to model the initiation and progression of disease in affected cell lineages.
Collapse
Affiliation(s)
- Zhaoyi Chen
- The Sprott Centre for Stem Cell Research; Regenerative Medicine Program; Ottawa Hospital Research Institute; Ottawa Ontario Canada K1H 8L6
- Department of Cellular and Molecular Medicine; University of Ottawa; Ottawa Ontario Canada
| | - Wing Y. Chang
- The Sprott Centre for Stem Cell Research; Regenerative Medicine Program; Ottawa Hospital Research Institute; Ottawa Ontario Canada K1H 8L6
| | - Alton Etheridge
- Pacific Northwest Diabetes Research Institute; 720 Broadway Seattle WA 98103 USA
| | - Hilmar Strickfaden
- Cross Cancer Institute and the Department of Experimental Oncology; Faculty of Medicine and Dentistry; University of Alberta; Edmonton Alberta Canada T6G 1Z2
| | - Zhigang Jin
- Cross Cancer Institute and the Department of Experimental Oncology; Faculty of Medicine and Dentistry; University of Alberta; Edmonton Alberta Canada T6G 1Z2
| | - Gareth Palidwor
- The Sprott Centre for Stem Cell Research; Regenerative Medicine Program; Ottawa Hospital Research Institute; Ottawa Ontario Canada K1H 8L6
- Ottawa Bioinformatics Core Facility; The Sprott Centre for Stem Cell Research; Ottawa Hospital Research Institute; Ottawa Ontario Canada K1H 8L6
| | - Ji-Hoon Cho
- Pacific Northwest Diabetes Research Institute; 720 Broadway Seattle WA 98103 USA
| | - Kai Wang
- Pacific Northwest Diabetes Research Institute; 720 Broadway Seattle WA 98103 USA
| | - Sarah Y. Kwon
- The Sprott Centre for Stem Cell Research; Regenerative Medicine Program; Ottawa Hospital Research Institute; Ottawa Ontario Canada K1H 8L6
- Department of Chemical Engineering; University of Toronto; Toronto Ontario Canada
| | - Carole Doré
- The Sprott Centre for Stem Cell Research; Regenerative Medicine Program; Ottawa Hospital Research Institute; Ottawa Ontario Canada K1H 8L6
| | - Angela Raymond
- The Sprott Centre for Stem Cell Research; Regenerative Medicine Program; Ottawa Hospital Research Institute; Ottawa Ontario Canada K1H 8L6
| | - Akitsu Hotta
- Center for iPS Cell Research and Application (CiRA); Kyoto University; Kyoto Japan
| | - James Ellis
- Program in Developmental and Stem Cell Biology; The Hospital for Sick Children; Toronto Ontario Canada
- Department of Molecular Genetics; University of Toronto; Toronto Ontario Canada
| | - Rita A. Kandel
- Pathology and Experimental Medicine; Mount Sinai Hospital; Toronto Ontario Canada
| | - F. Jeffrey Dilworth
- The Sprott Centre for Stem Cell Research; Regenerative Medicine Program; Ottawa Hospital Research Institute; Ottawa Ontario Canada K1H 8L6
- Department of Cellular and Molecular Medicine; University of Ottawa; Ottawa Ontario Canada
- Ottawa Institute of Systems Biology; Ottawa Ontario Canada
| | - Theodore J. Perkins
- The Sprott Centre for Stem Cell Research; Regenerative Medicine Program; Ottawa Hospital Research Institute; Ottawa Ontario Canada K1H 8L6
- Ottawa Bioinformatics Core Facility; The Sprott Centre for Stem Cell Research; Ottawa Hospital Research Institute; Ottawa Ontario Canada K1H 8L6
- Ottawa Institute of Systems Biology; Ottawa Ontario Canada
| | - Michael J. Hendzel
- Cross Cancer Institute and the Department of Experimental Oncology; Faculty of Medicine and Dentistry; University of Alberta; Edmonton Alberta Canada T6G 1Z2
| | - David J. Galas
- Pacific Northwest Diabetes Research Institute; 720 Broadway Seattle WA 98103 USA
| | - William L. Stanford
- The Sprott Centre for Stem Cell Research; Regenerative Medicine Program; Ottawa Hospital Research Institute; Ottawa Ontario Canada K1H 8L6
- Department of Cellular and Molecular Medicine; University of Ottawa; Ottawa Ontario Canada
- Department of Chemical Engineering; University of Toronto; Toronto Ontario Canada
- Department of Biochemistry, Microbiology and Immunology; University of Ottawa; Ottawa Ontario Canada
- Ottawa Institute of Systems Biology; Ottawa Ontario Canada
| |
Collapse
|
68
|
Epigenetics and Oxidative Stress in Aging. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:9175806. [PMID: 28808499 PMCID: PMC5541801 DOI: 10.1155/2017/9175806] [Citation(s) in RCA: 107] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Revised: 05/24/2017] [Accepted: 06/12/2017] [Indexed: 11/25/2022]
Abstract
Aging is a multifactorial process characterized by the progressive loss of physiological functions, leading to an increased vulnerability to age-associated diseases and finally to death. Several theories have been proposed to explain the nature of aging. One of the most known identifies the free radicals produced by the mitochondrial metabolism as the cause of cellular and DNA damage. However, there are also several evidences supporting that epigenetic modifications, such as DNA methylation, noncoding RNAs, and histone modifications, play a critical role in the molecular mechanism of aging. In this review, we explore the significance of these findings and argue how the interlinked effects of oxidative stress and epigenetics can explain the cause of age-related declines.
Collapse
|
69
|
Turgay Y, Medalia O. The structure of lamin filaments in somatic cells as revealed by cryo-electron tomography. Nucleus 2017. [PMID: 28635493 DOI: 10.1080/19491034.2017.1337622] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
Abstract
Metazoan nuclei are equipped with nuclear lamina - a thin layer of intermediate filaments (IFs) mostly built of nuclear lamins facing the inner nuclear membrane (INM). The nuclear lamina serves as an interaction hub for INM-proteins, soluble nuclear factors and DNA. It confers structural and mechanical stability to the nucleus, transduces mechanical forces and biochemical signals across the nuclear envelope (NE) and regulates the organization of chromatin. By using cryo-electron tomography (cryo-ET), we recently provided an unprecedented view into the 3D organization of lamin filaments within the lamina meshwork in mammalian somatic cells. Through implementation of averaging procedures, we resolved the rod and globular Ig-fold domains of lamin filaments. The density maps suggested that they assemble into 3.5 nm thick filaments. Our analysis revealed interesting structural differences between nucleoplasmic and cytoplasmic intermediate filaments, raising the question of which molecular cues define their assembly modes inside the cell.
Collapse
Affiliation(s)
- Y Turgay
- a Department of Biochemistry , University of Zurich , Zurich , Switzerland
| | - O Medalia
- a Department of Biochemistry , University of Zurich , Zurich , Switzerland.,b Department of Life Sciences and the National Institute for Biotechnology in the Negev , Ben-Gurion University , Beer-Sheva , Israel
| |
Collapse
|
70
|
Abstract
Chromosome ends are complex structures, which require a panel of factors for their elongation, replication, and protection. We describe here the mechanics of mammalian telomeres, dynamics and maintainance in relation to lamins. Multiple biochemical connections, including association of telomeres to the nuclear envelope and matrix, of telomeric proteins to lamins, and of lamin-associated proteins to chromosome ends, underline the interplay between lamins and telomeres. Paths toward senescence, such as defective telomere replication, altered heterochromatin organization, and impaired DNA repair, are common to lamins' and telomeres' dysfunction. The convergence of phenotypes can be interpreted through a model of dynamic, lamin-controlled functional platforms dedicated to the function of telomeres as fragile sites. The features of telomeropathies and laminopathies, and of animal models underline further overlapping aspects, including the alteration of stem cell compartments. We expect that future studies of basic biology and on aging will benefit from the analysis of this telomere-lamina interplay.
Collapse
Affiliation(s)
- Romina Burla
- a Dipartimento di Biologia e Biotecnologie "C. Darwin," Sapienza Università di Roma , Rome, Italy.,b Istituto di Biologia e Patologia Molecolari del CNR , Rome , Italy
| | - Mattia La Torre
- a Dipartimento di Biologia e Biotecnologie "C. Darwin," Sapienza Università di Roma , Rome, Italy
| | - Isabella Saggio
- a Dipartimento di Biologia e Biotecnologie "C. Darwin," Sapienza Università di Roma , Rome, Italy.,b Istituto di Biologia e Patologia Molecolari del CNR , Rome , Italy.,c Istituto Pasteur Fondazione Cenci Bolognetti , Rome , Italy
| |
Collapse
|
71
|
Successful Treatment of an Unusual Case of FPLD2: The Role of Roux-en-Y Gastric Bypass-Case Report and Literature Review. J Gastrointest Surg 2017; 21:739-743. [PMID: 27778252 DOI: 10.1007/s11605-016-3300-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Accepted: 10/04/2016] [Indexed: 02/07/2023]
Abstract
Familial partial lipodystrophy type 2 (FPLD2) is a rare disorder associated with LMNA gene mutations. It is usually marked by loss of subcutaneous fat on the limbs and trunk and severe insulin resistance. Scattered reports have indicated that Roux-en-Y bypass helps to control the diabetes mellitus in these patients. We present here a very unusual patient with FPLD2 who had life-threatening retroperitoneal and renal fat accumulation accompanied by bilateral renal cancers. Following cryotherapy of one renal cancer and a contralateral nephrectomy with debulking of the retroperitoneal fat, Roux-en-Y gastric bypass (RYGB) has successfully controlled the disease for 3 years. The clinical presentations and causes of FPLD are reviewed and the role of RYGB is discussed.
Collapse
|
72
|
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
|
73
|
Cofilin Regulates Nuclear Architecture through a Myosin-II Dependent Mechanotransduction Module. Sci Rep 2017; 7:40953. [PMID: 28102353 PMCID: PMC5244421 DOI: 10.1038/srep40953] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Accepted: 12/14/2016] [Indexed: 01/02/2023] Open
Abstract
Structural features of the nucleus including shape, size and deformability impact its function affecting normal cellular processes such as cell differentiation and pathological conditions such as tumor cell migration. Despite the fact that abnormal nuclear morphology has long been a defining characteristic for diseases such as cancer relatively little is known about the mechanisms that control normal nuclear architecture. Mounting evidence suggests close coupling between F-actin cytoskeletal organization and nuclear morphology however, mechanisms regulating this coupling are lacking. Here we identify that Cofilin/ADF-family F-actin remodeling proteins are essential for normal nuclear structure in different cell types. siRNA mediated silencing of Cofilin/ADF provokes striking nuclear defects including aberrant shapes, nuclear lamina disruption and reductions to peripheral heterochromatin. We provide evidence that these anomalies are primarily due to Rho kinase (ROCK) controlled excessive contractile myosin-II activity and not to elevated F-actin polymerization. Furthermore, we demonstrate a requirement for nuclear envelope LINC (linker of nucleoskeleton and cytoskeleton) complex proteins together with lamin A/C for nuclear aberrations induced by Cofilin/ADF loss. Our study elucidates a pivotal regulatory mechanism responsible for normal nuclear structure and which is expected to fundamentally influence nuclear function.
Collapse
|
74
|
Fuller HR, Gillingwater TH, Wishart TM. Commonality amid diversity: Multi-study proteomic identification of conserved disease mechanisms in spinal muscular atrophy. Neuromuscul Disord 2016; 26:560-9. [PMID: 27460344 DOI: 10.1016/j.nmd.2016.06.004] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Accepted: 06/03/2016] [Indexed: 01/09/2023]
Abstract
The neuromuscular disease spinal muscular atrophy (SMA) is a leading genetic cause of infant mortality, resulting from low levels of full-length survival motor neuron (SMN) protein. Despite having a good understanding of the underlying genetics of SMA, the molecular pathways downstream of SMN that regulate disease pathogenesis remain unclear. The identification of molecular perturbations downstream of SMN is required in order to fully understand the fundamental biological role(s) for SMN in cells and tissues of the body, as well as to develop a range of therapeutic targets for developing novel treatments for SMA. Recent developments in proteomic screening technologies have facilitated proteome-wide investigations of a range of SMA models and tissues, generating novel insights into disease mechanisms by highlighting conserved changes in a range of molecular pathways. Comparative analysis of distinct proteomic datasets reveals conserved changes in pathways converging on GAP43, GAPDH, NCAM, UBA1, LMNA, ANXA2 and COL6A3. Proteomic studies therefore represent a leading tool with which to dissect the molecular mechanisms of disease pathogenesis in SMA, serving to identify potentially attractive targets for the development of novel therapies.
Collapse
Affiliation(s)
- Heidi R Fuller
- Wolfson Centre for Inherited Neuromuscular Disease, RJAH Orthopaedic Hospital, Oswestry SY10 7AG, UK; Institute for Science and Technology in Medicine, Keele University, Staffordshire ST5 5BG, UK.
| | - Thomas H Gillingwater
- Centre for Integrative Physiology, University of Edinburgh, UK; Euan MacDonald Centre for Motor Neurone Disease Research, University of Edinburgh, UK
| | - Thomas M Wishart
- Euan MacDonald Centre for Motor Neurone Disease Research, University of Edinburgh, UK; Division of Neurobiology, The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, UK.
| |
Collapse
|
75
|
Robin JD, Magdinier F. Physiological and Pathological Aging Affects Chromatin Dynamics, Structure and Function at the Nuclear Edge. Front Genet 2016; 7:153. [PMID: 27602048 PMCID: PMC4993774 DOI: 10.3389/fgene.2016.00153] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2016] [Accepted: 08/08/2016] [Indexed: 01/29/2023] Open
Abstract
Lamins are intermediate filaments that form a complex meshwork at the inner nuclear membrane. Mammalian cells express two types of Lamins, Lamins A/C and Lamins B, encoded by three different genes, LMNA, LMNB1, and LMNB2. Mutations in the LMNA gene are associated with a group of phenotypically diverse diseases referred to as laminopathies. Lamins interact with a large number of binding partners including proteins of the nuclear envelope but also chromatin-associated factors. Lamins not only constitute a scaffold for nuclear shape, rigidity and resistance to stress but also contribute to the organization of chromatin and chromosomal domains. We will discuss here the impact of A-type Lamins loss on alterations of chromatin organization and formation of chromatin domains and how disorganization of the lamina contributes to the patho-physiology of premature aging syndromes.
Collapse
Affiliation(s)
- Jérôme D Robin
- IRCAN, CNRS UMR 7284/INSERM U1081, Faculté de Médecine Nice, France
| | | |
Collapse
|
76
|
All-trans retinoic acid and rapamycin normalize Hutchinson Gilford progeria fibroblast phenotype. Oncotarget 2016; 6:29914-28. [PMID: 26359359 PMCID: PMC4745772 DOI: 10.18632/oncotarget.4939] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Accepted: 07/31/2015] [Indexed: 11/25/2022] Open
Abstract
Hutchinson Gilford progeria syndrome is a fatal disorder characterized by accelerated aging, bone resorption and atherosclerosis, caused by a LMNA mutation which produces progerin, a mutant lamin A precursor. Progeria cells display progerin and prelamin A nuclear accumulation, altered histone methylation pattern, heterochromatin loss, increased DNA damage and cell cycle alterations. Since the LMNA promoter contains a retinoic acid responsive element, we investigated if all-trans retinoic acid administration could lower progerin levels in cultured fibroblasts. We also evaluated the effect of associating rapamycin, which induces autophagic degradation of progerin and prelamin A. We demonstrate that all-trans retinoic acid acts synergistically with low-dosage rapamycin reducing progerin and prelamin A, via transcriptional downregulation associated with protein degradation, and increasing the lamin A to progerin ratio. These effects rescue cell dynamics and cellular proliferation through recovery of DNA damage response factor PARP1 and chromatin-associated nuclear envelope proteins LAP2α and BAF. The combined all-trans retinoic acid-rapamycin treatment is dramatically efficient, highly reproducible, represents a promising new approach in Hutchinson-Gilford Progeria therapy and deserves investigation in ageing-associated disorders.
Collapse
|
77
|
Skeletal Muscle Laminopathies: A Review of Clinical and Molecular Features. Cells 2016; 5:cells5030033. [PMID: 27529282 PMCID: PMC5040975 DOI: 10.3390/cells5030033] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Revised: 06/01/2016] [Accepted: 06/08/2016] [Indexed: 01/12/2023] Open
Abstract
LMNA-related disorders are caused by mutations in the LMNA gene, which encodes for the nuclear envelope proteins, lamin A and C, via alternative splicing. Laminopathies are associated with a wide range of disease phenotypes, including neuromuscular, cardiac, metabolic disorders and premature aging syndromes. The most frequent diseases associated with mutations in the LMNA gene are characterized by skeletal and cardiac muscle involvement. This review will focus on genetics and clinical features of laminopathies affecting primarily skeletal muscle. Although only symptomatic treatment is available for these patients, many achievements have been made in clarifying the pathogenesis and improving the management of these diseases.
Collapse
|
78
|
Modulation of TGFbeta 2 levels by lamin A in U2-OS osteoblast-like cells: understanding the osteolytic process triggered by altered lamins. Oncotarget 2016; 6:7424-37. [PMID: 25823658 PMCID: PMC4480690 DOI: 10.18632/oncotarget.3232] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Accepted: 01/28/2015] [Indexed: 01/17/2023] Open
Abstract
Transforming growth factor beta (TGFbeta) plays an essential role in bone homeostasis and deregulation of TGFbeta occurs in bone pathologies. Patients affected by Mandibuloacral Dysplasia (MADA), a progeroid disease linked to LMNA mutations, suffer from an osteolytic process. Our previous work showed that MADA osteoblasts secrete excess amount of TGFbeta 2, which in turn elicits differentiation of human blood precursors into osteoclasts. Here, we sought to determine how altered lamin A affects TGFbeta signaling. Our results show that wild-type lamin A negatively modulates TGFbeta 2 levels in osteoblast-like U2-OS cells, while the R527H mutated prelamin A as well as farnesylated prelamin A do not, ultimately leading to increased secretion of TGFbeta 2. TGFbeta 2 in turn, triggers the Akt/mTOR pathway and upregulates osteoprotegerin and cathepsin K. TGFbeta 2 neutralization rescues Akt/mTOR activation and the downstream transcriptional effects, an effect also obtained by statins or RAD001 treatment. Our results unravel an unexpected role of lamin A in TGFbeta 2 regulation and indicate rapamycin analogs and neutralizing antibodies to TGFbeta 2 as new potential therapeutic tools for MADA.
Collapse
|
79
|
Abstract
Mechanoresponses in mesenchymal stem cells (MSCs) guide both differentiation and function. In this review, we focus on advances in0 our understanding of how the cytoplasmic cytoskeleton, nuclear envelope and nucleoskeleton, which are connected via LINC (Linker of Nucleoskeleton and Cytoskeleton) complexes, are emerging as an integrated dynamic signaling platform to regulate MSC mechanobiology. This dynamic interconnectivity affects mechanical signaling and transfer of signals into the nucleus. In this way, nuclear and LINC-mediated cytoskeletal connectivity play a critical role in maintaining mechanical signaling that affects MSC fate by serving as both mechanosensory and mechanoresponsive structures. We review disease and age related compromises of LINC complexes and nucleoskeleton that contribute to the etiology of musculoskeletal diseases. Finally we invite the idea that acquired dysfunctions of LINC might be a contributing factor to conditions such as aging, microgravity and osteoporosis and discuss potential mechanical strategies to modulate LINC connectivity to combat these conditions.
Collapse
|
80
|
Milone MR, Pucci B, Bifulco K, Iannelli F, Lombardi R, Ciardiello C, Bruzzese F, Carriero MV, Budillon A. Proteomic analysis of zoledronic-acid resistant prostate cancer cells unveils novel pathways characterizing an invasive phenotype. Oncotarget 2016; 6:5324-41. [PMID: 25481874 PMCID: PMC4467152 DOI: 10.18632/oncotarget.2694] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2014] [Accepted: 11/04/2014] [Indexed: 12/22/2022] Open
Abstract
Proteomic analysis identified differentially expressed proteins between zoledronic acid-resistant and aggressive DU145R80 prostate cancer (PCa) cells and their parental DU145 cells. Ingenuity Pathway Analysis (IPA) showed a strong relationship between the identified proteins within a network associated with cancer and with homogeneous cellular functions prevalently related with regulation of cell organization, movement and consistent with the smaller and reduced cell-cell contact morphology of DU145R80 cells. The identified proteins correlated in publically available human PCa genomic data with increased tumor expression and aggressiveness. DU145R80 exhibit also a clear increase of alpha-v-(αv) integrin, and of urokinase receptor (uPAR), both included within the same network of the identified proteins. Interestingly, the actin-rich structures localized at the cell periphery of DU145R80 cells are rich of Filamin A, one of the identified proteins and uPAR which, in turn, co-localizes with αv-integrin, in podosomes and/or invadopodia. Notably, the invasive feature of DU145R80 may be prevented by blocking anti-αv antibody. Overall, we unveil a signaling network that physically links the interior of the nucleus via the cytoskeleton to the extracellular matrix and that could dictate PCa aggressiveness suggesting novel potential prognostic markers and therapeutic targets for PCa patients.
Collapse
Affiliation(s)
- Maria Rita Milone
- Centro Ricerche Oncologiche Mercogliano, Istituto Nazionale Tumori Fondazione G. Pascale - IRCCS, Naples, Italy
| | - Biagio Pucci
- Centro Ricerche Oncologiche Mercogliano, Istituto Nazionale Tumori Fondazione G. Pascale - IRCCS, Naples, Italy
| | - Katia Bifulco
- Neoplastic Progression Unit, Istituto Nazionale Tumori Fondazione G. Pascale - IRCCS, Naples, Italy
| | - Federica Iannelli
- Centro Ricerche Oncologiche Mercogliano, Istituto Nazionale Tumori Fondazione G. Pascale - IRCCS, Naples, Italy
| | - Rita Lombardi
- Centro Ricerche Oncologiche Mercogliano, Istituto Nazionale Tumori Fondazione G. Pascale - IRCCS, Naples, Italy
| | - Chiara Ciardiello
- Experimental Pharmacology Unit, Istituto Nazionale Tumori Fondazione G. Pascale - IRCCS, Naples, Italy
| | - Francesca Bruzzese
- Experimental Pharmacology Unit, Istituto Nazionale Tumori Fondazione G. Pascale - IRCCS, Naples, Italy
| | - Maria Vincenza Carriero
- Neoplastic Progression Unit, Istituto Nazionale Tumori Fondazione G. Pascale - IRCCS, Naples, Italy
| | - Alfredo Budillon
- Centro Ricerche Oncologiche Mercogliano, Istituto Nazionale Tumori Fondazione G. Pascale - IRCCS, Naples, Italy.,Experimental Pharmacology Unit, Istituto Nazionale Tumori Fondazione G. Pascale - IRCCS, Naples, Italy
| |
Collapse
|
81
|
Frost B, Bardai FH, Feany MB. Lamin Dysfunction Mediates Neurodegeneration in Tauopathies. Curr Biol 2015; 26:129-36. [PMID: 26725200 DOI: 10.1016/j.cub.2015.11.039] [Citation(s) in RCA: 168] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Revised: 10/19/2015] [Accepted: 11/11/2015] [Indexed: 10/22/2022]
Abstract
The filamentous meshwork formed by the lamin nucleoskeleton provides a scaffold for the anchoring of highly condensed heterochromatic DNA to the nuclear envelope, thereby establishing the three-dimensional architecture of the genome [1]. Insight into the importance of lamins to cellular viability can be gleaned from laminopathies, severe disorders caused by mutations in genes encoding lamins. A cellular consequence of lamin dysfunction in laminopathies is relaxation of heterochromatic DNA [1]. Similarly, we have recently reported the widespread relaxation of heterochromatin in tauopathies [1]: age-related progressive neurodegenerative disorders, including Alzheimer's disease, that are pathologically characterized by aggregates of phosphorylated tau protein in the brain [2, 3]. Here we demonstrate that acquired lamin misregulation though aberrant cytoskeletal-nucleoskeletal coupling promotes relaxation of heterochromatin and neuronal death in an in vivo model of neurodegenerative tauopathy. Genetic manipulation of lamin function significantly modifies neurodegeneration in vivo, demonstrating that lamin pathology plays a causal role in tau-mediated neurotoxicity. We show that lamin dysfunction is conserved in human tauopathy, as super-resolution microscopy reveals a significantly disrupted nuclear lamina in postmortem tissue from human Alzheimer's disease brain. Our study provides strong evidence that tauopathies are neurodegenerative laminopathies and identifies a new pathway mediating neuronal death in currently untreatable human neurodegenerative disorders, including Alzheimer's disease.
Collapse
Affiliation(s)
- Bess Frost
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Farah H Bardai
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Mel B Feany
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
82
|
Lattanzi G. Chromatin dynamics and in vitro biomarkers in laminopathies: an overview. Orphanet J Rare Dis 2015. [PMCID: PMC4652542 DOI: 10.1186/1750-1172-10-s2-o12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
|
83
|
Makarov AA, Rizzotto A, Meinke P, Schirmer EC. Purification of Lamins and Soluble Fragments of NETs. Methods Enzymol 2015; 569:79-100. [PMID: 26778554 DOI: 10.1016/bs.mie.2015.09.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Lamins and associated nuclear envelope transmembrane proteins (NETs) present unique problems for biochemical studies. Lamins form insoluble intermediate filament networks, associate with chromatin, and are also connected via specific NETs to the cytoskeleton, thus further complicating their isolation and purification from mammalian cells. Adding to this complexity, NETs at the inner nuclear membrane function in three distinct environments: (a) their nucleoplasmic domain(s) can bind lamins, chromatin, and transcriptional regulators; (b) they possess one or more integral transmembrane domains; and (c) their lumenal domain(s) function in the unique reducing environment of the nuclear envelope/ER lumen. This chapter describes strategic considerations and protocols to facilitate biochemical studies of lamins and NET proteins in vitro. Studying these proteins in vitro typically involves first expressing specific polypeptide fragments in bacteria and optimizing conditions to purify each fragment. We describe parameters for choosing specific fragments and designing purification strategies and provide detailed purification protocols. Biochemical studies can provide fundamental knowledge including binding strengths and the molecular consequences of disease-causing mutations that will be essential to understand nuclear envelope-genome interactions and nuclear envelope linked disease mechanisms.
Collapse
Affiliation(s)
- Alexandr A Makarov
- Wellcome Trust Centre for Cell Biology, University of Edinburgh, Edinburgh, United Kingdom
| | - Andrea Rizzotto
- Wellcome Trust Centre for Cell Biology, University of Edinburgh, Edinburgh, United Kingdom
| | - Peter Meinke
- Wellcome Trust Centre for Cell Biology, University of Edinburgh, Edinburgh, United Kingdom
| | - Eric C Schirmer
- Wellcome Trust Centre for Cell Biology, University of Edinburgh, Edinburgh, United Kingdom.
| |
Collapse
|
84
|
Awazu A. Nuclear dynamical deformation induced hetero- and euchromatin positioning. PHYSICAL REVIEW. E, STATISTICAL, NONLINEAR, AND SOFT MATTER PHYSICS 2015; 92:032709. [PMID: 26465500 DOI: 10.1103/physreve.92.032709] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Indexed: 06/05/2023]
Abstract
We studied the role of active deformation dynamics in cell nuclei in chromatin positioning. Model chains containing two types of regions, with high (euchromatic) or low (heterochromatic) mobility, were confined in a pulsating container simulating a nucleus showing dynamic deformations. Brownian dynamic simulations show that the positioning of low mobility regions changes from sites near the periphery to the center if the affinity between these regions and the container periphery disappears. The former and latter positionings are similar to the "conventional" and "inverted" chromatin positionings in nuclei of normal differentiated cells and cells lacking Lamin-related proteins. Additionally, nuclear dynamical deformation played essential roles in "inverted" chromatin positioning.
Collapse
Affiliation(s)
- Akinori Awazu
- Department of Mathematical and Life Sciences, Hiroshima University and Research Center for Mathematics on Chromatin Live Dynamics, Kagami-yama 1-3-1, Higashi-Hiroshima 739-8526, Japan
| |
Collapse
|
85
|
Mattout A, Cabianca DS, Gasser SM. Chromatin states and nuclear organization in development--a view from the nuclear lamina. Genome Biol 2015; 16:174. [PMID: 26303512 PMCID: PMC4549078 DOI: 10.1186/s13059-015-0747-5] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
The spatial distribution of chromatin domains in interphase nuclei changes dramatically during development in multicellular organisms. A crucial question is whether nuclear organization is a cause or a result of differentiation. Genetic perturbation of lamina–heterochromatin interactions is helping to reveal the cross-talk between chromatin states and nuclear organization.
Collapse
Affiliation(s)
- Anna Mattout
- Friedrich Miescher Institute for Biomedical Research, Maulbeerstrasse 66, CH-4058, Basel, Switzerland.
| | - Daphne S Cabianca
- Friedrich Miescher Institute for Biomedical Research, Maulbeerstrasse 66, CH-4058, Basel, Switzerland.
| | - Susan M Gasser
- Friedrich Miescher Institute for Biomedical Research, Maulbeerstrasse 66, CH-4058, Basel, Switzerland. .,University of Basel, Faculty of Natural Sciences, Klingelbergstrasse 50, CH-4056, Basel, Switzerland.
| |
Collapse
|
86
|
Krause M, Wolf K. Cancer cell migration in 3D tissue: negotiating space by proteolysis and nuclear deformability. Cell Adh Migr 2015; 9:357-66. [PMID: 26301444 DOI: 10.1080/19336918.2015.1061173] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Efficient tumor cell invasion into the surrounding desmoplastic stroma is a hallmark of cancer progression and involves the navigation through available small tissue spaces existent within the dense stromal network. Such navigation includes the reciprocal adaptation of the moving tumor cell, including the nucleus as largest and stiffest organelle, to pre-existent or de-novo generated extracellular matrix (ECM) gaps, pores and trails within stromal compartments. Within the context of migration, we briefly summarize physiological and tumor-related changes in ECM geometries as well as tissue proteolysis. We then focus on mechanisms that ensure the successful translocation of a nucleus through a confining pore by cytoskeleton-mediated coupling, as well as regulators of cell and nuclear deformability such as chromatin organization and nuclear lamina expression. In summary, understanding dynamic nuclear mechanics during migration in response to confined space will add to a better conceptual appreciation of cancer invasion and progression.
Collapse
Affiliation(s)
- Marina Krause
- a Department of Cell Biology ; Radboud University Medical Center ; Nijmegen , The Netherlands
| | - Katarina Wolf
- a Department of Cell Biology ; Radboud University Medical Center ; Nijmegen , The Netherlands
| |
Collapse
|
87
|
Piva R, Lambertini E, Manferdini C, Capanni C, Penolazzi L, Gabusi E, Paolella F, Lolli A, Angelozzi M, Lattanzi G, Lisignoli G. Slug transcription factor and nuclear Lamin B1 are upregulated in osteoarthritic chondrocytes. Osteoarthritis Cartilage 2015; 23:1226-30. [PMID: 25797039 DOI: 10.1016/j.joca.2015.03.015] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2014] [Revised: 02/20/2015] [Accepted: 03/12/2015] [Indexed: 02/02/2023]
Abstract
OBJECTIVE To contribute to clarify molecular mechanisms supporting senescence and de-differentiation of chondrocytes in chondrocyte pathologies such as osteoarthritis (OA). Specifically, we investigated the relationship between the nuclear lamina protein Lamin B1 and the negative regulator of chondrogenesis Slug transcription factor in osteoarthritic chondrocytes. METHODS Lamin B1 and Slug proteins were analyzed in cartilage explants from normal subjects and OA patients by immunohistochemical technique. Their expression was confirmed on isolated chondrocytes both at passage 0 and passage 2 (de-differentiated chondrocytes) by immunofluorescence and western blot. Subsequently, we explored the "in vivo" binding of Slug on LMNB1 promoter by chromatin immunoprecipitation assay (ChIP). RESULTS In this study we demonstrated that nuclear lamina protein Lamin B1 and anti-chondrogenic Slug transcription factor are upregulated in cartilage and OA chondrocytes. Furthermore, we found that Slug is "in vivo" recruited by LMNB1 gene promoter mostly when chondrocytes undergo de-differentiation or OA degeneration. CONCLUSIONS We described for the first time a potential regulatory role of Slug on the LMNB1 gene expression in OA chondrocytes. These findings may have important implications for the study of premature senescence, and degeneration of cartilage, and may contribute to develop effective therapeutic strategies against signals supporting cartilage damage in different subsets of patients.
Collapse
Affiliation(s)
- R Piva
- Department of Biomedical and Specialty Surgical Sciences, University of Ferrara, Ferrara, Italy.
| | - E Lambertini
- Department of Biomedical and Specialty Surgical Sciences, University of Ferrara, Ferrara, Italy
| | - C Manferdini
- Laboratorio di Immunoreumatologia e Rigenerazione Tissutale, IOR, Bologna, Italy; Laboratorio RAMSES, IOR, Bologna, Italy
| | - C Capanni
- Rizzoli Orthopedic Institute, Laboratory of Musculoskeletal Cell Biology, and CNR Institute for Molecular Genetics, Bologna, Italy
| | - L Penolazzi
- Department of Biomedical and Specialty Surgical Sciences, University of Ferrara, Ferrara, Italy
| | - E Gabusi
- Laboratorio RAMSES, IOR, Bologna, Italy
| | | | - A Lolli
- Department of Biomedical and Specialty Surgical Sciences, University of Ferrara, Ferrara, Italy
| | - M Angelozzi
- Department of Biomedical and Specialty Surgical Sciences, University of Ferrara, Ferrara, Italy
| | - G Lattanzi
- Rizzoli Orthopedic Institute, Laboratory of Musculoskeletal Cell Biology, and CNR Institute for Molecular Genetics, Bologna, Italy
| | - G Lisignoli
- Laboratorio di Immunoreumatologia e Rigenerazione Tissutale, IOR, Bologna, Italy; Laboratorio RAMSES, IOR, Bologna, Italy
| |
Collapse
|
88
|
The genetic basis for inherited forms of sinoatrial dysfunction and atrioventricular node dysfunction. J Interv Card Electrophysiol 2015; 43:121-34. [PMID: 25863800 PMCID: PMC4486151 DOI: 10.1007/s10840-015-9998-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Accepted: 03/13/2015] [Indexed: 01/01/2023]
Abstract
The sinoatrial node (SAN) and the atrioventricular node (AVN) are the anatomical and functional regions of the heart which play critical roles in the generation and conduction of the electrical impulse. Their functions are ensured by peculiar structural cytological properties and specific collections of ion channels. Impairment of SAN and AVN activity is generally acquired,but in some cases familial inheritance has been established and therefore a genetic cause is involved. In recent years, combined efforts of clinical practice and experimental basic science studies have identified and characterized several causative gene mutations associated with the nodal syndromes. Channelopathies, i.e., diseases associated with defective ion channels, remain the major cause of genetically determined nodal arrhythmias; however, it is becoming increasingly evident that mutations in other classes of regulatory and structural proteins also have profound pathophysiological roles. In this review, we will present some aspects of the genetic identification of the molecular mechanism underlying both SAN and AVN dysfunctions with a particular focus on mutations of the Na, pacemaker (HCN), and Ca channels. Genetic defects in regulatory proteins and calcium-handling proteins will be also considered. In conclusion, the identification of the genetic defects associated with familial nodal dysfunction is an essential step for implementing an appropriate therapeutic treatment.
Collapse
|
89
|
Torvaldson E, Kochin V, Eriksson JE. Phosphorylation of lamins determine their structural properties and signaling functions. Nucleus 2015; 6:166-71. [PMID: 25793944 DOI: 10.1080/19491034.2015.1017167] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Lamin A/C is part of the nuclear lamina, a meshwork of intermediate filaments underlying the inner nuclear membrane. The lamin network is anchoring a complex set of structural and linker proteins and is either directly or through partner proteins also associated or interacting with a number of signaling protein and transcription factors. During mitosis the nuclear lamina is dissociated by well established phosphorylation- dependent mechanisms. A-type lamins are, however, also phosphorylated during interphase. A recent study identified 20 interphase phosphorylation sites on lamin A/C and explored their functions related to lamin dynamics; movements, localization and solubility. Here we discuss these findings in the light of lamin functions in health and disease.
Collapse
Affiliation(s)
- Elin Torvaldson
- a Department of Biosciences; Åbo Akademi University ; Turku , Finland
| | | | | |
Collapse
|