1
|
Wang Y, Ruf S, Wang L, Heimerl T, Bange G, Groeger S. The Dual Roles of Lamin A/C in Macrophage Mechanotransduction. Cell Prolif 2025; 58:e13794. [PMID: 39710429 PMCID: PMC12099221 DOI: 10.1111/cpr.13794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 11/12/2024] [Accepted: 12/04/2024] [Indexed: 12/24/2024] Open
Abstract
Cellular mechanotransduction is a complex physiological process that integrates alterations in the external environment with cellular behaviours. In recent years, the role of the nucleus in mechanotransduction has gathered increased attention. Our research investigated the involvement of lamin A/C, a component of the nuclear envelope, in the mechanotransduction of macrophages under compressive force. We discovered that hydrostatic compressive force induces heterochromatin formation, decreases SUN1/SUN2 levels, and transiently downregulates lamin A/C. Notably, downregulated lamin A/C increased nuclear permeability to yes-associated protein 1 (YAP1), thereby amplifying certain effects of force, such as inflammation induction and proliferation inhibition. Additionally, lamin A/C deficiency detached the linker of nucleoskeleton and cytoskeleton (LINC) complex from nuclear envelope, consequently reducing force-induced DNA damage and IRF4 expression. In summary, lamin A/C exerted dual effects on macrophage responses to mechanical compression, promoting certain outcomes while inhibiting others. It operated through two distinct mechanisms: enhancing nuclear permeability and impairing intracellular mechanotransmission. The results of this study support the understanding of the mechanisms of intracellular mechanotransduction and may assist in identifying potential therapeutic targets for mechanotransduction-related diseases.
Collapse
Affiliation(s)
- Yao Wang
- Department of Orthodontics, Faculty of MedicineJustus Liebig UniversityGiessenGermany
| | - Sabine Ruf
- Department of Orthodontics, Faculty of MedicineJustus Liebig UniversityGiessenGermany
| | - Lei Wang
- Department of Orthodontics, Faculty of MedicineJustus Liebig UniversityGiessenGermany
- Department of Oral and Maxillofacial Surgery, Affiliated Stomatological HospitalSouthwest Medical UniversityLuzhouP. R. China
| | - Thomas Heimerl
- Center for Synthetic Microbiology (SYNMIKRO)Philipps‐University MarburgMarburgGermany
| | - Gert Bange
- Center for Synthetic Microbiology (SYNMIKRO)Philipps‐University MarburgMarburgGermany
| | - Sabine Groeger
- Department of Orthodontics, Faculty of MedicineJustus Liebig UniversityGiessenGermany
| |
Collapse
|
2
|
Marvi MV, Evangelisti C, Cerchier CB, Fazio A, Neri I, Koufi FD, Blalock W, Cenni V, Zoli M, Asioli S, Morandi L, Franceschi E, Manzoli L, Capanni C, Ratti S. Combining prelamin A accumulation and oxidative stress: A strategy to target glioblastoma. Eur J Cell Biol 2025; 104:151491. [PMID: 40305992 DOI: 10.1016/j.ejcb.2025.151491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Revised: 04/02/2025] [Accepted: 04/03/2025] [Indexed: 05/02/2025] Open
Abstract
Glioblastoma is the most aggressive and prevalent tumor of the Central Nervous System (CNS) with limited treatment options and poor patient outcomes. Standard therapies, including surgery, radiation, and chemotherapy, provide only modest survival benefits, highlighting the need for innovative therapeutic approaches. This study investigates a novel strategy targeting prelamin A processing in glioblastoma cells. By inhibiting the farnesyltransferase enzyme using SCH66336 (Lonafarnib), we promote the accumulation of lamin A precursor (prelamin A) in glioblastoma cells, thereby increasing their susceptibility to oxidative stress induced by Menadione administration, while sparing normal human astrocytes. Notably, the combined SCH66336-Menadione treatment reduced cell proliferation, modified the expression of stemness markers, and decreased viability in patient-derived glioblastoma stem cells, which represent the population responsible for tumor aggressiveness and recurrence. These findings indicate that inhibiting prelamin A processing could be a potential strategy to reduce glioblastoma aggressiveness and enhance therapeutic outcomes, particularly for treatment-resistant glioblastoma stem cell populations. This approach shows potential for integrating prelamin A processing disruption as a complementary strategy in glioblastoma therapy.
Collapse
Affiliation(s)
- Maria Vittoria Marvi
- Cellular Signalling Laboratory, Anatomy Centre, Department of Biomedical and Neuromotor Sciences (DIBINEM), Alma Mater Studiorum-University of Bologna, Bologna, Italy
| | - Camilla Evangelisti
- Cellular Signalling Laboratory, Anatomy Centre, Department of Biomedical and Neuromotor Sciences (DIBINEM), Alma Mater Studiorum-University of Bologna, Bologna, Italy
| | - Camilla Bruna Cerchier
- CNR Institute of Molecular Genetics "Luigi Luca Cavalli-Sforza", Unit of Bologna, Bologna, Italy; IRCCS Rizzoli Orthopedic Institute, Bologna, Italy
| | - Antonietta Fazio
- Cellular Signalling Laboratory, Anatomy Centre, Department of Biomedical and Neuromotor Sciences (DIBINEM), Alma Mater Studiorum-University of Bologna, Bologna, Italy
| | - Irene Neri
- Cellular Signalling Laboratory, Anatomy Centre, Department of Biomedical and Neuromotor Sciences (DIBINEM), Alma Mater Studiorum-University of Bologna, Bologna, Italy
| | - Foteini-Dionysia Koufi
- Cellular Signalling Laboratory, Anatomy Centre, Department of Biomedical and Neuromotor Sciences (DIBINEM), Alma Mater Studiorum-University of Bologna, Bologna, Italy
| | - William Blalock
- CNR Institute of Molecular Genetics "Luigi Luca Cavalli-Sforza", Unit of Bologna, Bologna, Italy; IRCCS Rizzoli Orthopedic Institute, Bologna, Italy
| | - Vittoria Cenni
- CNR Institute of Molecular Genetics "Luigi Luca Cavalli-Sforza", Unit of Bologna, Bologna, Italy; IRCCS Rizzoli Orthopedic Institute, Bologna, Italy
| | - Matteo Zoli
- Department of Biomedical and Neuromotor Sciences (DIBINEM), Alma Mater Studiorum-University of Bologna, Bologna, Italy; IRCCS Istituto delle Scienze Neurologiche di Bologna, Programma Neurochirurgia Ipofisi-Pituitary Unit, Bologna, Italy
| | - Sofia Asioli
- Department of Biomedical and Neuromotor Sciences (DIBINEM), Alma Mater Studiorum-University of Bologna, Bologna, Italy; IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
| | - Luca Morandi
- Department of Biomedical and Neuromotor Sciences (DIBINEM), Alma Mater Studiorum-University of Bologna, Bologna, Italy; Functional and Molecular Neuroimaging Unit, IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
| | - Enrico Franceschi
- Nervous System Medical Oncology Department, IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
| | - Lucia Manzoli
- Cellular Signalling Laboratory, Anatomy Centre, Department of Biomedical and Neuromotor Sciences (DIBINEM), Alma Mater Studiorum-University of Bologna, Bologna, Italy
| | - Cristina Capanni
- CNR Institute of Molecular Genetics "Luigi Luca Cavalli-Sforza", Unit of Bologna, Bologna, Italy; IRCCS Rizzoli Orthopedic Institute, Bologna, Italy.
| | - Stefano Ratti
- Cellular Signalling Laboratory, Anatomy Centre, Department of Biomedical and Neuromotor Sciences (DIBINEM), Alma Mater Studiorum-University of Bologna, Bologna, Italy.
| |
Collapse
|
3
|
Fu X, Zhang F, Dong X, Pu L, Feng Y, Xu Y, Gao F, Liang T, Kang J, Sun H, Hong T, Liu Y, Zhou H, Jiang J, Yin D, Hu X, Wang DZ, Ding J, Chen J. Adapting cytoskeleton-mitochondria patterning with myocyte differentiation by promyogenic PRR33. Cell Death Differ 2025; 32:177-193. [PMID: 39147882 PMCID: PMC11742405 DOI: 10.1038/s41418-024-01363-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 08/02/2024] [Accepted: 08/09/2024] [Indexed: 08/17/2024] Open
Abstract
Coordinated cytoskeleton-mitochondria organization during myogenesis is crucial for muscle development and function. Our understanding of the underlying regulatory mechanisms remains inadequate. Here, we identified a novel muscle-enriched protein, PRR33, which is upregulated during myogenesis and acts as a promyogenic factor. Depletion of Prr33 in C2C12 represses myoblast differentiation. Genetic deletion of Prr33 in mice reduces myofiber size and decreases muscle strength. The Prr33 mutant mice also exhibit impaired myogenesis and defects in muscle regeneration in response to injury. Interactome and transcriptome analyses reveal that PRR33 regulates cytoskeleton and mitochondrial function. Remarkably, PRR33 interacts with DESMIN, a key regulator of cytoskeleton-mitochondria organization in muscle cells. Abrogation of PRR33 in myocytes substantially abolishes the interaction of DESMIN filaments with mitochondria, leading to abnormal intracellular accumulation of DESMIN and mitochondrial disorganization/dysfunction in myofibers. Together, our findings demonstrate that PRR33 and DESMIN constitute an important regulatory module coordinating mitochondrial organization with muscle differentiation.
Collapse
Affiliation(s)
- Xuyang Fu
- Department of Cardiology of Second Affiliated Hospital, State Key Laboratory of Transvascular Implantation Devices, Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, 310009, China
- Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, 310029, China
| | - Feng Zhang
- Department of Cardiology of Second Affiliated Hospital, State Key Laboratory of Transvascular Implantation Devices, Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, 310009, China
- Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, 310029, China
| | - Xiaoxuan Dong
- Department of Cardiology of Second Affiliated Hospital, State Key Laboratory of Transvascular Implantation Devices, Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, 310009, China
- Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, 310029, China
| | - Linbin Pu
- Department of Cardiology of Second Affiliated Hospital, State Key Laboratory of Transvascular Implantation Devices, Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, 310009, China
- Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, 310029, China
| | - Yan Feng
- School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Yang Xu
- School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Feng Gao
- Department of Cardiology of Second Affiliated Hospital, State Key Laboratory of Transvascular Implantation Devices, Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, 310009, China
- Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, 310029, China
| | - Tian Liang
- Department of Cardiology of Second Affiliated Hospital, State Key Laboratory of Transvascular Implantation Devices, Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, 310009, China
- Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, 310029, China
| | - Jianmeng Kang
- Department of Cardiology of Second Affiliated Hospital, State Key Laboratory of Transvascular Implantation Devices, Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, 310009, China
- Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, 310029, China
| | - Hongke Sun
- Department of Cardiology of Second Affiliated Hospital, State Key Laboratory of Transvascular Implantation Devices, Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, 310009, China
- Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, 310029, China
| | - Tingting Hong
- Department of Cardiology of Second Affiliated Hospital, State Key Laboratory of Transvascular Implantation Devices, Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Yunxia Liu
- Department of Cardiology of Second Affiliated Hospital, State Key Laboratory of Transvascular Implantation Devices, Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Hongmei Zhou
- Department of Cardiology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Jun Jiang
- Department of Cardiology of Second Affiliated Hospital, State Key Laboratory of Transvascular Implantation Devices, Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Deling Yin
- Department of Cardiology of Second Affiliated Hospital, State Key Laboratory of Transvascular Implantation Devices, Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Xinyang Hu
- Department of Cardiology of Second Affiliated Hospital, State Key Laboratory of Transvascular Implantation Devices, Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Da-Zhi Wang
- University of South Florida Health Heart Institute, Center for Regenerative Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, 33602, USA
| | - Jian Ding
- School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China.
| | - Jinghai Chen
- Department of Cardiology of Second Affiliated Hospital, State Key Laboratory of Transvascular Implantation Devices, Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, 310009, China.
- Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, 310029, China.
| |
Collapse
|
4
|
Cenni V, Evangelisti C, Santi S, Sabatelli P, Neri S, Cavallo M, Lattanzi G, Mattioli E. Desmin and Plectin Recruitment to the Nucleus and Nuclei Orientation Are Lost in Emery-Dreifuss Muscular Dystrophy Myoblasts Subjected to Mechanical Stimulation. Cells 2024; 13:162. [PMID: 38247853 PMCID: PMC10814836 DOI: 10.3390/cells13020162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 01/08/2024] [Accepted: 01/11/2024] [Indexed: 01/23/2024] Open
Abstract
In muscle cells subjected to mechanical stimulation, LINC complex and cytoskeletal proteins are basic to preserve cellular architecture and maintain nuclei orientation and positioning. In this context, the role of lamin A/C remains mostly elusive. This study demonstrates that in human myoblasts subjected to mechanical stretching, lamin A/C recruits desmin and plectin to the nuclear periphery, allowing a proper spatial orientation of the nuclei. Interestingly, in Emery-Dreifuss Muscular Dystrophy (EDMD2) myoblasts exposed to mechanical stretching, the recruitment of desmin and plectin to the nucleus and nuclear orientation were impaired, suggesting that a functional lamin A/C is crucial for the response to mechanical strain. While describing a new mechanism of action headed by lamin A/C, these findings show a structural alteration that could be involved in the onset of the muscle defects observed in muscular laminopathies.
Collapse
Affiliation(s)
- Vittoria Cenni
- CNR Institute of Molecular Genetics “Luigi Luca Cavalli-Sforza”, Unit of Bologna, 40136 Bologna, Italy; (V.C.); (S.S.); (P.S.)
- IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy
| | - Camilla Evangelisti
- Cellular Signalling Laboratory, Department of Biochemical and Neuromotor Sciences, Alma Mater Studiorum, University of Bologna, 40138 Bologna, Italy;
| | - Spartaco Santi
- CNR Institute of Molecular Genetics “Luigi Luca Cavalli-Sforza”, Unit of Bologna, 40136 Bologna, Italy; (V.C.); (S.S.); (P.S.)
- IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy
| | - Patrizia Sabatelli
- CNR Institute of Molecular Genetics “Luigi Luca Cavalli-Sforza”, Unit of Bologna, 40136 Bologna, Italy; (V.C.); (S.S.); (P.S.)
- IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy
| | - Simona Neri
- Medicine and Rheumatology Unit, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy;
| | - Marco Cavallo
- Shoulder-Elbow Surgery Unit, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy;
| | - Giovanna Lattanzi
- CNR Institute of Molecular Genetics “Luigi Luca Cavalli-Sforza”, Unit of Bologna, 40136 Bologna, Italy; (V.C.); (S.S.); (P.S.)
- IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy
| | - Elisabetta Mattioli
- CNR Institute of Molecular Genetics “Luigi Luca Cavalli-Sforza”, Unit of Bologna, 40136 Bologna, Italy; (V.C.); (S.S.); (P.S.)
- IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy
| |
Collapse
|
5
|
Perales S, Sigamani V, Rajasingh S, Czirok A, Rajasingh J. Hutchinson-Gilford progeria patient-derived cardiomyocyte model of carrying LMNA gene variant c.1824 C > T. Cell Tissue Res 2023; 394:189-207. [PMID: 37572165 PMCID: PMC11956677 DOI: 10.1007/s00441-023-03813-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 07/12/2023] [Indexed: 08/14/2023]
Abstract
Cardiovascular diseases, atherosclerosis, and strokes are the most common causes of death in patients with Hutchinson-Gilford progeria syndrome (HGPS). The LMNA variant c.1824C > T accounts for ~ 90% of HGPS cases. The detailed molecular mechanisms of Lamin A in the heart remain elusive due to the lack of appropriate in vitro models. We hypothesize that HGPS patient's induced pluripotent stem cell (iPSC)-derived cardiomyocytes (iCMCs) will provide a model platform to study the cardio-pathologic mechanisms associated with HGPS. To elucidate the effects of progerin in cardiomyocytes, we first obtained skin fibroblasts (SFs) from a de-identified HGPS patient (hPGP1, proband) and both parents from the Progeria Research Foundation. Through Sanger sequencing and restriction fragment length polymorphism, with the enzyme EciI, targeting Lamin A, we characterized hPGP1-SFs as heterozygous mutants for the LMNA variant c.1824 C > T. Additionally, we performed LMNA exon 11 bisulfite sequencing to analyze the methylation status of the progeria cells. Furthermore, we reprogrammed the three SFs into iPSCs and differentiated them into iCMCs, which gained a beating on day 7. Through particle image velocimetry analysis, we found that hPGP1-iCMCs had an irregular contractile function and decreased cardiac-specific gene and protein expressions by qRT-PCR and Western blot. Our progeria-patient-derived iCMCs were found to be functionally and structurally defective when compared to normal iCMCs. This in vitro model will help in elucidating the role of Lamin A in cardiac diseases and the cardio-pathologic mechanisms associated with progeria. It provides a new platform for researchers to study novel treatment approaches for progeria-associated cardiac diseases.
Collapse
Affiliation(s)
- Selene Perales
- Department of Bioscience Research, University of Tennessee Health Science Center, 847 Monroe Avenue, Memphis, TN 38163, USA
| | - Vinoth Sigamani
- Department of Bioscience Research, University of Tennessee Health Science Center, 847 Monroe Avenue, Memphis, TN 38163, USA
- Department of Microbiology, Immunology, and Biochemistry, University of Tennessee Health Science Center, 847 Monroe Avenue, Memphis, TN 38163, USA
| | - Sheeja Rajasingh
- Department of Bioscience Research, University of Tennessee Health Science Center, 847 Monroe Avenue, Memphis, TN 38163, USA
| | - Andras Czirok
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Johnson Rajasingh
- Department of Bioscience Research, University of Tennessee Health Science Center, 847 Monroe Avenue, Memphis, TN 38163, USA.
- Department of Microbiology, Immunology, and Biochemistry, University of Tennessee Health Science Center, 847 Monroe Avenue, Memphis, TN 38163, USA.
- Department of Medicine-Cardiology, University of Tennessee Health Science Center, 847 Monroe Avenue, Memphis, TN, 38163, USA.
| |
Collapse
|
6
|
Abdelrahman SA, Khattab MA, Youssef MS, Mahmoud AA. Granulocyte-colony stimulating factor ameliorates di-ethylhexyl phthalate-induced cardiac muscle injury via stem cells recruitment, Desmin protein regulation, antifibrotic and antiapoptotic mechanisms. J Mol Histol 2023; 54:349-363. [PMID: 37428366 PMCID: PMC10412672 DOI: 10.1007/s10735-023-10137-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 06/25/2023] [Indexed: 07/11/2023]
Abstract
Phthalates are common plasticizers present in medical-grade plastics and other everyday products. Di-ethylhexyl phthalate (DEHP) has been noted as a causative risk factor for the initiation and augmentation of cardiovascular functional disorders. G-CSF is a glycoprotein found in numerous tissues throughout the body and is currently applied in clinical practice and has been tested in congestive heart failure. We aimed to examine in depth the effect of DEHP on the histological and biochemical structure of the cardiac muscle in adult male albino rats and the mechanisms underlying the possible ameliorative effect of G-CSF. Forty-eight adult male albino rats were divided into control group, DEHP group, DEHP+ G-CSF group and DEHP-recovery group. We measured serum levels of aspartate aminotransferase (AST), creatine kinase MB isoenzyme (CK-MB) and lactate dehydrogenase (LDH). Left ventricular sections were processed for light and electron microscope examination, and immunohistochemical staining of Desmin, activated Caspase-3 and CD34. DEHP significantly increased enzyme levels, markedly distorted the normal architecture of cardiac muscle fibers, downregulated Desmin protein levels and enhanced fibrosis, and apoptosis. G-CSF treatment significantly decreased the enzyme levels compared to DEHP group. It enhanced CD34 positive stem cells recruitment to injured cardiac muscle, therefore improved the ultrastructural features of most cardiac muscle fibers via anti-fibrotic and anti-apoptotic effects in addition to increased Desmin protein expression levels. The recovery group showed partial improvement due to persistent DEHP effect. In conclusion, administration of G-CSF effectively corrected the histopathological, immunohistochemical and biochemical alterations in the cardiac muscle after DEHP administration by stem cells recruitment, Desmin protein regulation, antifibrotic and antiapoptotic mechanisms.
Collapse
Affiliation(s)
- Shaimaa A Abdelrahman
- Medical Histology and Cell Biology Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt.
| | - Maha A Khattab
- Medical Histology and Cell Biology Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Marian S Youssef
- Medical Histology and Cell Biology Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Abeer A Mahmoud
- Medical Histology and Cell Biology Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| |
Collapse
|
7
|
Capanni C, Schena E, Di Giampietro ML, Montecucco A, Mattioli E, Lattanzi G. The role of prelamin A post-translational maturation in stress response and 53BP1 recruitment. Front Cell Dev Biol 2022; 10:1018102. [PMID: 36467410 PMCID: PMC9709412 DOI: 10.3389/fcell.2022.1018102] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Accepted: 10/24/2022] [Indexed: 11/25/2023] Open
Abstract
Lamin A is a main constituent of the nuclear lamina and contributes to nuclear shaping, mechano-signaling transduction and gene regulation, thus affecting major cellular processes such as cell cycle progression and entry into senescence, cellular differentiation and stress response. The role of lamin A in stress response is particularly intriguing, yet not fully elucidated, and involves prelamin A post-translational processing. Here, we propose prelamin A as the tool that allows lamin A plasticity during oxidative stress response and permits timely 53BP1 recruitment to DNA damage foci. We show that while PCNA ubiquitination, p21 decrease and H2AX phosphorylation occur soon after stress induction in the absence of prelamin A, accumulation of non-farnesylated prelamin A follows and triggers recruitment of 53BP1 to lamin A/C complexes. Then, the following prelamin A processing steps causing transient accumulation of farnesylated prelamin A and maturation to lamin A reduce lamin A affinity for 53BP1 and favor its release and localization to DNA damage sites. Consistent with these observations, accumulation of prelamin A forms in cells under basal conditions impairs histone H2AX phosphorylation, PCNA ubiquitination and p21 degradation, thus affecting the early stages of stress response. As a whole, our results are consistent with a physiological function of prelamin A modulation during stress response aimed at timely recruitment/release of 53BP1 and other molecules required for DNA damage repair. In this context, it becomes more obvious how farnesylated prelamin A accumulation to toxic levels alters timing of DNA damage signaling and 53BP1 recruitment, thus contributing to cellular senescence and accelerated organismal aging as observed in progeroid laminopathies.
Collapse
Affiliation(s)
- Cristina Capanni
- CNR Institute of Molecular Genetics “Luigi Luca Cavalli-Sforza”, Unit of Bologna, Bologna, Italy
- IRCCS Rizzoli Orthopedic Institute, Bologna, Italy
| | - Elisa Schena
- CNR Institute of Molecular Genetics “Luigi Luca Cavalli-Sforza”, Unit of Bologna, Bologna, Italy
- IRCCS Rizzoli Orthopedic Institute, Bologna, Italy
| | | | | | - Elisabetta Mattioli
- CNR Institute of Molecular Genetics “Luigi Luca Cavalli-Sforza”, Unit of Bologna, Bologna, Italy
- IRCCS Rizzoli Orthopedic Institute, Bologna, Italy
| | - Giovanna Lattanzi
- CNR Institute of Molecular Genetics “Luigi Luca Cavalli-Sforza”, Unit of Bologna, Bologna, Italy
- IRCCS Rizzoli Orthopedic Institute, Bologna, Italy
| |
Collapse
|
8
|
Wu X, Chu M, Ma X, Pei J, Xiong L, Guo X, Liang C, Yan P. Genome-Wide Identification of RNA Editing Sites Affecting Muscle Development in Yak. Front Vet Sci 2022; 9:871814. [PMID: 35836505 PMCID: PMC9274240 DOI: 10.3389/fvets.2022.871814] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Accepted: 06/07/2022] [Indexed: 12/24/2022] Open
Abstract
Skeletal muscle growth and development is a complicated process that is regulated at multiple steps and by numerous myogenesis genes. RNA editing represents one of the events at the post-transcriptional level, which contributes to the diversity of transcriptome and proteome by altering the nucleotides of RNAs. However, RNA editing events in the skeletal muscle of yaks are still not well defined. This study conducted whole-genome RNA-editing identification in skeletal muscle of yaks at embryonic stage (ES) and adult stage (AS). We found a total of 11,168 unique RNA editing sites, most of which were detected in the intergenic region. After annotation, we totally identified 2,718 editing sites within coding regions, among which 858 were missense changes. Moreover, totally 322 editing sites in the 3′ untranslated regions (UTR) were also predicted to alter the set of miRNA target sites, indicating that RNA editing may be involved in translational repression or mRNA degradation. We found 838 RNA editing sites (involving 244 common genes) that are edited differentially in ES as compared to AS. According to the KEGG enrichment analysis, these differentially edited genes were mainly involved in pathways highly related to skeletal muscle development and myogenesis, including MAPK, AMPK, Wnt, and PI3K-Akt signaling pathways. Altogether, our work presents the first characterization of RNA editing sites within yak skeletal muscles on a genome-wide scale and enhances our understanding of the mechanism of skeletal muscle development and myogenesis.
Collapse
|
9
|
Chiarini F, Paganelli F, Balestra T, Capanni C, Fazio A, Manara MC, Landuzzi L, Petrini S, Evangelisti C, Lollini PL, Martelli AM, Lattanzi G, Scotlandi K. Lamin A and the LINC complex act as potential tumor suppressors in Ewing Sarcoma. Cell Death Dis 2022; 13:346. [PMID: 35422060 PMCID: PMC9010457 DOI: 10.1038/s41419-022-04729-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 03/07/2022] [Accepted: 03/16/2022] [Indexed: 12/14/2022]
Abstract
Lamin A, a main constituent of the nuclear lamina, is involved in mechanosignaling and cell migration through dynamic interactions with the LINC complex, formed by the nuclear envelope proteins SUN1, SUN2 and the nesprins. Here, we investigated lamin A role in Ewing Sarcoma (EWS), an aggressive bone tumor affecting children and young adults. In patients affected by EWS, we found a significant inverse correlation between LMNA gene expression and tumor aggressiveness. Accordingly, in experimental in vitro models, low lamin A expression correlated with enhanced cell migration and invasiveness and, in vivo, with an increased metastatic load. At the molecular level, this condition was linked to altered expression and anchorage of nuclear envelope proteins and increased nuclear retention of YAP/TAZ, a mechanosignaling effector. Conversely, overexpression of lamin A rescued LINC complex organization, thus reducing YAP/TAZ nuclear recruitment and preventing cell invasiveness. These effects were also obtained through modulation of lamin A maturation by a statin-based pharmacological treatment that further elicited a more differentiated phenotype in EWS cells. These results demonstrate that drugs inducing nuclear envelope remodeling could be exploited to improve therapeutic strategies for EWS.
Collapse
Affiliation(s)
- Francesca Chiarini
- CNR Institute of Molecular Genetics "Luigi Luca Cavalli-Sforza", Unit of Bologna, 40136, Bologna, Italy. .,IRCCS Istituto Ortopedico Rizzoli, 40136, Bologna, Italy.
| | - Francesca Paganelli
- CNR Institute of Molecular Genetics "Luigi Luca Cavalli-Sforza", Unit of Bologna, 40136, Bologna, Italy.,Alma Mater Studiorum, University of Bologna, Department of Biomedical and Neuromotor Sciences, 40136, Bologna, Italy
| | - Tommaso Balestra
- IRCCS Istituto Ortopedico Rizzoli, Experimental Oncology Laboratory, 40136, Bologna, Italy.,Alma Mater Studiorum, University of Bologna, Department of Experimental, Diagnostic and Specialty Medicine, 40138, Bologna, Italy
| | - Cristina Capanni
- CNR Institute of Molecular Genetics "Luigi Luca Cavalli-Sforza", Unit of Bologna, 40136, Bologna, Italy.,IRCCS Istituto Ortopedico Rizzoli, 40136, Bologna, Italy
| | - Antonietta Fazio
- Alma Mater Studiorum, University of Bologna, Department of Biomedical and Neuromotor Sciences, 40136, Bologna, Italy
| | - Maria Cristina Manara
- IRCCS Istituto Ortopedico Rizzoli, Experimental Oncology Laboratory, 40136, Bologna, Italy
| | - Lorena Landuzzi
- IRCCS Istituto Ortopedico Rizzoli, Experimental Oncology Laboratory, 40136, Bologna, Italy
| | - Stefania Petrini
- Confocal Microscopy Core Facility, Research Center, Bambino Gesu' Children's Hospital IRCCS, 00146, Rome, Italy
| | - Camilla Evangelisti
- Alma Mater Studiorum, University of Bologna, Department of Biomedical and Neuromotor Sciences, 40136, Bologna, Italy
| | - Pier-Luigi Lollini
- Alma Mater Studiorum, University of Bologna, Department of Experimental, Diagnostic and Specialty Medicine, 40138, Bologna, Italy
| | - Alberto M Martelli
- Alma Mater Studiorum, University of Bologna, Department of Biomedical and Neuromotor Sciences, 40136, Bologna, Italy
| | - Giovanna Lattanzi
- CNR Institute of Molecular Genetics "Luigi Luca Cavalli-Sforza", Unit of Bologna, 40136, Bologna, Italy. .,IRCCS Istituto Ortopedico Rizzoli, 40136, Bologna, Italy.
| | - Katia Scotlandi
- IRCCS Istituto Ortopedico Rizzoli, Experimental Oncology Laboratory, 40136, Bologna, Italy.
| |
Collapse
|
10
|
Clinical Profile, Arrhythmias, and Adverse Cardiac Outcomes in Emery–Dreifuss Muscular Dystrophies: A Systematic Review of the Literature. BIOLOGY 2022; 11:biology11040530. [PMID: 35453731 PMCID: PMC9031530 DOI: 10.3390/biology11040530] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 03/23/2022] [Accepted: 03/24/2022] [Indexed: 11/16/2022]
Abstract
Cardiolaminopathies are a heterogeneous group of disorders which are due to mutations in the genes encoding for nuclear lamins or their binding proteins. The whole spectrum of cardiac manifestations encompasses atrial arrhythmias, conduction disturbances, progressive systolic dysfunction, and malignant ventricular arrhythmias. Despite the prognostic significance of cardiac involvement in this setting, the current recommendations lack strong evidence. The aim of our work was to systematically review the current data on the main cardiovascular outcomes in cardiolaminopathies. We searched PubMed/Embase for studies focusing on cardiovascular outcomes in LMNA mutation carriers (atrial arrhythmias, ventricular arrhythmias, sudden cardiac death, conduction disturbances, thromboembolic events, systolic dysfunction, heart transplantation, and all-cause and cardiovascular mortality). In total, 11 studies were included (1070 patients, mean age between 26–45 years, with follow-up periods ranging from 2.5 years up to 45 ± 12). When available, data on the EMD-mutated population were separately reported (40 patients). The incidence rates (IR) were individually assessed for the outcomes of interest. The IR for atrial fibrillation/atrial flutter/atrial tachycardia ranged between 6.1 and 13.9 events/100 pts–year. The IR of atrial standstill ranged between 0 and 2 events/100 pts-year. The IR for malignant ventricular arrhythmias reached 10.2 events/100 pts–year and 15.6 events/100 pts–year for appropriate implantable cardioverter–defibrillator (ICD) interventions. The IR for advanced conduction disturbances ranged between 3.2 and 7.7 events/100 pts–year. The IR of thromboembolic events reached up to 8.9 events/100 pts–year. Our results strengthen the need for periodic cardiological evaluation focusing on the early recognition of atrial arrhythmias, and possibly for the choice of preventive strategies for thromboembolic events. The frequent need for cardiac pacing due to advanced conduction disturbances should be counterbalanced with the high risk of malignant ventricular arrhythmias that would justify ICD over pacemaker implantation.
Collapse
|
11
|
Drosophila Nesprin-1 Isoforms Differentially Contribute to Muscle Function. Cells 2021; 10:cells10113061. [PMID: 34831284 PMCID: PMC8616381 DOI: 10.3390/cells10113061] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 10/27/2021] [Accepted: 10/28/2021] [Indexed: 12/26/2022] Open
Abstract
Nesprin-1 is a large scaffold protein connecting nuclei to the actin cytoskeleton via its KASH and Calponin Homology domains, respectively. Nesprin-1 disconnection from nuclei results in altered muscle function and myonuclei mispositioning. Furthermore, Nesprin-1 mutations are associated with muscular pathologies such as Emery Dreifuss muscular dystrophy and arthrogryposis. Nesprin-1 was thus proposed to mainly contribute to muscle function by controlling nuclei position. However, Nesprin-1′s localisation at sarcomere’s Z-discs, its involvement in organelles’ subcellular localization, as well as the description of numerous isoforms presenting different combinations of Calponin Homology (CH) and KASH domains, suggest that the contribution of Nesprin-1 to muscle functions is more complex. Here, we investigate the roles of Nesprin-1/Msp300 isoforms in muscle function and subcellular organisation using Drosophila larvae as a model. Subsets of Msp300 isoform were down-regulated by muscle-specific RNAi expression and muscle global function and morphology were assessed. We show that nuclei anchoring in mature muscle and global muscle function are disconnected functions associated with different Msp300 isoforms. Our work further uncovers a new and unsuspected role of Msp300 in myofibril registration and nuclei peripheral displacement supported by Msp300 CH containing isoforms, a function performed by Desmin in mammals.
Collapse
|
12
|
Ghasemizadeh A, Christin E, Guiraud A, Couturier N, Abitbol M, Risson V, Girard E, Jagla C, Soler C, Laddada L, Sanchez C, Jaque-Fernandez FI, Jacquemond V, Thomas JL, Lanfranchi M, Courchet J, Gondin J, Schaeffer L, Gache V. MACF1 controls skeletal muscle function through the microtubule-dependent localization of extra-synaptic myonuclei and mitochondria biogenesis. eLife 2021; 10:e70490. [PMID: 34448452 PMCID: PMC8500715 DOI: 10.7554/elife.70490] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 08/10/2021] [Indexed: 01/02/2023] Open
Abstract
Skeletal muscles are composed of hundreds of multinucleated muscle fibers (myofibers) whose myonuclei are regularly positioned all along the myofiber's periphery except the few ones clustered underneath the neuromuscular junction (NMJ) at the synaptic zone. This precise myonuclei organization is altered in different types of muscle disease, including centronuclear myopathies (CNMs). However, the molecular machinery regulating myonuclei position and organization in mature myofibers remains largely unknown. Conversely, it is also unclear how peripheral myonuclei positioning is lost in the related muscle diseases. Here, we describe the microtubule-associated protein, MACF1, as an essential and evolutionary conserved regulator of myonuclei positioning and maintenance, in cultured mammalian myotubes, in Drosophila muscle, and in adult mammalian muscle using a conditional muscle-specific knockout mouse model. In vitro, we show that MACF1 controls microtubules dynamics and contributes to microtubule stabilization during myofiber's maturation. In addition, we demonstrate that MACF1 regulates the microtubules density specifically around myonuclei, and, as a consequence, governs myonuclei motion. Our in vivo studies show that MACF1 deficiency is associated with alteration of extra-synaptic myonuclei positioning and microtubules network organization, both preceding NMJ fragmentation. Accordingly, MACF1 deficiency results in reduced muscle excitability and disorganized triads, leaving voltage-activated sarcoplasmic reticulum Ca2+ release and maximal muscle force unchanged. Finally, adult MACF1-KO mice present an improved resistance to fatigue correlated with a strong increase in mitochondria biogenesis.
Collapse
Affiliation(s)
- Alireza Ghasemizadeh
- Institut NeuroMyoGène, CNRS UMR5310, INSERM U1217, Faculté de Médecine Rockefeller, Université Claude Bernard Lyon ILyon CedexFrance
| | - Emilie Christin
- Institut NeuroMyoGène, CNRS UMR5310, INSERM U1217, Faculté de Médecine Rockefeller, Université Claude Bernard Lyon ILyon CedexFrance
| | - Alexandre Guiraud
- Institut NeuroMyoGène, CNRS UMR5310, INSERM U1217, Faculté de Médecine Rockefeller, Université Claude Bernard Lyon ILyon CedexFrance
| | - Nathalie Couturier
- Institut NeuroMyoGène, CNRS UMR5310, INSERM U1217, Faculté de Médecine Rockefeller, Université Claude Bernard Lyon ILyon CedexFrance
| | - Marie Abitbol
- Institut NeuroMyoGène, CNRS UMR5310, INSERM U1217, Faculté de Médecine Rockefeller, Université Claude Bernard Lyon ILyon CedexFrance
- Université Marcy l’Etoile, VetAgro SupLyonFrance
| | - Valerie Risson
- Institut NeuroMyoGène, CNRS UMR5310, INSERM U1217, Faculté de Médecine Rockefeller, Université Claude Bernard Lyon ILyon CedexFrance
| | - Emmanuelle Girard
- Institut NeuroMyoGène, CNRS UMR5310, INSERM U1217, Faculté de Médecine Rockefeller, Université Claude Bernard Lyon ILyon CedexFrance
| | - Christophe Jagla
- GReD Laboratory, Clermont-Auvergne University, INSERM U1103, CNRSClermont-FerrandFrance
| | - Cedric Soler
- GReD Laboratory, Clermont-Auvergne University, INSERM U1103, CNRSClermont-FerrandFrance
| | - Lilia Laddada
- GReD Laboratory, Clermont-Auvergne University, INSERM U1103, CNRSClermont-FerrandFrance
| | - Colline Sanchez
- Institut NeuroMyoGène, CNRS UMR5310, INSERM U1217, Faculté de Médecine Rockefeller, Université Claude Bernard Lyon ILyon CedexFrance
| | - Francisco-Ignacio Jaque-Fernandez
- Institut NeuroMyoGène, CNRS UMR5310, INSERM U1217, Faculté de Médecine Rockefeller, Université Claude Bernard Lyon ILyon CedexFrance
| | - Vincent Jacquemond
- Institut NeuroMyoGène, CNRS UMR5310, INSERM U1217, Faculté de Médecine Rockefeller, Université Claude Bernard Lyon ILyon CedexFrance
| | - Jean-Luc Thomas
- Institut NeuroMyoGène, CNRS UMR5310, INSERM U1217, Faculté de Médecine Rockefeller, Université Claude Bernard Lyon ILyon CedexFrance
| | - Marine Lanfranchi
- Institut NeuroMyoGène, CNRS UMR5310, INSERM U1217, Faculté de Médecine Rockefeller, Université Claude Bernard Lyon ILyon CedexFrance
| | - Julien Courchet
- Institut NeuroMyoGène, CNRS UMR5310, INSERM U1217, Faculté de Médecine Rockefeller, Université Claude Bernard Lyon ILyon CedexFrance
| | - Julien Gondin
- Institut NeuroMyoGène, CNRS UMR5310, INSERM U1217, Faculté de Médecine Rockefeller, Université Claude Bernard Lyon ILyon CedexFrance
| | - Laurent Schaeffer
- Institut NeuroMyoGène, CNRS UMR5310, INSERM U1217, Faculté de Médecine Rockefeller, Université Claude Bernard Lyon ILyon CedexFrance
| | - Vincent Gache
- Institut NeuroMyoGène, CNRS UMR5310, INSERM U1217, Faculté de Médecine Rockefeller, Université Claude Bernard Lyon ILyon CedexFrance
| |
Collapse
|
13
|
Bin Imtiaz MK, Jaeger BN, Bottes S, Machado RAC, Vidmar M, Moore DL, Jessberger S. Declining lamin B1 expression mediates age-dependent decreases of hippocampal stem cell activity. Cell Stem Cell 2021; 28:967-977.e8. [PMID: 33631115 DOI: 10.1016/j.stem.2021.01.015] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 11/19/2020] [Accepted: 01/21/2021] [Indexed: 01/04/2023]
Abstract
Neural stem cells (NSCs) generate neurons throughout life in the hippocampal dentate gyrus. With advancing age, levels of neurogenesis sharply drop, which has been associated with a decline in hippocampal memory function. However, cell-intrinsic mechanisms mediating age-related changes in NSC activity remain largely unknown. Here, we show that the nuclear lamina protein lamin B1 (LB1) is downregulated with age in mouse hippocampal NSCs, whereas protein levels of SUN-domain containing protein 1 (SUN1), previously implicated in Hutchinson-Gilford progeria syndrome (HGPS), increase. Balancing the levels of LB1 and SUN1 in aged NSCs restores the strength of the endoplasmic reticulum diffusion barrier that is associated with segregation of aging factors in proliferating NSCs. Virus-based restoration of LB1 expression in aged NSCs enhances stem cell activity in vitro and increases progenitor cell proliferation and neurogenesis in vivo. Thus, we here identify a mechanism that mediates age-related decline of neurogenesis in the mammalian hippocampus.
Collapse
Affiliation(s)
- Muhammad Khadeesh Bin Imtiaz
- Laboratory of Neural Plasticity, Faculties of Medicine and Science, Brain Research Institute, University of Zurich, 8057 Zurich, Switzerland
| | - Baptiste N Jaeger
- Laboratory of Neural Plasticity, Faculties of Medicine and Science, Brain Research Institute, University of Zurich, 8057 Zurich, Switzerland
| | - Sara Bottes
- Laboratory of Neural Plasticity, Faculties of Medicine and Science, Brain Research Institute, University of Zurich, 8057 Zurich, Switzerland
| | - Raquel A C Machado
- Laboratory of Neural Plasticity, Faculties of Medicine and Science, Brain Research Institute, University of Zurich, 8057 Zurich, Switzerland
| | - Mojca Vidmar
- Laboratory of Neural Plasticity, Faculties of Medicine and Science, Brain Research Institute, University of Zurich, 8057 Zurich, Switzerland
| | - Darcie L Moore
- Department of Neuroscience, University of Wisconsin-Madison, Madison, WI 53705, USA.
| | - Sebastian Jessberger
- Laboratory of Neural Plasticity, Faculties of Medicine and Science, Brain Research Institute, University of Zurich, 8057 Zurich, Switzerland.
| |
Collapse
|
14
|
Skeletal and Cardiac Muscle Disorders Caused by Mutations in Genes Encoding Intermediate Filament Proteins. Int J Mol Sci 2021; 22:ijms22084256. [PMID: 33923914 PMCID: PMC8073371 DOI: 10.3390/ijms22084256] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 04/12/2021] [Accepted: 04/15/2021] [Indexed: 02/08/2023] Open
Abstract
Intermediate filaments are major components of the cytoskeleton. Desmin and synemin, cytoplasmic intermediate filament proteins and A-type lamins, nuclear intermediate filament proteins, play key roles in skeletal and cardiac muscle. Desmin, encoded by the DES gene (OMIM *125660) and A-type lamins by the LMNA gene (OMIM *150330), have been involved in striated muscle disorders. Diseases include desmin-related myopathy and cardiomyopathy (desminopathy), which can be manifested with dilated, restrictive, hypertrophic, arrhythmogenic, or even left ventricular non-compaction cardiomyopathy, Emery–Dreifuss Muscular Dystrophy (EDMD2 and EDMD3, due to LMNA mutations), LMNA-related congenital Muscular Dystrophy (L-CMD) and LMNA-linked dilated cardiomyopathy with conduction system defects (CMD1A). Recently, mutations in synemin (SYNM gene, OMIM *606087) have been linked to cardiomyopathy. This review will summarize clinical and molecular aspects of desmin-, lamin- and synemin-related striated muscle disorders with focus on LMNA and DES-associated clinical entities and will suggest pathogenetic hypotheses based on the interplay of desmin and lamin A/C. In healthy muscle, such interplay is responsible for the involvement of this network in mechanosignaling, nuclear positioning and mitochondrial homeostasis, while in disease it is disturbed, leading to myocyte death and activation of inflammation and the associated secretome alterations.
Collapse
|
15
|
Charar C, Metsuyanim-Cohen S, Gruenbaum Y, Bar DZ. Exploring the nuclear lamina in health and pathology using C. elegans. Curr Top Dev Biol 2021; 144:91-110. [PMID: 33992162 DOI: 10.1016/bs.ctdb.2020.12.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The eukaryotic genome inside the nucleus is enveloped by two membranes, the Outer Nuclear Membrane (ONM) and the Inner Nuclear Membrane (INM). Tethered to the INM is the nuclear lamina, a fibrillar network composed of lamins-the nuclear intermediate filaments, and membrane associated proteins. The nuclear lamina interacts with several nuclear structures, including chromatin. As most nuclear functions, including regulation of gene expression, chromosome segregation and duplication as well as nuclear structure, are highly conserved in metazoans, the Caenorhabditis elegans nematode serves as a powerful model organism to study nuclear processes and architecture. This translucent organism can easily be observed under a microscope as a live embryo, larvae and even adult. Here we will review the data on nuclear lamina composition and functions gathered from studies using C. elegans model organisms: We will discuss genome spatial organization and its contribution to gene expression. We will review both the interaction between the cytoplasm and the nucleus and mechanotransduction mechanism. Finally, we will discuss disease causing mutation in nuclear lamins, including the use of this animal model in diseases research.
Collapse
Affiliation(s)
- Chayki Charar
- Department of Genetics, Institute of Life Sciences, Hebrew University of Jerusalem, Jerusalem, Israel; Department of Oral Biology, The Goldschleger School of Dental Medicine, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Sally Metsuyanim-Cohen
- Department of Oral Biology, The Goldschleger School of Dental Medicine, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Yosef Gruenbaum
- Department of Genetics, Institute of Life Sciences, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Daniel Z Bar
- Department of Oral Biology, The Goldschleger School of Dental Medicine, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
16
|
Squarzoni S, Schena E, Sabatelli P, Mattioli E, Capanni C, Cenni V, D'Apice MR, Andrenacci D, Sarli G, Pellegrino V, Festa A, Baruffaldi F, Storci G, Bonafè M, Barboni C, Sanapo M, Zaghini A, Lattanzi G. Interleukin-6 neutralization ameliorates symptoms in prematurely aged mice. Aging Cell 2021; 20:e13285. [PMID: 33393189 PMCID: PMC7811841 DOI: 10.1111/acel.13285] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 10/23/2020] [Accepted: 11/14/2020] [Indexed: 12/17/2022] Open
Abstract
Hutchinson-Gilford progeria syndrome (HGPS) causes premature aging in children, with adipose tissue, skin and bone deterioration, and cardiovascular impairment. In HGPS cells and mouse models, high levels of interleukin-6, an inflammatory cytokine linked to aging processes, have been detected. Here, we show that inhibition of interleukin-6 activity by tocilizumab, a neutralizing antibody raised against interleukin-6 receptors, counteracts progeroid features in both HGPS fibroblasts and LmnaG609G / G609G progeroid mice. Tocilizumab treatment limits the accumulation of progerin, the toxic protein produced in HGPS cells, rescues nuclear envelope and chromatin abnormalities, and attenuates the hyperactivated DNA damage response. In vivo administration of tocilizumab reduces aortic lesions and adipose tissue dystrophy, delays the onset of lipodystrophy and kyphosis, avoids motor impairment, and preserves a good quality of life in progeroid mice. This work identifies tocilizumab as a valuable tool in HGPS therapy and, speculatively, in the treatment of a variety of aging-related disorders.
Collapse
Affiliation(s)
- Stefano Squarzoni
- CNR Institute of Molecular Genetics “Luigi Luca Cavalli‐Sforza” Unit of Bologna Bologna Italy
- IRCCS Istituto Ortopedico Rizzoli Bologna Italy
| | - Elisa Schena
- CNR Institute of Molecular Genetics “Luigi Luca Cavalli‐Sforza” Unit of Bologna Bologna Italy
- IRCCS Istituto Ortopedico Rizzoli Bologna Italy
| | - Patrizia Sabatelli
- CNR Institute of Molecular Genetics “Luigi Luca Cavalli‐Sforza” Unit of Bologna Bologna Italy
- IRCCS Istituto Ortopedico Rizzoli Bologna Italy
| | - Elisabetta Mattioli
- CNR Institute of Molecular Genetics “Luigi Luca Cavalli‐Sforza” Unit of Bologna Bologna Italy
- IRCCS Istituto Ortopedico Rizzoli Bologna Italy
| | - Cristina Capanni
- CNR Institute of Molecular Genetics “Luigi Luca Cavalli‐Sforza” Unit of Bologna Bologna Italy
- IRCCS Istituto Ortopedico Rizzoli Bologna Italy
| | - Vittoria Cenni
- CNR Institute of Molecular Genetics “Luigi Luca Cavalli‐Sforza” Unit of Bologna Bologna Italy
- IRCCS Istituto Ortopedico Rizzoli Bologna Italy
| | | | - Davide Andrenacci
- CNR Institute of Molecular Genetics “Luigi Luca Cavalli‐Sforza” Unit of Bologna Bologna Italy
- IRCCS Istituto Ortopedico Rizzoli Bologna Italy
| | - Giuseppe Sarli
- Department of Veterinary Medical Sciences University of Bologna Bologna Italy
| | - Valeria Pellegrino
- Department of Veterinary Medical Sciences University of Bologna Bologna Italy
| | - Anna Festa
- Laboratory of Medical Technology IRCCS Istituto Ortopedico Rizzoli Bologna Italy
| | - Fabio Baruffaldi
- Laboratory of Medical Technology IRCCS Istituto Ortopedico Rizzoli Bologna Italy
| | - Gianluca Storci
- Department of Experimental, Diagnostic and Specialty Medicine University of Bologna Bologna Italy
| | - Massimiliano Bonafè
- Department of Experimental, Diagnostic and Specialty Medicine University of Bologna Bologna Italy
| | - Catia Barboni
- Department of Veterinary Medical Sciences University of Bologna Bologna Italy
| | - Mara Sanapo
- Department of Veterinary Medical Sciences University of Bologna Bologna Italy
| | - Anna Zaghini
- Department of Veterinary Medical Sciences University of Bologna Bologna Italy
| | - Giovanna Lattanzi
- CNR Institute of Molecular Genetics “Luigi Luca Cavalli‐Sforza” Unit of Bologna Bologna Italy
- IRCCS Istituto Ortopedico Rizzoli Bologna Italy
| |
Collapse
|
17
|
Cenni V, Capanni C, Mattioli E, Schena E, Squarzoni S, Bacalini MG, Garagnani P, Salvioli S, Franceschi C, Lattanzi G. Lamin A involvement in ageing processes. Ageing Res Rev 2020; 62:101073. [PMID: 32446955 DOI: 10.1016/j.arr.2020.101073] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 03/05/2020] [Accepted: 04/11/2020] [Indexed: 12/29/2022]
Abstract
Lamin A, a main constituent of the nuclear lamina, is the major splicing product of the LMNA gene, which also encodes lamin C, lamin A delta 10 and lamin C2. Involvement of lamin A in the ageing process became clear after the discovery that a group of progeroid syndromes, currently referred to as progeroid laminopathies, are caused by mutations in LMNA gene. Progeroid laminopathies include Hutchinson-Gilford Progeria, Mandibuloacral Dysplasia, Atypical Progeria and atypical-Werner syndrome, disabling and life-threatening diseases with accelerated ageing, bone resorption, lipodystrophy, skin abnormalities and cardiovascular disorders. Defects in lamin A post-translational maturation occur in progeroid syndromes and accumulated prelamin A affects ageing-related processes, such as mTOR signaling, epigenetic modifications, stress response, inflammation, microRNA activation and mechanosignaling. In this review, we briefly describe the role of these pathways in physiological ageing and go in deep into lamin A-dependent mechanisms that accelerate the ageing process. Finally, we propose that lamin A acts as a sensor of cell intrinsic and environmental stress through transient prelamin A accumulation, which triggers stress response mechanisms. Exacerbation of lamin A sensor activity due to stably elevated prelamin A levels contributes to the onset of a permanent stress response condition, which triggers accelerated ageing.
Collapse
Affiliation(s)
- Vittoria Cenni
- CNR Institute of Molecular Genetics "Luigi Luca Cavalli-Sforza", Unit of Bologna, Bologna, Italy; IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Cristina Capanni
- CNR Institute of Molecular Genetics "Luigi Luca Cavalli-Sforza", Unit of Bologna, Bologna, Italy; IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Elisabetta Mattioli
- CNR Institute of Molecular Genetics "Luigi Luca Cavalli-Sforza", Unit of Bologna, Bologna, Italy; IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Elisa Schena
- CNR Institute of Molecular Genetics "Luigi Luca Cavalli-Sforza", Unit of Bologna, Bologna, Italy; IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Stefano Squarzoni
- CNR Institute of Molecular Genetics "Luigi Luca Cavalli-Sforza", Unit of Bologna, Bologna, Italy; IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | | | - Paolo Garagnani
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy; Clinical Chemistry, Department of Laboratory Medicine, Karolinska Institutet at Huddinge, University Hospital, Stockholm, Sweden
| | - Stefano Salvioli
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy; Interdepartmental Center Alma Mater Research Institute on Global Challenges and Climate Changes, University of Bologna, Bologna, Italy
| | - Claudio Franceschi
- Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russia
| | - Giovanna Lattanzi
- CNR Institute of Molecular Genetics "Luigi Luca Cavalli-Sforza", Unit of Bologna, Bologna, Italy; IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy.
| |
Collapse
|
18
|
Liu J, Huang ZP, Nie M, Wang G, Silva WJ, Yang Q, Freire PP, Hu X, Chen H, Deng Z, Pu WT, Wang DZ. Regulation of myonuclear positioning and muscle function by the skeletal muscle-specific CIP protein. Proc Natl Acad Sci U S A 2020; 117:19254-19265. [PMID: 32719146 PMCID: PMC7430979 DOI: 10.1073/pnas.1922911117] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
The appropriate arrangement of myonuclei within skeletal muscle myofibers is of critical importance for normal muscle function, and improper myonuclear localization has been linked to a variety of skeletal muscle diseases, such as centronuclear myopathy and muscular dystrophies. However, the molecules that govern myonuclear positioning remain elusive. Here, we report that skeletal muscle-specific CIP (sk-CIP) is a regulator of nuclear positioning. Genetic deletion of sk-CIP in mice results in misalignment of myonuclei along the myofibers and at specialized structures such as neuromuscular junctions (NMJs) and myotendinous junctions (MTJs) in vivo, impairing myonuclear positioning after muscle regeneration, leading to severe muscle dystrophy in mdx mice, a mouse model of Duchenne muscular dystrophy. sk-CIP is localized to the centrosome in myoblasts and relocates to the outer nuclear envelope in myotubes upon differentiation. Mechanistically, we found that sk-CIP interacts with the Linker of Nucleoskeleton and Cytoskeleton (LINC) complex and the centriole Microtubule Organizing Center (MTOC) proteins to coordinately modulate myonuclear positioning and alignment. These findings indicate that sk-CIP may function as a muscle-specific anchoring protein to regulate nuclear position in multinucleated muscle cells.
Collapse
MESH Headings
- Animals
- Carrier Proteins/genetics
- Carrier Proteins/metabolism
- Cell Nucleus/genetics
- Cell Nucleus/metabolism
- Co-Repressor Proteins
- Humans
- Mice
- Mice, Inbred mdx
- Mice, Knockout
- Muscle, Skeletal/metabolism
- Muscle, Skeletal/physiopathology
- Myoblasts/metabolism
- Myopathies, Structural, Congenital/genetics
- Myopathies, Structural, Congenital/metabolism
- Myopathies, Structural, Congenital/physiopathology
- Nuclear Proteins/genetics
- Nuclear Proteins/metabolism
- Organ Specificity
Collapse
Affiliation(s)
- Jianming Liu
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115
| | - Zhan-Peng Huang
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115
- Center for Translational Medicine, National Health Commission (NHC) Key Laboratory of Assisted Circulation, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510275, China
| | - Mao Nie
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115
- Department of Orthopaedic Surgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, 400010, China
| | - Gang Wang
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115
| | - William J Silva
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115
- Laboratório de Biologia Celular e Molecular do Músculo Estriado, University of São Paulo, CEP 05508-000 Cidade Universitária, São Paulo, Brazil
| | - Qiumei Yang
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115
- Department of Animal Sciences, Sichuan Agriculture University, Chengdu, 611130, China
| | - Paula P Freire
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115
- Department of Morphology, Institute of Biosciences, São Paulo State University, CEP 18618-000, Botucatu, São Paulo, Brazil
| | - Xiaoyun Hu
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115
| | - Huaqun Chen
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115
- Department of Biology, Nanjing Normal University, Nanjing, 225300, China
| | - Zhongliang Deng
- Department of Orthopaedic Surgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, 400010, China
| | - William T Pu
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138
| | - Da-Zhi Wang
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115;
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138
| |
Collapse
|
19
|
PCAF Involvement in Lamin A/C-HDAC2 Interplay during the Early Phase of Muscle Differentiation. Cells 2020; 9:cells9071735. [PMID: 32698523 PMCID: PMC7409167 DOI: 10.3390/cells9071735] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 07/09/2020] [Accepted: 07/17/2020] [Indexed: 02/07/2023] Open
Abstract
Lamin A/C has been implicated in the epigenetic regulation of muscle gene expression through dynamic interaction with chromatin domains and epigenetic enzymes. We previously showed that lamin A/C interacts with histone deacetylase 2 (HDAC2). In this study, we deepened the relevance and regulation of lamin A/C-HDAC2 interaction in human muscle cells. We present evidence that HDAC2 binding to lamina A/C is related to HDAC2 acetylation on lysine 75 and expression of p300-CBP associated factor (PCAF), an acetyltransferase known to acetylate HDAC2. Our findings show that lamin A and farnesylated prelamin A promote PCAF recruitment to the nuclear lamina and lamin A/C binding in human myoblasts committed to myogenic differentiation, while protein interaction is decreased in differentiating myotubes. Interestingly, PCAF translocation to the nuclear envelope, as well as lamin A/C-PCAF interaction, are reduced by transient expression of lamin A mutated forms causing Emery Dreifuss muscular dystrophy. Consistent with this observation, lamin A/C interaction with both PCAF and HDAC2 is significantly reduced in Emery-Dreifuss muscular dystrophy myoblasts. Overall, these results support the view that, by recruiting PCAF and HDAC2 in a molecular platform, lamin A/C might contribute to regulate their epigenetic activity required in the early phase of muscle differentiation.
Collapse
|
20
|
Muscle cell differentiation and development pathway defects in Emery-Dreifuss muscular dystrophy. Neuromuscul Disord 2020; 30:443-456. [DOI: 10.1016/j.nmd.2020.04.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 03/20/2020] [Accepted: 04/15/2020] [Indexed: 12/12/2022]
|
21
|
Bostani M, Rahmati M, Mard SA. The effect of endurance training on levels of LINC complex proteins in skeletal muscle fibers of STZ-induced diabetic rats. Sci Rep 2020; 10:8738. [PMID: 32457392 PMCID: PMC7251114 DOI: 10.1038/s41598-020-65793-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Accepted: 05/11/2020] [Indexed: 02/07/2023] Open
Abstract
The changes of the linker of nucleoskeleton and cytoskeleton (LINC) complex have been studied in many muscular abnormality conditions; however, the effects of diabetes and physical activities on it have still remained to be defined. Therefore, the purpose of the this study was to evaluate the impacts of a six-week endurance training on the levels of SUN1 and Nesprin-1 proteins in Soleus and EDL muscles from diabetic wistar rats. A total number of 48 male Wistar rats (10 weeks, 200-250 gr) were randomly divided into healthy control (HC, N = 12), healthy trained (HT, N = 12), diabetic control (DC, N = 12), and diabetic trained (DT, N = 12) groups. Diabetes was also induced by a single intraperitoneally injection of streptozocin (45 mg/kg). The training groups ran a treadmill for five consecutive days within six weeks. The levels of the SUN1 and the Nesprin-1 proteins were further determined via ELISA method. The induction of diabetes had significantly decreased the levels of Nesprin-1 protein in the soleus and EDL muscles but it had no effects on the SUN1 in these muscles. As well, the findings revealed that six weeks of endurance training had significantly increased the levels of Nesprin-1 in DT and HT groups in the soleus as well as the EDL muscles; however, it had no impacts on the SUN1 in these muscles. The muscle fiber cross-sectional area (CSA) and myonuclei also decreased in diabetic control rats in both studied muscles. The training further augmented these parameters in both studied muscles in HT and DT groups. The present study provides new evidence that diabetes changes Nesprin-1 protein levels in skeletal muscle and endurance exercise training can modify it.
Collapse
Affiliation(s)
- Mehdi Bostani
- Department of Physical Education, Ahvaz Branch, Islamic Azad University, Ahvaz, Iran
| | - Masoud Rahmati
- Department of Physical Education and Sport Sciences, Faculty of Literature and Human Sciences, Lorestan University, Khoramabad, Iran.
| | - Seyyed Ali Mard
- Alimentary Tract Research Center and Physiology Research Center, Department of Physiology, The School of Medicine, Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
22
|
Gómez-Domínguez D, Epifano C, de Miguel F, Castaño AG, Vilaplana-Martí B, Martín A, Amarilla-Quintana S, Bertrand AT, Bonne G, Ramón-Azcón J, Rodríguez-Milla MA, Pérez de Castro I. Consequences of Lmna Exon 4 Mutations in Myoblast Function. Cells 2020; 9:cells9051286. [PMID: 32455813 PMCID: PMC7291140 DOI: 10.3390/cells9051286] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 05/06/2020] [Accepted: 05/16/2020] [Indexed: 02/06/2023] Open
Abstract
Laminopathies are causally associated with mutations on the Lamin A/C gene (LMNA). To date, more than 400 mutations in LMNA have been reported in patients. These mutations are widely distributed throughout the entire gene and are associated with a wide range of phenotypes. Unfortunately, little is known about the mechanisms underlying the effect of the majority of these mutations. This is the case of more than 40 mutations that are located at exon 4. Using CRISPR/Cas9 technology, we generated a collection of Lmna exon 4 mutants in mouse C2C12 myoblasts. These cell models included different types of exon 4 deletions and the presence of R249W mutation, one of the human variants associated with a severe type of laminopathy, LMNA-associated congenital muscular dystrophy (L-CMD). We characterized these clones by measuring their nuclear circularity, myogenic differentiation capacity in 2D and 3D conditions, DNA damage, and levels of p-ERK and p-AKT (phosphorylated Mitogen-Activated Protein Kinase 1/3 and AKT serine/threonine kinase 1). Our results indicated that Lmna exon 4 mutants showed abnormal nuclear morphology. In addition, levels and/or subcellular localization of different members of the lamin and LINC (LInker of Nucleoskeleton and Cytoskeleton) complex were altered in all these mutants. Whereas no significant differences were observed for ERK and AKT activities, the accumulation of DNA damage was associated to the Lmna p.R249W mutant myoblasts. Finally, significant myogenic differentiation defects were detected in the Lmna exon 4 mutants. These results have key implications in the development of future therapeutic strategies for the treatment of laminopathies.
Collapse
Affiliation(s)
- Déborah Gómez-Domínguez
- Instituto de Investigación de Enfermedades Raras, Instituto de Salud Carlos III, Ctra. Majadahonda-Pozuelo km2.2, E-28029 Madrid, Spain; (D.G.-D.); (F.d.M.); (B.V.-M.); (A.M.); (M.A.R.-M.)
| | - Carolina Epifano
- Fundación Andrés Marcio, niños contra la laminopatía, C/Núñez de Balboa, 11, E-28001 Madrid, Spain;
| | - Fernando de Miguel
- Instituto de Investigación de Enfermedades Raras, Instituto de Salud Carlos III, Ctra. Majadahonda-Pozuelo km2.2, E-28029 Madrid, Spain; (D.G.-D.); (F.d.M.); (B.V.-M.); (A.M.); (M.A.R.-M.)
- Universidad Europea de Madrid, C/ Tajo, s/n, E-28670 Villaviciosa de Odón, Spain
| | - Albert García Castaño
- Institute for Bioengineering of Catalonia (IBEC), C/Baldiri Reixac, 10-12, E-08028 Barcelona, Spain; (A.G.C.); (J.R.-A.)
| | - Borja Vilaplana-Martí
- Instituto de Investigación de Enfermedades Raras, Instituto de Salud Carlos III, Ctra. Majadahonda-Pozuelo km2.2, E-28029 Madrid, Spain; (D.G.-D.); (F.d.M.); (B.V.-M.); (A.M.); (M.A.R.-M.)
| | - Alberto Martín
- Instituto de Investigación de Enfermedades Raras, Instituto de Salud Carlos III, Ctra. Majadahonda-Pozuelo km2.2, E-28029 Madrid, Spain; (D.G.-D.); (F.d.M.); (B.V.-M.); (A.M.); (M.A.R.-M.)
| | - Sandra Amarilla-Quintana
- Fundación de Investigación HM Hospitales, Plaza del Conde Valle Suchil, 2, E-28015 Madrid, Spain;
| | - Anne T Bertrand
- UMRS 974, Center of Research in Myology, Institut de Myologie, Sorbonne Université, INSERM, 75013 Paris, France; (A.T.B.); (G.B.)
| | - Gisèle Bonne
- UMRS 974, Center of Research in Myology, Institut de Myologie, Sorbonne Université, INSERM, 75013 Paris, France; (A.T.B.); (G.B.)
| | - Javier Ramón-Azcón
- Institute for Bioengineering of Catalonia (IBEC), C/Baldiri Reixac, 10-12, E-08028 Barcelona, Spain; (A.G.C.); (J.R.-A.)
- ICREA-Institució Catalana de Recerca i Estudis Avançats, 08010 Barcelona, Spain
| | - Miguel A Rodríguez-Milla
- Instituto de Investigación de Enfermedades Raras, Instituto de Salud Carlos III, Ctra. Majadahonda-Pozuelo km2.2, E-28029 Madrid, Spain; (D.G.-D.); (F.d.M.); (B.V.-M.); (A.M.); (M.A.R.-M.)
| | - Ignacio Pérez de Castro
- Instituto de Investigación de Enfermedades Raras, Instituto de Salud Carlos III, Ctra. Majadahonda-Pozuelo km2.2, E-28029 Madrid, Spain; (D.G.-D.); (F.d.M.); (B.V.-M.); (A.M.); (M.A.R.-M.)
- Correspondence: ; Tel.: +34-918223188
| |
Collapse
|
23
|
Lamin A and Prelamin A Counteract Migration of Osteosarcoma Cells. Cells 2020; 9:cells9030774. [PMID: 32235738 PMCID: PMC7140691 DOI: 10.3390/cells9030774] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Revised: 03/16/2020] [Accepted: 03/19/2020] [Indexed: 12/11/2022] Open
Abstract
A type lamins are fundamental components of the nuclear lamina. Changes in lamin A expression correlate with malignant transformation in several cancers. However, the role of lamin A has not been explored in osteosarcoma (OS). Here, we wanted to investigate the role of lamin A in normal osteoblasts (OBs) and OS cells. Thus, we studied the expression of lamin A/C in OS cells compared to OBs and evaluated the effects of lamin A overexpression in OS cell lines. We show that, while lamin A expression increases during osteoblast differentiation, all examined OS cell lines express lower lamin A levels relative to differentiated OBs. The condition of low LMNA expression confers to OS cells a significant increase in migration potential, while overexpression of lamin A reduces migration ability of OS cells. Moreover, overexpression of unprocessable prelamin A also reduces cell migration. In agreement with the latter finding, OS cells which accumulate the highest prelamin A levels upon inhibition of lamin A maturation by statins, had significantly reduced migration ability. Importantly, OS cells subjected to statin treatment underwent apoptotic cell death in a RAS-independent, lamin A-dependent manner. Our results show that pro-apoptotic effects of statins and statin inhibitory effect on OS cell migration are comparable to those obtained by prelamin A accumulation and further suggest that modulation of lamin A expression and post-translational processing can be a tool to decrease migration potential in OS cells.
Collapse
|
24
|
Cavaliere V, Lattanzi G, Andrenacci D. Silencing of Euchromatic Transposable Elements as a Consequence of Nuclear Lamina Dysfunction. Cells 2020; 9:cells9030625. [PMID: 32151001 PMCID: PMC7140440 DOI: 10.3390/cells9030625] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Revised: 02/28/2020] [Accepted: 03/02/2020] [Indexed: 02/07/2023] Open
Abstract
Transposable elements (TEs) are mobile genomic sequences that are normally repressed to avoid proliferation and genome instability. Gene silencing mechanisms repress TEs by RNA degradation or heterochromatin formation. Heterochromatin maintenance is therefore important to keep TEs silent. Loss of heterochromatic domains has been linked to lamin mutations, which have also been associated with derepression of TEs. In fact, lamins are structural components of the nuclear lamina (NL), which is considered a pivotal structure in the maintenance of heterochromatin domains at the nuclear periphery in a silent state. Here, we show that a lethal phenotype associated with Lamin loss-of-function mutations is influenced by Drosophila gypsy retrotransposons located in euchromatic regions, suggesting that NL dysfunction has also effects on active TEs located in euchromatic loci. In fact, expression analysis of different long terminal repeat (LTR) retrotransposons and of one non-LTR retrotransposon located near active genes shows that Lamin inactivation determines the silencing of euchromatic TEs. Furthermore, we show that the silencing effect on euchromatic TEs spreads to the neighboring genomic regions, with a repressive effect on nearby genes. We propose that NL dysfunction may have opposed regulatory effects on TEs that depend on their localization in active or repressed regions of the genome.
Collapse
Affiliation(s)
- Valeria Cavaliere
- Dipartimento di Farmacia e Biotecnologie, Alma Mater Studiorum Università di Bologna, 40126 Bologna, Italy;
| | - Giovanna Lattanzi
- CNR Institute of Molecular Genetics “Luigi-Luca Cavalli-Sforza”, Unit of Bologna, 40136 Bologna, Italy;
- IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy
| | - Davide Andrenacci
- CNR Institute of Molecular Genetics “Luigi-Luca Cavalli-Sforza”, Unit of Bologna, 40136 Bologna, Italy;
- IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy
- Correspondence:
| |
Collapse
|
25
|
Zironi I, Gavoçi E, Lattanzi G, Virelli A, Amorini F, Remondini D, Castellani G. BK channel overexpression on plasma membrane of fibroblasts from Hutchinson-Gilford progeria syndrome. Aging (Albany NY) 2019; 10:3148-3160. [PMID: 30398975 PMCID: PMC6286842 DOI: 10.18632/aging.101621] [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: 02/05/2018] [Accepted: 10/27/2018] [Indexed: 12/15/2022]
Abstract
Hutchinson-Gilford progeria syndrome (HGPS) is an extremely rare genetic disorder wherein symptoms resembling aspects of aging are manifested at a very early age. It is a genetic condition that occurs due to a de novo mutation in the LMNA gene encoding for the nuclear structural protein lamin A. The lamin family of proteins are thought to be involved in nuclear stability, chromatin structure and gene expression and this leads to heavy effects on the regulation and functionality of the cell machinery. The functional role of the large-conductance calcium-activated potassium channels (BKCa) is still unclear, but has been recently described a strong relationship with their membrane expression, progerin nuclear levels and the ageing process. In this study, we found that: i) the outward potassium membrane current amplitude and the fluorescence intensity of the BKCa channel probe showed higher values in human dermal fibroblast obtained from patients affected by HGPS if compared to that from healthy young subjects; ii) this result appears to correlate with a basic cellular activity such as the replicative boost. We suggest that studying the HGPS also from the electrophysiological point of view might reveal new clues about the normal process of aging.
Collapse
Affiliation(s)
- Isabella Zironi
- Department of Physics and Astronomy (D.I.F.A.) University of Bologna, Bologna, Italy.,Interdepartmental Centre "L. Galvani" for integrated studies of Bioinformatics, Biophysics and Biocomplexity (C.I.G.) University of Bologna, Bologna, Italy.,National Institute for Nuclear Physics (INFN), Bologna, Italy
| | - Entelë Gavoçi
- National Institute for Nuclear Physics (INFN), Bologna, Italy
| | - Giovanna Lattanzi
- CNR Institute of Molecular Genetics, Unit of Bologna, Bologna, Italy.,Rizzoli Orthopedic Institute, Bologna, Italy
| | - Angela Virelli
- Department of Physics and Astronomy (D.I.F.A.) University of Bologna, Bologna, Italy
| | - Fabrizio Amorini
- Department of Physics and Astronomy (D.I.F.A.) University of Bologna, Bologna, Italy
| | - Daniel Remondini
- Department of Physics and Astronomy (D.I.F.A.) University of Bologna, Bologna, Italy.,Interdepartmental Centre "L. Galvani" for integrated studies of Bioinformatics, Biophysics and Biocomplexity (C.I.G.) University of Bologna, Bologna, Italy.,National Institute for Nuclear Physics (INFN), Bologna, Italy
| | - Gastone Castellani
- Department of Physics and Astronomy (D.I.F.A.) University of Bologna, Bologna, Italy.,Interdepartmental Centre "L. Galvani" for integrated studies of Bioinformatics, Biophysics and Biocomplexity (C.I.G.) University of Bologna, Bologna, Italy.,National Institute for Nuclear Physics (INFN), Bologna, Italy
| |
Collapse
|
26
|
Bergqvist C, Niss F, Figueroa RA, Beckman M, Maksel D, Jafferali MH, Kulyté A, Ström AL, Hallberg E. Monitoring of chromatin organization in live cells by FRIC. Effects of the inner nuclear membrane protein Samp1. Nucleic Acids Res 2019; 47:e49. [PMID: 30793190 PMCID: PMC6511872 DOI: 10.1093/nar/gkz123] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 01/17/2019] [Accepted: 02/19/2019] [Indexed: 12/24/2022] Open
Abstract
In most cells, transcriptionally inactive heterochromatin is preferentially localized in the nuclear periphery and transcriptionally active euchromatin is localized in the nuclear interior. Different cell types display characteristic chromatin distribution patterns, which change dramatically during cell differentiation, proliferation, senescence and different pathological conditions. Chromatin organization has been extensively studied on a cell population level, but there is a need to understand dynamic reorganization of chromatin at the single cell level, especially in live cells. We have developed a novel image analysis tool that we term Fluorescence Ratiometric Imaging of Chromatin (FRIC) to quantitatively monitor dynamic spatiotemporal distribution of euchromatin and total chromatin in live cells. A vector (pTandemH) assures stoichiometrically constant expression of the histone variants Histone 3.3 and Histone 2B, fused to EGFP and mCherry, respectively. Quantitative ratiometric (H3.3/H2B) imaging displayed a concentrated distribution of heterochromatin in the periphery of U2OS cell nuclei. As proof of concept, peripheral heterochromatin responded to experimental manipulation of histone acetylation. We also found that peripheral heterochromatin depended on the levels of the inner nuclear membrane protein Samp1, suggesting an important role in promoting peripheral heterochromatin. Taken together, FRIC is a powerful and robust new tool to study dynamic chromatin redistribution in live cells.
Collapse
Affiliation(s)
- Cecilia Bergqvist
- Department of Biochemistry and Biophysics, Stockholm University, Svante Arrhenius väg 16B, SE-106 91 Stockholm, Sweden
| | - Frida Niss
- Department of Biochemistry and Biophysics, Stockholm University, Svante Arrhenius väg 16B, SE-106 91 Stockholm, Sweden
| | - Ricardo A Figueroa
- Department of Biochemistry and Biophysics, Stockholm University, Svante Arrhenius väg 16B, SE-106 91 Stockholm, Sweden
| | - Marie Beckman
- Department of Biochemistry and Biophysics, Stockholm University, Svante Arrhenius väg 16B, SE-106 91 Stockholm, Sweden.,Institute of Environmental Medicine, Karolinska Institutet SE-171 77 Sweden
| | - Danuta Maksel
- Monash Molecular Crystallisation Facility (MMCF), Monash University, VIC 3800, Australia
| | - Mohammed H Jafferali
- Department of Biochemistry and Biophysics, Stockholm University, Svante Arrhenius väg 16B, SE-106 91 Stockholm, Sweden
| | - Agné Kulyté
- Lipid laboratory, Department of Medicine, Karolinska Institutet, SE-141 57 Huddinge, Sweden
| | - Anna-Lena Ström
- Department of Biochemistry and Biophysics, Stockholm University, Svante Arrhenius väg 16B, SE-106 91 Stockholm, Sweden
| | - Einar Hallberg
- Department of Biochemistry and Biophysics, Stockholm University, Svante Arrhenius väg 16B, SE-106 91 Stockholm, Sweden
| |
Collapse
|
27
|
Loo TH, Ye X, Chai RJ, Ito M, Bonne G, Ferguson-Smith AC, Stewart CL. The mammalian LINC complex component SUN1 regulates muscle regeneration by modulating drosha activity. eLife 2019; 8:e49485. [PMID: 31686651 PMCID: PMC6853637 DOI: 10.7554/elife.49485] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Accepted: 10/27/2019] [Indexed: 01/13/2023] Open
Abstract
Here we show that a major muscle specific isoform of the murine LINC complex protein SUN1 is required for efficient muscle regeneration. The nucleoplasmic domain of the isoform specifically binds to and inhibits Drosha, a key component of the microprocessor complex required for miRNA synthesis. Comparison of the miRNA profiles between wildtype and SUN1 null myotubes identified a cluster of miRNAs encoded by a non-translated retrotransposon-like one antisense (Rtl1as) transcript that are decreased in the WT myoblasts due to SUN1 inhibition of Drosha. One of these miRNAs miR-127 inhibits the translation of the Rtl1 sense transcript, that encodes the retrotransposon-like one protein (RTL1), which is also required for muscle regeneration and is expressed in regenerating/dystrophic muscle. The LINC complex may therefore regulate gene expression during muscle regeneration by controlling miRNA processing. This provides new insights into the molecular pathology underlying muscular dystrophies and how the LINC complex may regulate mechanosignaling.
Collapse
Affiliation(s)
- Tsui Han Loo
- Developmental and Regenerative BiologyInstitute of Medical BiologySingaporeSingapore
| | - Xiaoqian Ye
- Developmental and Regenerative BiologyInstitute of Medical BiologySingaporeSingapore
| | - Ruth Jinfen Chai
- Developmental and Regenerative BiologyInstitute of Medical BiologySingaporeSingapore
| | - Mitsuteru Ito
- Department of GeneticsUniversity of CambridgeCambridgeUnited Kingdom
| | - Gisèle Bonne
- Center of Research in Myology, Institut de MyologieSorbonne Universités, UPMC Univ Paris 06, INSERM UMRS 974, CNRS FRE 3617ParisFrance
| | | | - Colin L Stewart
- Developmental and Regenerative BiologyInstitute of Medical BiologySingaporeSingapore
| |
Collapse
|
28
|
Bernasconi P, Carboni N, Ricci G, Siciliano G, Politano L, Maggi L, Mongini T, Vercelli L, Rodolico C, Biagini E, Boriani G, Ruggiero L, Santoro L, Schena E, Prencipe S, Evangelisti C, Pegoraro E, Morandi L, Columbaro M, Lanzuolo C, Sabatelli P, Cavalcante P, Cappelletti C, Bonne G, Muchir A, Lattanzi G. Elevated TGF β2 serum levels in Emery-Dreifuss Muscular Dystrophy: Implications for myocyte and tenocyte differentiation and fibrogenic processes. Nucleus 2019; 9:292-304. [PMID: 29693488 PMCID: PMC5973167 DOI: 10.1080/19491034.2018.1467722] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Among rare diseases caused by mutations in LMNA gene, Emery-Dreifuss Muscular Dystrophy type 2 and Limb-Girdle muscular Dystrophy 1B are characterized by muscle weakness and wasting, joint contractures, cardiomyopathy with conduction system disorders. Circulating biomarkers for these pathologies have not been identified. Here, we analyzed the secretome of a cohort of patients affected by these muscular laminopathies in the attempt to identify a common signature. Multiplex cytokine assay showed that transforming growth factor beta 2 (TGF β2) and interleukin 17 serum levels are consistently elevated in the vast majority of examined patients, while interleukin 6 and basic fibroblast growth factor are altered in subgroups of patients. Levels of TGF β2 are also increased in fibroblast and myoblast cultures established from patient biopsies as well as in serum from mice bearing the H222P Lmna mutation causing Emery-Dreifuss Muscular Dystrophy in humans. Both patient serum and fibroblast conditioned media activated a TGF β2-dependent fibrogenic program in normal human myoblasts and tenocytes and inhibited myoblast differentiation. Consistent with these results, a TGF β2 neutralizing antibody avoided fibrogenic marker activation and myogenesis impairment. Cell intrinsic TGF β2-dependent mechanisms were also determined in laminopathic cells, where TGF β2 activated AKT/mTOR phosphorylation. These data show that TGF β2 contributes to the pathogenesis of Emery-Dreifuss Muscular Dystrophy type 2 and Limb-Girdle muscular Dystrophy 1B and can be considered a potential biomarker of those diseases. Further, the evidence of TGF β2 pathogenetic effects in tenocytes provides the first mechanistic insight into occurrence of joint contractures in muscular laminopathies.
Collapse
Affiliation(s)
- Pia Bernasconi
- a Neurology IV - Neuroimmunology and Neuromuscular Diseases Unit , Foundation IRCCS Neurological Institute "Carlo Besta" , Milan , Italy
| | - Nicola Carboni
- b Neurology Department , Hospital San Francesco of Nuoro , Nuoro , Italy
| | - Giulia Ricci
- c Department of Clinical and Experimental Medicine , University of Pisa , Pisa , Italy
| | - Gabriele Siciliano
- c Department of Clinical and Experimental Medicine , University of Pisa , Pisa , Italy
| | - Luisa Politano
- d Cardiomyology and Medical Genetics, Department of Experimental Medicine , Campania University "Luigi Vanvitelli" (former denomination: Second University of Naples) , Italy
| | - Lorenzo Maggi
- a Neurology IV - Neuroimmunology and Neuromuscular Diseases Unit , Foundation IRCCS Neurological Institute "Carlo Besta" , Milan , Italy
| | - Tiziana Mongini
- e Department of Neurosciences "Rita Levi Montalcini" , University of Turin , Turin , Italy
| | - Liliana Vercelli
- e Department of Neurosciences "Rita Levi Montalcini" , University of Turin , Turin , Italy
| | - Carmelo Rodolico
- f Institute of Applied Sciences and Intelligent Systems "ISASI Edoardo Caianello", National Research Council of Italy , Messina , Italy
| | - Elena Biagini
- g Istituto di Cardiologia, Università di Bologna, Policlinico S.Orsola-Malpighi , Bologna , Italy
| | - Giuseppe Boriani
- h Cardiology Division, Department of Diagnostics , Clinical and Public Health Medicine, University of Modena and Reggio Emilia, Policlinico di Modena , Modena , Italy
| | - Lucia Ruggiero
- i Department of Neurosciences , Odontostomatological and Reproductive Sciences, University of Naples "Federico II" , Naples , Italy
| | - Lucio Santoro
- i Department of Neurosciences , Odontostomatological and Reproductive Sciences, University of Naples "Federico II" , Naples , Italy
| | - Elisa Schena
- j Institute of Molecular Genetics (IGM)-CNR, Unit of Bologna , Bologna , Italy.,k Laboratory of Musculoskeletal Cell Biology , Rizzoli Orthopaedic Institute , Bologna , Italy
| | - Sabino Prencipe
- j Institute of Molecular Genetics (IGM)-CNR, Unit of Bologna , Bologna , Italy.,k Laboratory of Musculoskeletal Cell Biology , Rizzoli Orthopaedic Institute , Bologna , Italy
| | - Camilla Evangelisti
- j Institute of Molecular Genetics (IGM)-CNR, Unit of Bologna , Bologna , Italy.,k Laboratory of Musculoskeletal Cell Biology , Rizzoli Orthopaedic Institute , Bologna , Italy
| | - Elena Pegoraro
- l Department of Neurosciences , Neuromuscular Center, University of Padova , Padova , Italy
| | - Lucia Morandi
- a Neurology IV - Neuroimmunology and Neuromuscular Diseases Unit , Foundation IRCCS Neurological Institute "Carlo Besta" , Milan , Italy
| | - Marta Columbaro
- k Laboratory of Musculoskeletal Cell Biology , Rizzoli Orthopaedic Institute , Bologna , Italy
| | - Chiara Lanzuolo
- m Istituto Nazionale di Genetica Molecolare "Romeo and Enrica Invernizzi" , Milan , Italy.,n Institute of Cell Biology and Neurobiology, IRCCS Santa Lucia Foundation , Rome , Italy
| | - Patrizia Sabatelli
- j Institute of Molecular Genetics (IGM)-CNR, Unit of Bologna , Bologna , Italy.,k Laboratory of Musculoskeletal Cell Biology , Rizzoli Orthopaedic Institute , Bologna , Italy
| | - Paola Cavalcante
- a Neurology IV - Neuroimmunology and Neuromuscular Diseases Unit , Foundation IRCCS Neurological Institute "Carlo Besta" , Milan , Italy
| | - Cristina Cappelletti
- a Neurology IV - Neuroimmunology and Neuromuscular Diseases Unit , Foundation IRCCS Neurological Institute "Carlo Besta" , Milan , Italy
| | - Gisèle Bonne
- o Sorbonne Universités , UPMC Univ Paris 06, INSERM UMRS974, CNRS FRE3617, Center for Research in Myology, Institut de Myologie, G.H. Pitié Salpêtrière , Paris Cedex 13, France
| | - Antoine Muchir
- o Sorbonne Universités , UPMC Univ Paris 06, INSERM UMRS974, CNRS FRE3617, Center for Research in Myology, Institut de Myologie, G.H. Pitié Salpêtrière , Paris Cedex 13, France
| | - Giovanna Lattanzi
- j Institute of Molecular Genetics (IGM)-CNR, Unit of Bologna , Bologna , Italy.,k Laboratory of Musculoskeletal Cell Biology , Rizzoli Orthopaedic Institute , Bologna , Italy
| |
Collapse
|
29
|
Ankrd2 in Mechanotransduction and Oxidative Stress Response in Skeletal Muscle: New Cues for the Pathogenesis of Muscular Laminopathies. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:7318796. [PMID: 31428229 PMCID: PMC6681624 DOI: 10.1155/2019/7318796] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 05/02/2019] [Accepted: 05/19/2019] [Indexed: 12/11/2022]
Abstract
Ankrd2 (ankyrin repeats containing domain 2) or Arpp (ankyrin repeat, PEST sequence, and proline-rich region) is a member of the muscle ankyrin repeat protein family. Ankrd2 is mostly expressed in skeletal muscle, where it plays an intriguing role in the transcriptional response to stress induced by mechanical stimulation as well as by cellular reactive oxygen species. Our studies in myoblasts from Emery-Dreifuss muscular dystrophy 2, a LMNA-linked disease affecting skeletal and cardiac muscles, demonstrated that Ankrd2 is a lamin A-binding protein and that mutated lamins found in Emery-Dreifuss muscular dystrophy change the dynamics of Ankrd2 nuclear import, thus affecting oxidative stress response. In this review, besides describing the latest advances related to Ankrd2 studies, including novel discoveries on Ankrd2 isoform-specific functions, we report the main findings on the relationship of Ankrd2 with A-type lamins and discuss known and potential mechanisms involving defective Ankrd2-lamin A interplay in the pathogenesis of muscular laminopathies.
Collapse
|
30
|
Abstract
Cellular behavior is continuously affected by microenvironmental forces through the process of mechanotransduction, in which mechanical stimuli are rapidly converted to biochemical responses. Mounting evidence suggests that the nucleus itself is a mechanoresponsive element, reacting to cytoskeletal forces and mediating downstream biochemical responses. The nucleus responds through a host of mechanisms, including partial unfolding, conformational changes, and phosphorylation of nuclear envelope proteins; modulation of nuclear import/export; and altered chromatin organization, resulting in transcriptional changes. It is unclear which of these events present direct mechanotransduction processes and which are downstream of other mechanotransduction pathways. We critically review and discuss the current evidence for nuclear mechanotransduction, particularly in the context of stem cell fate, a largely unexplored topic, and in disease, where an improved understanding of nuclear mechanotransduction is beginning to open new treatment avenues. Finally, we discuss innovative technological developments that will allow outstanding questions in the rapidly growing field of nuclear mechanotransduction to be answered.
Collapse
Affiliation(s)
- Melanie Maurer
- Meinig School of Biomedical Engineering and Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, New York 14853, USA; ,
| | - Jan Lammerding
- Meinig School of Biomedical Engineering and Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, New York 14853, USA; ,
| |
Collapse
|
31
|
The Cutting Edge: The Role of mTOR Signaling in Laminopathies. Int J Mol Sci 2019; 20:ijms20040847. [PMID: 30781376 PMCID: PMC6412338 DOI: 10.3390/ijms20040847] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 02/11/2019] [Accepted: 02/12/2019] [Indexed: 12/29/2022] Open
Abstract
The mechanistic target of rapamycin (mTOR) is a ubiquitous serine/threonine kinase that regulates anabolic and catabolic processes, in response to environmental inputs. The existence of mTOR in numerous cell compartments explains its specific ability to sense stress, execute growth signals, and regulate autophagy. mTOR signaling deregulation is closely related to aging and age-related disorders, among which progeroid laminopathies represent genetically characterized clinical entities with well-defined phenotypes. These diseases are caused by LMNA mutations and feature altered bone turnover, metabolic dysregulation, and mild to severe segmental progeria. Different LMNA mutations cause muscular, adipose tissue and nerve pathologies in the absence of major systemic involvement. This review explores recent advances on mTOR involvement in progeroid and tissue-specific laminopathies. Indeed, hyper-activation of protein kinase B (AKT)/mTOR signaling has been demonstrated in muscular laminopathies, and rescue of mTOR-regulated pathways increases lifespan in animal models of Emery-Dreifuss muscular dystrophy. Further, rapamycin, the best known mTOR inhibitor, has been used to elicit autophagy and degradation of mutated lamin A or progerin in progeroid cells. This review focuses on mTOR-dependent pathogenetic events identified in Emery-Dreifuss muscular dystrophy, LMNA-related cardiomyopathies, Hutchinson-Gilford Progeria, mandibuloacral dysplasia, and type 2 familial partial lipodystrophy. Pharmacological application of mTOR inhibitors in view of therapeutic strategies is also discussed.
Collapse
|
32
|
Wu J, Guan F, Luo W, Yuan Z, Chen R, Gou X, Shi X, Guo H, Fang K. Retracted
: Prelamin A overexpression promotes detrusor calcification/aging in urinary incontinence via prelamin A accumulation. J Cell Physiol 2019; 234:17800-17811. [DOI: 10.1002/jcp.28406] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Revised: 01/09/2019] [Accepted: 01/10/2019] [Indexed: 12/26/2022]
Affiliation(s)
- Jing Wu
- Department of Biochemistry and Molecular Biology The Primary Medicine School of Kunming Medical University Kunming China
| | - Fei Guan
- Department of Urology The Second Affiliated Hospital of Kunming Medical University Kunming P. R. China
| | - Wei Luo
- Department of Urology The Second Affiliated Hospital of Kunming Medical University Kunming P. R. China
| | - Zhi‐Wei Yuan
- Department of Pathology The Second Affiliated Hospital of Kunming Medical University Kunming P. R. China
| | - Rong‐Qiong Chen
- Department of Urology The Second Affiliated Hospital of Kunming Medical University Kunming P. R. China
| | - Xin Gou
- Department of Urology The Second Affiliated Hospital of Kunming Medical University Kunming P. R. China
| | - Xin Shi
- Department of Urology The Second Affiliated Hospital of Kunming Medical University Kunming P. R. China
| | - Hai‐Xiang Guo
- Department of Urology The Second Affiliated Hospital of Kunming Medical University Kunming P. R. China
| | - Ke‐Wei Fang
- Department of Urology The Second Affiliated Hospital of Kunming Medical University Kunming P. R. China
| |
Collapse
|
33
|
Samp1 Mislocalization in Emery-Dreifuss Muscular Dystrophy. Cells 2018; 7:cells7100170. [PMID: 30326651 PMCID: PMC6210792 DOI: 10.3390/cells7100170] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Revised: 10/01/2018] [Accepted: 10/11/2018] [Indexed: 12/27/2022] Open
Abstract
LMNA linked-Emery-Dreifuss muscular dystrophy (EDMD2) is a rare disease characterized by muscle weakness, muscle wasting, and cardiomyopathy with conduction defects. The mutated protein lamin A/C binds several nuclear envelope components including the Linker of Nucleoskeleton and Cytoskeleton (LINC) complex and the inner nuclear membrane protein Samp1 (Spindle Associated Membrane Protein 1). Considering that Samp1 is upregulated during muscle cell differentiation and it is involved in nuclear movement, we hypothesized that it could be part of the protein platform formed by LINC proteins and prelamin A at the myotube nuclear envelope and, as previously demonstrated for those proteins, could be affected in EDMD2. Our results show that Samp1 is uniformly distributed at the nuclear periphery of normal human myotubes and committed myoblasts, but its anchorage at the nuclear poles is related to the presence of farnesylated prelamin A and it is disrupted by the loss of prelamin A farnesylation. Moreover, Samp1 is absent from the nuclear poles in EDMD2 myotubes, which shows that LMNA mutations associated with muscular dystrophy, due to reduced prelamin A levels in muscle cell nuclei, impair Samp1 anchorage. Conversely, SUN1 pathogenetic mutations do not alter Samp1 localization in myotubes, which suggests that Samp1 lies upstream of SUN1 in nuclear envelope protein complexes. The hypothesis that Samp1 is part of the protein platform that regulates microtubule nucleation from the myotube nuclear envelope in concert with pericentrin and LINC components warrants future investigation. As a whole, our data identify Samp1 as a new contributor to EDMD2 pathogenesis and our data are relevant to the understanding of nuclear clustering occurring in laminopathic muscle.
Collapse
|
34
|
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: 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: 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
|
35
|
Pałka M, Tomczak A, Grabowska K, Machowska M, Piekarowicz K, Rzepecka D, Rzepecki R. Laminopathies: what can humans learn from fruit flies. Cell Mol Biol Lett 2018; 23:32. [PMID: 30002683 PMCID: PMC6034310 DOI: 10.1186/s11658-018-0093-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Accepted: 06/05/2018] [Indexed: 01/01/2023] Open
Abstract
Lamin proteins are type V intermediate filament proteins (IFs) located inside the cell nucleus. They are evolutionarily conserved and have similar domain organization and properties to cytoplasmic IFs. Lamins provide a skeletal network for chromatin, the nuclear envelope, nuclear pore complexes and the entire nucleus. They are also responsible for proper connections between the karyoskeleton and structural elements in the cytoplasm: actin and the microtubule and cytoplasmic IF networks. Lamins affect transcription and splicing either directly or indirectly. Translocation of active genes into the close proximity of nuclear lamina is thought to result in their transcriptional silencing. Mutations in genes coding for lamins and interacting proteins in humans result in various genetic disorders, called laminopathies. Human genes coding for A-type lamin (LMNA) are the most frequently mutated. The resulting phenotypes include muscle, cardiac, neuronal, lipodystrophic and metabolic pathologies, early aging phenotypes, and combined complex phenotypes. The Drosophila melanogaster genome codes for lamin B-type (lamin Dm), lamin A-type (lamin C), and for LEM-domain proteins, BAF, LINC-complex proteins and all typical nuclear proteins. The fruit fly system is simpler than the vertebrate one since in flies there is only single lamin B-type and single lamin A-type protein, as opposed to the complex system of B- and A-type lamins in Danio, Xenopus and Mus musculus. This offers a unique opportunity to study laminopathies. Applying genetic tools based on Gal4 and in vitro nuclear assembly system to the fruit fly model may successfully advance knowledge of laminopathies. Here, we review studies of the laminopathies in the fly model system.
Collapse
Affiliation(s)
- Marta Pałka
- Laboratory of Nuclear Proteins, Faculty of Biotechnology, University of Wroclaw, Fryderyka Joliot-Curie 14a, 50-383 Wroclaw, Poland
| | - Aleksandra Tomczak
- Laboratory of Nuclear Proteins, Faculty of Biotechnology, University of Wroclaw, Fryderyka Joliot-Curie 14a, 50-383 Wroclaw, Poland
| | - Katarzyna Grabowska
- Laboratory of Nuclear Proteins, Faculty of Biotechnology, University of Wroclaw, Fryderyka Joliot-Curie 14a, 50-383 Wroclaw, Poland
| | - Magdalena Machowska
- Laboratory of Nuclear Proteins, Faculty of Biotechnology, University of Wroclaw, Fryderyka Joliot-Curie 14a, 50-383 Wroclaw, Poland
| | - Katarzyna Piekarowicz
- Laboratory of Nuclear Proteins, Faculty of Biotechnology, University of Wroclaw, Fryderyka Joliot-Curie 14a, 50-383 Wroclaw, Poland
| | - Dorota Rzepecka
- Laboratory of Nuclear Proteins, Faculty of Biotechnology, University of Wroclaw, Fryderyka Joliot-Curie 14a, 50-383 Wroclaw, Poland
| | - Ryszard Rzepecki
- Laboratory of Nuclear Proteins, Faculty of Biotechnology, University of Wroclaw, Fryderyka Joliot-Curie 14a, 50-383 Wroclaw, Poland
| |
Collapse
|
36
|
Aberrant Compartment Formation by HSPB2 Mislocalizes Lamin A and Compromises Nuclear Integrity and Function. Cell Rep 2018; 20:2100-2115. [PMID: 28854361 PMCID: PMC5583511 DOI: 10.1016/j.celrep.2017.08.018] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Revised: 07/10/2017] [Accepted: 08/01/2017] [Indexed: 11/23/2022] Open
Abstract
Small heat shock proteins (HSPBs) contain intrinsically disordered regions (IDRs), but the functions of these IDRs are still unknown. Here, we report that, in mammalian cells, HSPB2 phase separates to form nuclear compartments with liquid-like properties. We show that phase separation requires the disordered C-terminal domain of HSPB2. We further demonstrate that, in differentiating myoblasts, nuclear HSPB2 compartments sequester lamin A. Increasing the nuclear concentration of HSPB2 causes the formation of aberrant nuclear compartments that mislocalize lamin A and chromatin, with detrimental consequences for nuclear function and integrity. Importantly, phase separation of HSPB2 is regulated by HSPB3, but this ability is lost in two identified HSPB3 mutants that are associated with myopathy. Our results suggest that HSPB2 phase separation is involved in reorganizing the nucleoplasm during myoblast differentiation. Furthermore, these findings support the idea that aberrant HSPB2 phase separation, due to HSPB3 loss-of-function mutations, contributes to myopathy. HSPB2 undergoes concentration-dependent liquid-liquid phase separation in cells HSPB2 phase separation requires its intrinsically disordered C-terminal tail Aberrant HSPB2 phase separation mislocalizes lamin A HSPB3, but not two HSPB3 myopathy mutants, inhibits HSPB2 phase separation
Collapse
|
37
|
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: 0.7] [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
|
38
|
Mandibuloacral dysplasia: A premature ageing disease with aspects of physiological ageing. Ageing Res Rev 2018; 42:1-13. [PMID: 29208544 DOI: 10.1016/j.arr.2017.12.001] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Revised: 11/09/2017] [Accepted: 12/01/2017] [Indexed: 01/12/2023]
Abstract
Mandibuloacral dysplasia (MAD) is a rare genetic condition characterized by bone abnormalities including localized osteolysis and generalized osteoporosis, skin pigmentation, lipodystrophic signs and mildly accelerated ageing. The molecular defects associated with MAD are mutations in LMNA or ZMPSTE24 (FACE1) gene, causing type A or type B MAD, respectively. Downstream of LMNA or ZMPSTE24 mutations, the lamin A precursor, prelamin A, is accumulated in cells and affects chromatin dynamics and stress response. A new form of mandibuloacral dysplasia has been recently associated with mutations in POLD1 gene, encoding DNA polymerase delta, a major player in DNA replication. Of note, involvement of prelamin A in chromatin dynamics and recruitment of DNA repair factors has been also determined under physiological conditions, at the border between stress response and cellular senescence. Here, we review current knowledge on MAD clinical and pathogenetic aspects and highlight aspects typical of physiological ageing.
Collapse
|
39
|
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: 1.4] [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]
|
40
|
Boriani G, Biagini E, Ziacchi M, Malavasi VL, Vitolo M, Talarico M, Mauro E, Gorlato G, Lattanzi G. Cardiolaminopathies from bench to bedside: challenges in clinical decision-making with focus on arrhythmia-related outcomes. Nucleus 2018; 9:442-459. [PMID: 30130999 PMCID: PMC6244733 DOI: 10.1080/19491034.2018.1506680] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Revised: 06/16/2018] [Accepted: 07/25/2018] [Indexed: 02/07/2023] Open
Abstract
Lamin A/C gene mutations can be associated with cardiac diseases, usually referred to as 'cardiolaminopathies' characterized by arrhythmic disorders and/or left ventricular or biventricular dysfunction up to an overt picture of heart failure. The phenotypic cardiac manifestations of laminopathies are frequently mixed in complex clinical patterns and specifically may include bradyarrhythmias (sinus node disease or atrioventricular blocks), atrial arrhythmias (atrial fibrillation, atrial flutter, atrial standstill), ventricular tachyarrhythmias and heart failure of variable degrees of severity. Family history, physical examination, laboratory findings (specifically serum creatine phosphokinase values) and ECG findings are often important 'red flags' in diagnosing a 'cardiolaminopathy'. Sudden arrhythmic death, thromboembolic events or stroke and severe heart failure requiring heart transplantation are the most dramatic complications of the evolution of cardiolaminopathies and appropriate risk stratification is clinically needed combined with clinical follow-up. Treatment with cardiac electrical implantable devices is indicated in case of bradyarrhythmias (implant of a device with pacemaker functions), risk of life-threatening ventricular tachyarrhythmias (implant of an ICD) or in case of heart failure with wide QRS interval (implant of a device for cardiac resynchronization). New technologies introduced in the last 5 years can help physicians to reduce device-related complications, thanks to the extension of device longevity and availability of leadless pacemakers or defibrillators, to be implanted in appropriately selected patients. An improved knowledge of the complex pathophysiological pathways involved in cardiolaminopathies and in the determinants of their progression to more severe forms will help to improve clinical management and to better target pharmacological and non-pharmacological treatments.
Collapse
Affiliation(s)
- Giuseppe Boriani
- Cardiology Division, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Policlinico di Modena, Modena, Italy
| | - Elena Biagini
- Institute of Cardiology, Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, S.Orsola-Malpighi University Hospital, Bologna, Italy
| | - Matteo Ziacchi
- Institute of Cardiology, Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, S.Orsola-Malpighi University Hospital, Bologna, Italy
| | - Vincenzo Livio Malavasi
- Cardiology Division, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Policlinico di Modena, Modena, Italy
| | - Marco Vitolo
- Cardiology Division, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Policlinico di Modena, Modena, Italy
| | - Marisa Talarico
- Cardiology Division, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Policlinico di Modena, Modena, Italy
| | - Erminio Mauro
- Cardiology Division, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Policlinico di Modena, Modena, Italy
| | - Giulia Gorlato
- Cardiology Division, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Policlinico di Modena, Modena, Italy
| | - Giovanna Lattanzi
- CNR Institute of Molecular Genetics, Unit of Bologna, Bologna, Italy
- Laboratory of Musculoskeletal Cell Biology, Rizzoli Orthopedic Institute, Bologna, Italy
| |
Collapse
|
41
|
Stroud MJ, Feng W, Zhang J, Veevers J, Fang X, Gerace L, Chen J. Nesprin 1α2 is essential for mouse postnatal viability and nuclear positioning in skeletal muscle. J Cell Biol 2017; 216:1915-1924. [PMID: 28533284 PMCID: PMC5496623 DOI: 10.1083/jcb.201612128] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2017] [Revised: 03/08/2017] [Accepted: 04/18/2017] [Indexed: 12/13/2022] Open
Abstract
The position of the nucleus in a cell is controlled by interactions between the linker of nucleoskeleton and cytoskeleton (LINC) complex and the cytoskeleton. Defects in nuclear positioning and abnormal aggregation of nuclei occur in many muscle diseases and correlate with muscle dysfunction. Nesprin 1, which includes multiple isoforms, is an integral component of the LINC complex, critical for nuclear positioning and anchorage in skeletal muscle, and is thought to provide an essential link between nuclei and actin. However, previous studies have yet to identify which isoform is responsible. To elucidate this, we generated a series of nesprin 1 mutant mice. We showed that the actin-binding domains of nesprin 1 were dispensable, whereas nesprin 1α2, which lacks actin-binding domains, was crucial for postnatal viability, nuclear positioning, and skeletal muscle function. Furthermore, we revealed that kinesin 1 was displaced in fibers of nesprin 1α2-knockout mice, suggesting that this interaction may play an important role in positioning of myonuclei and functional skeletal muscle.
Collapse
Affiliation(s)
- Matthew J Stroud
- Department of Medicine, University of California, San Diego, La Jolla, CA
| | - Wei Feng
- Department of Medicine, University of California, San Diego, La Jolla, CA
| | - Jianlin Zhang
- Department of Medicine, University of California, San Diego, La Jolla, CA
| | - Jennifer Veevers
- Department of Medicine, University of California, San Diego, La Jolla, CA
| | - Xi Fang
- Department of Medicine, University of California, San Diego, La Jolla, CA
| | - Larry Gerace
- Department of Cell and Molecular Biology, The Scripps Research Institute, La Jolla, CA
| | - Ju Chen
- Department of Medicine, University of California, San Diego, La Jolla, CA
| |
Collapse
|
42
|
Hickey R, Pelling AE. The rotation of mouse myoblast nuclei is dependent on substrate elasticity. Cytoskeleton (Hoboken) 2017; 74:184-194. [PMID: 28236372 DOI: 10.1002/cm.21357] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Revised: 02/15/2017] [Accepted: 02/21/2017] [Indexed: 11/09/2022]
Abstract
The complex interplay of biochemical signaling and mechanical traction forces regulate the position of cellular nuclei. Although the phenomenon of nuclear rotation has been observed for many years, the influence of substrate elasticity was unknown. We discovered another layer of complexity to this phenomenon: nuclear rotation is dependent on substrate elasticity. Nuclear rotation is drastically reduced on physiologically relevant stiffnesses. Here, we studied nuclear rotation in mouse C2C12 myoblasts cultured on soft substrates designed to mimic resting tissue (∼26 kPa) and on hard glass substrates. We examined the roles of the actin and microtubule cytoskeleton on the presence and dynamics of nuclear rotation in these two different microenvironments. We demonstrated the clear dependence of nuclear rotation dynamics on matrix stiffness. These results will have important implications for the design of future studies of nuclear rotation and our understanding of the phenomenon as a whole. Unnaturally, hard substrates do not only fail to mimic the in vivo microenvironment, but can also induce cellular processes that would not normally occur in the natural cellular environment.
Collapse
Affiliation(s)
- Ryan Hickey
- Centre for Interdisciplinary NanoPhysics, Department of Physics, University of Ottawa, MacDonald Hall, 150 Louis Pasteur, Ottawa, ON, K1N5N5, Canada
| | - Andrew E Pelling
- Centre for Interdisciplinary NanoPhysics, Department of Physics, University of Ottawa, MacDonald Hall, 150 Louis Pasteur, Ottawa, ON, K1N5N5, Canada.,Department of Biology, University of Ottawa, Gendron Hall, 30 Marie Curie, Ottawa, ON, K1N5N5, Canada.,Institute for Science Society and Policy, Simard Hall, 60 University, University of Ottawa, Ottawa, ON, K1N5N5, Canada.,SymbioticA, School of Anatomy, Physiology and Human Biology, University of Western Australia, Perth, WA, 6009
| |
Collapse
|
43
|
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: 2.6] [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
|
44
|
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: 6.7] [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
|
45
|
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: 2.6] [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
|
46
|
Camozzi D, Capanni C, Cenni V, Mattioli E, Columbaro M, Squarzoni S, Lattanzi G. Diverse lamin-dependent mechanisms interact to control chromatin dynamics. Focus on laminopathies. Nucleus 2015; 5:427-40. [PMID: 25482195 PMCID: PMC4164485 DOI: 10.4161/nucl.36289] [Citation(s) in RCA: 86] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Interconnected functional strategies govern chromatin dynamics in eukaryotic cells. In this context, A and B type lamins, the nuclear intermediate filaments, act on diverse platforms involved in tissue homeostasis. On the nuclear side, lamins elicit large scale or fine chromatin conformational changes, affect DNA damage response factors and transcription factor shuttling. On the cytoplasmic side, bridging-molecules, the LINC complex, associate with lamins to coordinate chromatin dynamics with cytoskeleton and extra-cellular signals.
Consistent with such a fine tuning, lamin mutations and/or defects in their expression or post-translational processing, as well as mutations in lamin partner genes, cause a heterogeneous group of diseases known as laminopathies. They include muscular dystrophies, cardiomyopathy, lipodystrophies, neuropathies, and progeroid syndromes. The study of chromatin dynamics under pathological conditions, which is summarized in this review, is shedding light on the complex and fascinating role of the nuclear lamina in chromatin regulation.
Collapse
Affiliation(s)
- Daria Camozzi
- a CNR Institute for Molecular Genetics; Unit of Bologna and SC Laboratory of Musculoskeletal Cell Biology; Rizzoli Orthopedic Institute; Bologna, Italy
| | | | | | | | | | | | | |
Collapse
|
47
|
Cenni V, Capanni C, Mattioli E, Columbaro M, Wehnert M, Ortolani M, Fini M, Novelli G, Bertacchini J, Maraldi NM, Marmiroli S, D'Apice MR, Prencipe S, Squarzoni S, Lattanzi G. Rapamycin treatment of Mandibuloacral dysplasia cells rescues localization of chromatin-associated proteins and cell cycle dynamics. Aging (Albany NY) 2015; 6:755-70. [PMID: 25324471 PMCID: PMC4233654 DOI: 10.18632/aging.100680] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Lamin A is a key component of the nuclear lamina produced through post-translational processing of its precursor known as prelamin A. LMNA mutations leading to farnesylated prelamin A accumulation are known to cause lipodystrophy, progeroid and developmental diseases, including Mandibuloacral dysplasia, a mild progeroid syndrome with partial lipodystrophy and altered bone turnover. Thus, degradation of prelamin A is expected to improve the disease phenotype. Here, we show different susceptibilities of prelamin A forms to proteolysis and further demonstrate that treatment with rapamycin efficiently and selectively triggers lysosomal degradation of farnesylated prelamin A, the most toxic processing intermediate. Importantly, rapamycin treatment of Mandibuloacral dysplasia cells, which feature very low levels of the NAD-dependent sirtuin SIRT-1 in the nuclear matrix, restores SIRT-1 localization and distribution of chromatin markers, elicits release of the transcription factor Oct-1 and determines shortening of the prolonged S-phase. These findings indicate the drug as a possible treatment for Mandibuloacral dysplasia.
Collapse
Affiliation(s)
- Vittoria Cenni
- National Research Council of Italy, Institute of Molecular Genetics, IGM-CNR-IOR, Bologna, Italy. Rizzoli Orthopedic Institute, Laboratory of Musculoskeletal Cell Biology, Bologna, Italy
| | - Cristina Capanni
- National Research Council of Italy, Institute of Molecular Genetics, IGM-CNR-IOR, Bologna, Italy. Rizzoli Orthopedic Institute, Laboratory of Musculoskeletal Cell Biology, Bologna, Italy
| | - Elisabetta Mattioli
- National Research Council of Italy, Institute of Molecular Genetics, IGM-CNR-IOR, Bologna, Italy. Rizzoli Orthopedic Institute, Laboratory of Musculoskeletal Cell Biology, Bologna, Italy
| | - Marta Columbaro
- Rizzoli Orthopedic Institute, Laboratory of Musculoskeletal Cell Biology, Bologna, Italy
| | - Manfred Wehnert
- Institute of Human Genetics, University of Greifswald, Germany
| | - Michela Ortolani
- National Research Council of Italy, Institute of Molecular Genetics, IGM-CNR-IOR, Bologna, Italy
| | - Milena Fini
- Rizzoli Orthopedic Institute, Laboratory of Preclinical and Surgical Studies and BITTA, RIT, Bologna, Italy
| | - Giuseppe Novelli
- Department of Biomedicine and Prevention, Tor Vergata University, Rome, Italy
| | - Jessika Bertacchini
- Department of Laboratory, CEIA, University of Modena and Reggio Emilia, Modena, Italy
| | - Nadir M Maraldi
- National Research Council of Italy, Institute of Molecular Genetics, IGM-CNR-IOR, Bologna, Italy
| | - Sandra Marmiroli
- Department of Laboratory, CEIA, University of Modena and Reggio Emilia, Modena, Italy
| | - Maria Rosaria D'Apice
- Department of Biomedicine and Prevention, Tor Vergata University, Rome, Italy. Fondazione Policlinico Tor Vergata, Rome, Italy
| | - Sabino Prencipe
- National Research Council of Italy, Institute of Molecular Genetics, IGM-CNR-IOR, Bologna, Italy. Rizzoli Orthopedic Institute, Laboratory of Musculoskeletal Cell Biology, Bologna, Italy
| | - Stefano Squarzoni
- National Research Council of Italy, Institute of Molecular Genetics, IGM-CNR-IOR, Bologna, Italy. Rizzoli Orthopedic Institute, Laboratory of Musculoskeletal Cell Biology, Bologna, Italy
| | - Giovanna Lattanzi
- National Research Council of Italy, Institute of Molecular Genetics, IGM-CNR-IOR, Bologna, Italy. Rizzoli Orthopedic Institute, Laboratory of Musculoskeletal Cell Biology, Bologna, Italy
| |
Collapse
|
48
|
Wilson MH, Holzbaur ELF. Nesprins anchor kinesin-1 motors to the nucleus to drive nuclear distribution in muscle cells. Development 2015; 142:218-28. [PMID: 25516977 DOI: 10.1242/dev.114769] [Citation(s) in RCA: 118] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
During skeletal muscle development, nuclei move dynamically through myotubes in a microtubule-dependent manner, driven by the microtubule motor protein kinesin-1. Loss of kinesin-1 leads to improperly positioned nuclei in culture and in vivo. Two models have been proposed to explain how kinesin-1 functions to move nuclei in myotubes. In the cargo model, kinesin-1 acts directly from the surface of the nucleus, whereas in an alternative model, kinesin-1 moves nuclei indirectly by sliding anti-parallel microtubules. Here, we test the hypothesis that an ensemble of Kif5B motors acts from the nuclear envelope to distribute nuclei throughout the length of syncytial myotubes. First, using an inducible dimerization system, we show that controlled recruitment of truncated, constitutively active kinesin-1 motors to the nuclear envelope is sufficient to prevent the nuclear aggregation resulting from depletion of endogenous kinesin-1. Second, we identify a conserved kinesin light chain (KLC)-binding motif in the nuclear envelope proteins nesprin-1 and nesprin-2, and show that recruitment of the motor complex to the nucleus via this LEWD motif is essential for nuclear distribution. Together, our findings demonstrate that the nucleus is a kinesin-1 cargo in myotubes and that nesprins function as nuclear cargo adaptors. The importance of achieving and maintaining proper nuclear position is not restricted to muscle fibers, suggesting that the nesprin-dependent recruitment of kinesin-1 to the nuclear envelope through the interaction of a conserved LEWD motif with kinesin light chain might be a general mechanism for cell-type-specific nuclear positioning during development.
Collapse
Affiliation(s)
- Meredith H Wilson
- Department of Physiology and the Pennsylvania Muscle Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Erika L F Holzbaur
- Department of Physiology and the Pennsylvania Muscle Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
49
|
Meinke P, Mattioli E, Haque F, Antoku S, Columbaro M, Straatman KR, Worman HJ, Gundersen GG, Lattanzi G, Wehnert M, Shackleton S. Muscular dystrophy-associated SUN1 and SUN2 variants disrupt nuclear-cytoskeletal connections and myonuclear organization. PLoS Genet 2014; 10:e1004605. [PMID: 25210889 PMCID: PMC4161305 DOI: 10.1371/journal.pgen.1004605] [Citation(s) in RCA: 133] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2013] [Accepted: 07/16/2014] [Indexed: 11/19/2022] Open
Abstract
Proteins of the nuclear envelope (NE) are associated with a range of inherited disorders, most commonly involving muscular dystrophy and cardiomyopathy, as exemplified by Emery-Dreifuss muscular dystrophy (EDMD). EDMD is both genetically and phenotypically variable, and some evidence of modifier genes has been reported. Six genes have so far been linked to EDMD, four encoding proteins associated with the LINC complex that connects the nucleus to the cytoskeleton. However, 50% of patients have no identifiable mutations in these genes. Using a candidate approach, we have identified putative disease-causing variants in the SUN1 and SUN2 genes, also encoding LINC complex components, in patients with EDMD and related myopathies. Our data also suggest that SUN1 and SUN2 can act as disease modifier genes in individuals with co-segregating mutations in other EDMD genes. Five SUN1/SUN2 variants examined impaired rearward nuclear repositioning in fibroblasts, confirming defective LINC complex function in nuclear-cytoskeletal coupling. Furthermore, myotubes from a patient carrying compound heterozygous SUN1 mutations displayed gross defects in myonuclear organization. This was accompanied by loss of recruitment of centrosomal marker, pericentrin, to the NE and impaired microtubule nucleation at the NE, events that are required for correct myonuclear arrangement. These defects were recapitulated in C2C12 myotubes expressing exogenous SUN1 variants, demonstrating a direct link between SUN1 mutation and impairment of nuclear-microtubule coupling and myonuclear positioning. Our findings strongly support an important role for SUN1 and SUN2 in muscle disease pathogenesis and support the hypothesis that defects in the LINC complex contribute to disease pathology through disruption of nuclear-microtubule association, resulting in defective myonuclear positioning. Emery-Dreifuss muscular dystrophy (EDMD) is an inherited disorder involving muscle wasting and weakness, accompanied by cardiac defects. The disease is variable in its severity and also in its genetic cause. So far, 6 genes have been linked to EDMD, most encoding proteins that form a structural network that supports the nucleus of the cell and connects it to structural elements of the cytoplasm. This network is particularly important in muscle cells, providing resistance to mechanical strain. Weakening of this network is thought to contribute to development of muscle disease in these patients. Despite rigorous screening, at least 50% of patients with EDMD have no detectable mutation in the 6 known genes. We therefore undertook screening and identified mutations in two additional genes that encode other components of the nuclear structural network, SUN1 and SUN2. Our findings add to the genetic complexity of this disease since some individuals carry mutations in more than one gene. We also show that the mutations disrupt connections between the nucleus and the structural elements of cytoplasm, leading to mis-positioning and clustering of nuclei in muscle cells. This nuclear mis-positioning is likely to be another factor contributing to pathogenesis of EDMD.
Collapse
Affiliation(s)
- Peter Meinke
- Institute of Human Genetics and Interfaculty Institute of Genetics and Functional Genomics, University of Greifswald, Greifswald, Germany
| | - Elisabetta Mattioli
- National Research Council of Italy - CNR - Institute for Molecular Genetics, Unit of Bologna IOR, Bologna, Italy
- Rizzoli Orthopaedic Institute, Laboratory of Musculoskeletal Cell Biology, Bologna, Italy
| | - Farhana Haque
- Department of Biochemistry, University of Leicester, Leicester, United Kingdom
| | - Susumu Antoku
- Department of Pathology and Cell Biology, College of Physicians and Surgeons, Columbia University, New York, New York, United States of America
| | - Marta Columbaro
- Rizzoli Orthopaedic Institute, Laboratory of Musculoskeletal Cell Biology, Bologna, Italy
| | - Kees R. Straatman
- Centre for Core Biotechnology Services, University of Leicester, Leicester, United Kingdom
| | - Howard J. Worman
- Department of Pathology and Cell Biology, College of Physicians and Surgeons, Columbia University, New York, New York, United States of America
- Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, New York, United States of America
| | - Gregg G. Gundersen
- Department of Pathology and Cell Biology, College of Physicians and Surgeons, Columbia University, New York, New York, United States of America
| | - Giovanna Lattanzi
- National Research Council of Italy - CNR - Institute for Molecular Genetics, Unit of Bologna IOR, Bologna, Italy
- Rizzoli Orthopaedic Institute, Laboratory of Musculoskeletal Cell Biology, Bologna, Italy
| | - Manfred Wehnert
- Institute of Human Genetics and Interfaculty Institute of Genetics and Functional Genomics, University of Greifswald, Greifswald, Germany
| | - Sue Shackleton
- Department of Biochemistry, University of Leicester, Leicester, United Kingdom
- * E-mail:
| |
Collapse
|
50
|
Luo YB, Mitrpant C, Adams AM, Johnsen RD, Fletcher S, Mastaglia FL, Wilton SD. Antisense oligonucleotide induction of progerin in human myogenic cells. PLoS One 2014; 9:e98306. [PMID: 24892300 PMCID: PMC4044034 DOI: 10.1371/journal.pone.0098306] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2014] [Accepted: 04/30/2014] [Indexed: 01/16/2023] Open
Abstract
We sought to use splice-switching antisense oligonucleotides to produce a model of accelerated ageing by enhancing expression of progerin, translated from a mis-spliced lamin A gene (LMNA) transcript in human myogenic cells. The progerin transcript (LMNA Δ150) lacks the last 150 bases of exon 11, and is translated into a truncated protein associated with the severe premature ageing disease, Hutchinson-Gilford progeria syndrome (HGPS). HGPS arises from de novo mutations that activate a cryptic splice site in exon 11 of LMNA and result in progerin accumulation in tissues of mesodermal origin. Progerin has also been proposed to play a role in the 'natural' ageing process in tissues. We sought to test this hypothesis by producing a model of accelerated muscle ageing in human myogenic cells. A panel of splice-switching antisense oligonucleotides were designed to anneal across exon 11 of the LMNA pre-mRNA, and these compounds were transfected into primary human myogenic cells. RT-PCR showed that the majority of oligonucleotides were able to modify LMNA transcript processing. Oligonucleotides that annealed within the 150 base region of exon 11 that is missing in the progerin transcript, as well as those that targeted the normal exon 11 donor site induced the LMNA Δ150 transcript, but most oligonucleotides also generated variable levels of LMNA transcript missing the entire exon 11. Upon evaluation of different oligomer chemistries, the morpholino phosphorodiamidate oligonucleotides were found to be more efficient than the equivalent sequences prepared as oligonucleotides with 2'-O-methyl modified bases on a phosphorothioate backbone. The morpholino oligonucleotides induced nuclear localised progerin, demonstrated by immunostaining, and morphological nuclear changes typical of HGPS cells. We show that it is possible to induce progerin expression in myogenic cells using splice-switching oligonucleotides to redirect splicing of LMNA. This may offer a model to investigate the role of progerin in premature muscle ageing.
Collapse
Affiliation(s)
- Yue-Bei Luo
- Centre for Neuromuscular and Neurological Disorders, Australian Neuro-Muscular Research Institute, University of Western Australia, Perth, Australia
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Chalermchai Mitrpant
- Centre for Neuromuscular and Neurological Disorders, Australian Neuro-Muscular Research Institute, University of Western Australia, Perth, Australia
- Department of Biochemistry, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Abbie M. Adams
- Centre for Neuromuscular and Neurological Disorders, Australian Neuro-Muscular Research Institute, University of Western Australia, Perth, Australia
- Centre for Comparative Genomics, Murdoch University, Perth, Australia
| | - Russell D. Johnsen
- Centre for Neuromuscular and Neurological Disorders, Australian Neuro-Muscular Research Institute, University of Western Australia, Perth, Australia
- Centre for Comparative Genomics, Murdoch University, Perth, Australia
| | - Sue Fletcher
- Centre for Neuromuscular and Neurological Disorders, Australian Neuro-Muscular Research Institute, University of Western Australia, Perth, Australia
- Centre for Comparative Genomics, Murdoch University, Perth, Australia
| | - Frank L. Mastaglia
- Centre for Neuromuscular and Neurological Disorders, Australian Neuro-Muscular Research Institute, University of Western Australia, Perth, Australia
- Institute for Immunology & Infectious Diseases, Murdoch University, Perth, Australia
| | - Steve D. Wilton
- Centre for Neuromuscular and Neurological Disorders, Australian Neuro-Muscular Research Institute, University of Western Australia, Perth, Australia
- Centre for Comparative Genomics, Murdoch University, Perth, Australia
| |
Collapse
|