1
|
Ji Z, Siu WYS, Dueñas ME, Müller L, Trost M, Carvalho P. Suppression of TGF-β/SMAD signaling by an inner nuclear membrane phosphatase complex. Nat Commun 2025; 16:3474. [PMID: 40216785 PMCID: PMC11992160 DOI: 10.1038/s41467-025-58681-x] [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: 08/08/2024] [Accepted: 03/28/2025] [Indexed: 04/14/2025] Open
Abstract
Cytokines of the TGF-β superfamily control essential cell fate decisions via receptor regulated SMAD (R-SMAD) transcription factors. Ligand-induced R-SMAD phosphorylation in the cytosol triggers their activation and nuclear accumulation. We determine how R-SMADs are inactivated by dephosphorylation in the cell nucleus to counteract signaling by TGF-β superfamily ligands. We show that R-SMAD dephosphorylation is mediated by an inner nuclear membrane associated complex containing the scaffold protein MAN1 and the CTDNEP1-NEP1R1 phosphatase. Structural prediction, domain mapping and mutagenesis reveals that MAN1 binds independently to the CTDNEP1-NEP1R1 phosphatase and R-SMADs to promote their inactivation by dephosphorylation. Disruption of this complex causes nuclear accumulation of R-SMADs and aberrant signaling, even in the absence of TGF-β ligands. These findings establish CTDNEP1-NEP1R1 as the R-SMAD phosphatase, reveal the mechanistic basis for TGF-β signaling inactivation and highlight how this process is disrupted by disease-associated MAN1 mutations.
Collapse
Affiliation(s)
- Zhe Ji
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| | | | - Maria Emilia Dueñas
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK
- The Kids Research Institute Australia, Perth Children's Hospital, Nedlands, Australia
| | - Leonie Müller
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Matthias Trost
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Pedro Carvalho
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK.
| |
Collapse
|
2
|
El Mossadeq L, Bellutti L, Le Borgne R, Canman JC, Pintard L, Verbavatz JM, Askjaer P, Dumont J. An interkinetic envelope surrounds chromosomes between meiosis I and II in C. elegans oocytes. J Cell Biol 2025; 224:e202403125. [PMID: 39724138 DOI: 10.1083/jcb.202403125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 09/24/2024] [Accepted: 11/18/2024] [Indexed: 12/28/2024] Open
Abstract
At the end of cell division, the nuclear envelope reassembles around the decondensing chromosomes. Female meiosis culminates in two consecutive cell divisions of the oocyte, meiosis I and II, which are separated by a brief transition phase known as interkinesis. Due to the absence of chromosome decondensation and the suppression of genome replication during interkinesis, it has been widely assumed that the nuclear envelope does not reassemble between meiosis I and II. By analyzing interkinesis in C. elegans oocytes, we instead show that an atypical structure made of two lipid bilayers, which we termed the interkinetic envelope, surrounds the surface of the segregating chromosomes. The interkinetic envelope shares common features with the nuclear envelope but also exhibits specific characteristics that distinguish it, including its lack of continuity with the endoplasmic reticulum, unique protein composition, assembly mechanism, and function in chromosome segregation. These distinct attributes collectively define the interkinetic envelope as a unique and specialized structure that has been previously overlooked.
Collapse
Affiliation(s)
| | - Laura Bellutti
- Université Paris Cité, CNRS, Institut Jacques Monod , Paris, France
| | - Rémi Le Borgne
- Université Paris Cité, CNRS, Institut Jacques Monod , Paris, France
| | - Julie C Canman
- Department of Pathology and Cell Biology, Columbia University, New York, NY, USA
| | - Lionel Pintard
- Université Paris Cité, CNRS, Institut Jacques Monod , Paris, France
| | | | - Peter Askjaer
- Andalusian Center for Developmental Biology, CSIC/JA/Universidad Pablo de Olavide , Seville, Spain
| | - Julien Dumont
- Université Paris Cité, CNRS, Institut Jacques Monod , Paris, France
| |
Collapse
|
3
|
Wang Y, Chen Z, Yang G, Yuan G. Unveiling the roles of LEMD proteins in cellular processes. Life Sci 2024; 357:123116. [PMID: 39374771 DOI: 10.1016/j.lfs.2024.123116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 09/30/2024] [Accepted: 10/03/2024] [Indexed: 10/09/2024]
Abstract
Proteins localized in the inner nuclear membrane (INM) engage in various fundamental cellular processes via their interactions with outer nuclear membrane (ONM) proteins and nuclear lamina. LAP2-emerin-MAN1 domain (LEMD) family proteins, predominantly positioned in the INM, participate in the maintenance of INM functions, including the reconstruction of the nuclear envelope during mitosis, mechanotransduction, and gene transcriptional modulation. Malfunction of LEMD proteins leads to severe tissue-restricted diseases, which may manifest as fatal deformities and defects. In this review, we summarize the significant roles of LEMD proteins in cellular processes, explains the mechanisms of LEMD protein-related diseases, and puts forward questions in less-explored areas like details in tissue-restricted phenotypes. It intends to sort out previous works about LEMD proteins and pave way for future researchers who might discover deeper mechanisms of and better treatment strategies for LEMD protein-related diseases.
Collapse
Affiliation(s)
- Yiyun Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei 430079, China; Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan 430071, China
| | - Zhi Chen
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei 430079, China; Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan 430071, China
| | - Guobin Yang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei 430079, China; Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan 430071, China
| | - Guohua Yuan
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei 430079, China; Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan 430071, China.
| |
Collapse
|
4
|
Yang D, Lai A, Davies A, Janssen AF, Ellis MO, Larrieu D. A novel role for CSA in the regulation of nuclear envelope integrity: uncovering a non-canonical function. Life Sci Alliance 2024; 7:e202402745. [PMID: 39209536 PMCID: PMC11361374 DOI: 10.26508/lsa.202402745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 08/14/2024] [Accepted: 08/20/2024] [Indexed: 09/04/2024] Open
Abstract
Cockayne syndrome (CS) is a premature ageing condition characterized by microcephaly, growth failure, and neurodegeneration. It is caused by mutations in ERCC6 or ERCC8 encoding for Cockayne syndrome B (CSB) and A (CSA) proteins, respectively. CSA and CSB have well-characterized roles in transcription-coupled nucleotide excision repair, responsible for removing bulky DNA lesions, including those caused by UV irradiation. Here, we report that CSA dysfunction causes defects in the nuclear envelope (NE) integrity. NE dysfunction is characteristic of progeroid disorders caused by a mutation in NE proteins, such as Hutchinson-Gilford progeria syndrome. However, it has never been reported in Cockayne syndrome. We observed CSA dysfunction affected LEMD2 incorporation at the NE and increased actin stress fibers that contributed to enhanced mechanical stress to the NE. Altogether, these led to NE abnormalities associated with the activation of the cGAS/STING pathway. Targeting the linker of the nucleoskeleton and cytoskeleton complex was sufficient to rescue these phenotypes. This work reveals NE dysfunction in a progeroid syndrome caused by mutations in a DNA damage repair protein, reinforcing the connection between NE deregulation and ageing.
Collapse
Affiliation(s)
- Denny Yang
- Department of Pharmacology, University of Cambridge, Cambridge, UK
- UK Dementia Research Institute, Island Research Building, Cambridge, UK
| | - Austin Lai
- Cambridge Institute for Medical Research, The Keith Peters Building, Cambridge, UK
| | - Amelie Davies
- Department of Pharmacology, University of Cambridge, Cambridge, UK
| | - Anne Fj Janssen
- Department of Pharmacology, University of Cambridge, Cambridge, UK
- Cambridge Institute for Medical Research, The Keith Peters Building, Cambridge, UK
| | - Matthew O Ellis
- UK Dementia Research Institute, Island Research Building, Cambridge, UK
| | - Delphine Larrieu
- Department of Pharmacology, University of Cambridge, Cambridge, UK
- Cambridge Institute for Medical Research, The Keith Peters Building, Cambridge, UK
| |
Collapse
|
5
|
Mossadeq LE, Bellutti L, Borgne RL, Canman JC, Pintard L, Verbavatz JM, Askjaer P, Dumont J. An interkinetic envelope surrounds chromosomes between meiosis I and II in C. elegans oocytes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.19.619195. [PMID: 39484525 PMCID: PMC11526925 DOI: 10.1101/2024.10.19.619195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
At the end of cell division, the nuclear envelope reassembles around the decondensing chromosomes. Female meiosis culminates in two consecutive cell divisions of the oocyte, meiosis I and II, which are separated by a brief transition phase known as interkinesis. Due to the absence of chromosome decondensation and the suppression of genome replication during interkinesis, it has been widely assumed that the nuclear envelope does not reassemble between meiosis I and II. By analyzing interkinesis in C. elegans oocytes, we instead show that an atypical structure made of two lipid bilayers, which we termed the interkinetic envelope, surrounds the surface of the segregating chromosomes. The interkinetic envelope shares common features with the nuclear envelope but also exhibits specific characteristics that distinguish it, including its lack of continuity with the endoplasmic reticulum, unique protein composition, assembly mechanism, and function in chromosome segregation. These distinct attributes collectively define the interkinetic envelope as a unique and specialized structure that has been previously overlooked.
Collapse
Affiliation(s)
- Layla El Mossadeq
- Université Paris Cité, CNRS, Institut Jacques Monod, F-75013 Paris, France
| | - Laura Bellutti
- Université Paris Cité, CNRS, Institut Jacques Monod, F-75013 Paris, France
| | - Rémi Le Borgne
- Université Paris Cité, CNRS, Institut Jacques Monod, F-75013 Paris, France
| | - Julie C. Canman
- Columbia University; Department of Pathology and Cell Biology, New York, NY 10032, USA
| | - Lionel Pintard
- Université Paris Cité, CNRS, Institut Jacques Monod, F-75013 Paris, France
| | | | - Peter Askjaer
- Andalusian Center for Developmental Biology (CABD), CSIC/JA/Universidad Pablo de Olavide, Seville, Spain
| | - Julien Dumont
- Université Paris Cité, CNRS, Institut Jacques Monod, F-75013 Paris, France
| |
Collapse
|
6
|
Matter A, Kaufman C, Zürcher N, Lenggenhager D, Grehten P, Bartholdi D, Horka L, Häberle J, Makris G. LEMD2-associated progeroid syndrome: Expanding the phenotype of the nuclear envelopathy caused by a defect in LEMD2 gene. Aging Cell 2024; 23:e14189. [PMID: 38757373 PMCID: PMC11320348 DOI: 10.1111/acel.14189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 03/27/2024] [Accepted: 04/19/2024] [Indexed: 05/18/2024] Open
Abstract
Nuclear envelopathies are rare genetic diseases that compromise the integrity of the nuclear envelope. Patients with a defect in LEM domain nuclear envelope protein 2 (LEMD2) leading to LEMD2-associated progeroid syndrome are exceedingly scarce in number, yet they exhibit shared clinical features including skeletal abnormalities and a prematurely-aged appearance. Our study broadens the understanding of LEMD2-associated progeroid syndrome by detailing its phenotypic and molecular characteristics in the first female and fourth reported case, highlighting a distinct impact on metabolic functions. The patient's history revealed growth delay, facial and skeletal abnormalities, and recurrent abdominal pain crises caused by hepatomegaly. Comparisons with the previously documented cases emphasized similarities in skeletal and facial features while showcasing unique variations, notably in cardiac and hepatic manifestations. In vitro experiments conducted on patient-derived peripheral blood and urinary epithelial cells and LEMD2-downregulated HepG2 cells confirmed abnormalities in the structure of the nuclear envelope in all three tissue-types. Overall, our work offers a comprehensive profile of a patient with LEMD2-related syndrome, emphasizing the hepatic involvement in the disease and broadening our understanding of clinical and molecular implications. This study not only contributes specific insights into LEMD2-related conditions but also underscores potential therapeutic paths for disorders affecting nuclear envelope dynamics.
Collapse
Affiliation(s)
- Alyssia Matter
- Division of Metabolism and Children's Research CenterUniversity Children's Hospital ZurichZurichSwitzerland
| | - Christina Kaufman
- Division of Metabolism and Children's Research CenterUniversity Children's Hospital ZurichZurichSwitzerland
| | - Nadia Zürcher
- Division of Metabolism and Children's Research CenterUniversity Children's Hospital ZurichZurichSwitzerland
| | - Daniela Lenggenhager
- Department of Pathology and Molecular PathologyUniversity Hospital ZurichZurichSwitzerland
| | - Patrice Grehten
- Department of Diagnostic ImagingUniversity Children's Hospital ZurichZurichSwitzerland
| | - Deborah Bartholdi
- Department of Human Genetics, InselspitalBern University HospitalBernSwitzerland
| | - Laura Horka
- Department of Endocrinology, Diabetology and Clinical NutritionUniversity Hospital ZurichZurichSwitzerland
| | - Johannes Häberle
- Division of Metabolism and Children's Research CenterUniversity Children's Hospital ZurichZurichSwitzerland
| | - Georgios Makris
- Division of Metabolism and Children's Research CenterUniversity Children's Hospital ZurichZurichSwitzerland
| |
Collapse
|
7
|
Keuper K, Bartek J, Maya-Mendoza A. The nexus of nuclear envelope dynamics, circular economy and cancer cell pathophysiology. Eur J Cell Biol 2024; 103:151394. [PMID: 38340500 DOI: 10.1016/j.ejcb.2024.151394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Revised: 02/01/2024] [Accepted: 02/02/2024] [Indexed: 02/12/2024] Open
Abstract
The nuclear envelope (NE) is a critical component in maintaining the function and structure of the eukaryotic nucleus. The NE and lamina are disassembled during each cell cycle to enable an open mitosis. Nuclear architecture construction and deconstruction is a prime example of a circular economy, as it fulfills a highly efficient recycling program bound to continuous assessment of the quality and functionality of the building blocks. Alterations in the nuclear dynamics and lamina structure have emerged as important contributors to both oncogenic transformation and cancer progression. However, the knowledge of the NE breakdown and reassembly is still limited to a fraction of participating proteins and complexes. As cancer cells contain highly diverse nuclei in terms of DNA content, but also in terms of nuclear number, size, and shape, it is of great interest to understand the intricate relationship between these nuclear features in cancer cell pathophysiology. In this review, we provide insights into how those NE dynamics are regulated, and how lamina destabilization processes may alter the NE circular economy. Moreover, we expand the knowledge of the lamina-associated domain region by using strategic algorithms, including Artificial Intelligence, to infer protein associations, assess their function and location, and predict cancer-type specificity with implications for the future of cancer diagnosis, prognosis and treatment. Using this approach we identified NUP98 and MECP2 as potential proteins that exhibit upregulation in Acute Myeloid Leukemia (LAML) patients with implications for early diagnosis.
Collapse
Affiliation(s)
- Kristina Keuper
- DNA Replication and Cancer Group, Danish Cancer Institute, Copenhagen, Denmark; Genome Integrity Group, Danish Cancer Institute, Copenhagen, Denmark
| | - Jiri Bartek
- Genome Integrity Group, Danish Cancer Institute, Copenhagen, Denmark; Division of Genome Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, SciLifeLab, Stockholm, Sweden
| | | |
Collapse
|
8
|
Rose M, Burgess JT, Cheong CM, Adams MN, Shahrouzi P, O’Byrne KJ, Richard DJ, Bolderson E. The expression and role of the Lem-D proteins Ankle2, Emerin, Lemd2, and TMPO in triple-negative breast cancer cell growth. Front Oncol 2024; 14:1222698. [PMID: 38720803 PMCID: PMC11076778 DOI: 10.3389/fonc.2024.1222698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 02/28/2024] [Indexed: 05/12/2024] Open
Abstract
Background Triple-negative breast cancer (TNBC) is a sub-classification of breast carcinomas, which leads to poor survival outcomes for patients. TNBCs do not possess the hormone receptors that are frequently targeted as a therapeutic in other cancer subtypes and, therefore, chemotherapy remains the standard treatment for TNBC. Nuclear envelope proteins are frequently dysregulated in cancer cells, supporting their potential as novel cancer therapy targets. The Lem-domain (Lem-D) (LAP2, Emerin, MAN1 domain, and Lem-D) proteins are a family of inner nuclear membrane proteins, which share a ~45-residue Lem-D. The Lem-D proteins, including Ankle2, Lemd2, TMPO, and Emerin, have been shown to be associated with many of the hallmarks of cancer. This study aimed to define the association between the Lem-D proteins and TNBC and determine whether these proteins could be promising therapeutic targets. Methods GENT2, TCGA, and KM plotter were utilized to investigate the expression and prognostic implications of several Lem-D proteins: Ankle2, TMPO, Emerin, and Lemd2 in publicly available breast cancer patient data. Immunoblotting and immunofluorescent analysis of immortalized non-cancerous breast cells and a panel of TNBC cells were utilized to establish whether protein expression of the Lem-D proteins was significantly altered in TNBC. SiRNA was used to decrease individual Lem-D protein expression, and functional assays, including proliferation assays and apoptosis assays, were conducted. Results The Lem-D proteins were generally overexpressed in TNBC patient samples at the mRNA level and showed variable expression at the protein level in TNBC cell lysates. Similarly, protein levels were generally negatively correlated with patient survival outcomes. siRNA-mediated depletion of the individual Lem-D proteins in TNBC cells induced aberrant nuclear morphology, decreased proliferation, and induced cell death. However, minimal effects on nuclear morphology or cell viability were observed following Lem-D depletion in non-cancerous MCF10A cells. Conclusion There is evidence to suggest that Ankle2, TMPO, Emerin, and Lemd2 expressions are correlated with breast cancer patient outcomes, but larger patient sample numbers are required to confirm this. siRNA-mediated depletion of these proteins was shown to specifically impair TNBC cell growth, suggesting that the Lem-D proteins may be a specific anti-cancer target.
Collapse
Affiliation(s)
- Maddison Rose
- Cancer and Ageing Research Program, Centre for Genomics and Personalised Health, School of Biomedical Sciences, Translational Research Institute, Queensland University of Technology, Brisbane, QLD, Australia
| | - Joshua T. Burgess
- Cancer and Ageing Research Program, Centre for Genomics and Personalised Health, School of Biomedical Sciences, Translational Research Institute, Queensland University of Technology, Brisbane, QLD, Australia
| | - Chee Man Cheong
- Cancer and Ageing Research Program, Centre for Genomics and Personalised Health, School of Biomedical Sciences, Translational Research Institute, Queensland University of Technology, Brisbane, QLD, Australia
| | - Mark N. Adams
- Cancer and Ageing Research Program, Centre for Genomics and Personalised Health, School of Biomedical Sciences, Translational Research Institute, Queensland University of Technology, Brisbane, QLD, Australia
| | - Parastoo Shahrouzi
- Department of Medical Genetics, Faculty of Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Kenneth J. O’Byrne
- Cancer and Ageing Research Program, Centre for Genomics and Personalised Health, School of Biomedical Sciences, Translational Research Institute, Queensland University of Technology, Brisbane, QLD, Australia
- Cancer Services, Princess Alexandra Hospital, Brisbane, QLD, Australia
| | - Derek J. Richard
- Cancer and Ageing Research Program, Centre for Genomics and Personalised Health, School of Biomedical Sciences, Translational Research Institute, Queensland University of Technology, Brisbane, QLD, Australia
| | - Emma Bolderson
- Cancer and Ageing Research Program, Centre for Genomics and Personalised Health, School of Biomedical Sciences, Translational Research Institute, Queensland University of Technology, Brisbane, QLD, Australia
| |
Collapse
|
9
|
Lu Z, Zhang W, Mao X, Li D, Chen X, Liu L, Lin Y. The third case of Marbach-Rustad progeroid syndrome caused by a de novo LEMD2 variant. Clin Genet 2024; 105:209-213. [PMID: 37867468 DOI: 10.1111/cge.14441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 10/05/2023] [Accepted: 10/08/2023] [Indexed: 10/24/2023]
Abstract
Marbach-Rustad progeroid syndrome is an extremely rare disease caused by a heterozygous variant in the LEMD2 gene. To date, only two patients and one LEMD2 pathogenic variant have been reported in Marbach-Rustad progeroid syndrome. Here we describe the third case of Marbach-Rustad progeroid syndrome worldwide, which is also the first case in China. The proband was affected with premature birth, failed to thrive, facial abnormalities, feeding difficulties, skull defects and delayed motor milestones, but had a normal intelligence and speech. Whole exome sequencing (WES) initially did not find a phenotype-causing variant when the proband was 1 year of age. The reanalysis of WES data 4 years later revealed the proband harbored a de novo heterozygous c.1436C>T(p.Ser479Phe) variant in the LEMD2 gene, which is known responsible for Marbach-Rustad progeroid syndrome. Sanger sequencing confirmed the presence of this variant in the proband and absence in his parents and two elder sisters. Our study provides accurate clinical diagnosis for the proband and adds a new patient with Marbach-Rustad progeroid syndrome. Our study suggests the LEMD2 c.1436C>T(p.Ser479Phe) variant as a hotspot. Our work also indicates reanalysis of WES data of negative cases might identify pathogenic variant and improve diagnostic efficiency.
Collapse
Affiliation(s)
- Zhikun Lu
- Department of Genetics and Endocrinology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, China
| | - Wen Zhang
- Department of Genetics and Endocrinology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, China
| | - Xiaojian Mao
- Department of Genetics and Endocrinology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, China
| | - Duan Li
- Department of Genetics and Endocrinology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, China
| | - Xiaodan Chen
- Department of Genetics and Endocrinology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, China
| | - Li Liu
- Department of Genetics and Endocrinology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, China
| | - Yunting Lin
- Department of Genetics and Endocrinology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, China
| |
Collapse
|
10
|
Lee GE, Byun J, Lee CJ, Cho YY. Molecular Mechanisms for the Regulation of Nuclear Membrane Integrity. Int J Mol Sci 2023; 24:15497. [PMID: 37895175 PMCID: PMC10607757 DOI: 10.3390/ijms242015497] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 10/19/2023] [Accepted: 10/22/2023] [Indexed: 10/29/2023] Open
Abstract
The nuclear membrane serves a critical role in protecting the contents of the nucleus and facilitating material and signal exchange between the nucleus and cytoplasm. While extensive research has been dedicated to topics such as nuclear membrane assembly and disassembly during cell division, as well as interactions between nuclear transmembrane proteins and both nucleoskeletal and cytoskeletal components, there has been comparatively less emphasis on exploring the regulation of nuclear morphology through nuclear membrane integrity. In particular, the role of type II integral proteins, which also function as transcription factors, within the nuclear membrane remains an area of research that is yet to be fully explored. The integrity of the nuclear membrane is pivotal not only during cell division but also in the regulation of gene expression and the communication between the nucleus and cytoplasm. Importantly, it plays a significant role in the development of various diseases. This review paper seeks to illuminate the biomolecules responsible for maintaining the integrity of the nuclear membrane. It will delve into the mechanisms that influence nuclear membrane integrity and provide insights into the role of type II membrane protein transcription factors in this context. Understanding these aspects is of utmost importance, as it can offer valuable insights into the intricate processes governing nuclear membrane integrity. Such insights have broad-reaching implications for cellular function and our understanding of disease pathogenesis.
Collapse
Affiliation(s)
- Ga-Eun Lee
- BK21-4th, and BRL, College of Pharmacy, The Catholic University of Korea, 43, Jibong-ro, Wonmi-gu, Bucheon-si 14662, Gyeonggi-do, Republic of Korea; (G.-E.L.); (J.B.)
| | - Jiin Byun
- BK21-4th, and BRL, College of Pharmacy, The Catholic University of Korea, 43, Jibong-ro, Wonmi-gu, Bucheon-si 14662, Gyeonggi-do, Republic of Korea; (G.-E.L.); (J.B.)
| | - Cheol-Jung Lee
- Research Center for Materials Analysis, Korea Basic Science Institute, 169-148, Gwahak-ro, Yuseong-gu, Daejeon 34133, Chungcheongnam-do, Republic of Korea
| | - Yong-Yeon Cho
- BK21-4th, and BRL, College of Pharmacy, The Catholic University of Korea, 43, Jibong-ro, Wonmi-gu, Bucheon-si 14662, Gyeonggi-do, Republic of Korea; (G.-E.L.); (J.B.)
- RCD Control and Material Research Institute, The Catholic University of Korea, 43, Jibong-ro, Wonmi-gu, Bucheon-si 14662, Gyeonggi-do, Republic of Korea
| |
Collapse
|
11
|
Liu C, Rex R, Lung Z, Wang JS, Wu F, Kim HJ, Zhang L, Sohn LL, Dernburg AF. A cooperative network at the nuclear envelope counteracts LINC-mediated forces during oogenesis in C. elegans. SCIENCE ADVANCES 2023; 9:eabn5709. [PMID: 37436986 PMCID: PMC10337908 DOI: 10.1126/sciadv.abn5709] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Accepted: 06/08/2023] [Indexed: 07/14/2023]
Abstract
Oogenesis involves transduction of mechanical forces from the cytoskeleton to the nuclear envelope (NE). In Caenorhabditis elegans, oocyte nuclei lacking the single lamin protein LMN-1 are vulnerable to collapse under forces mediated through LINC (linker of nucleoskeleton and cytoskeleton) complexes. Here, we use cytological analysis and in vivo imaging to investigate the balance of forces that drive this collapse and protect oocyte nuclei. We also use a mechano-node-pore sensing device to directly measure the effect of genetic mutations on oocyte nuclear stiffness. We find that nuclear collapse is not a consequence of apoptosis. It is promoted by dynein, which induces polarization of a LINC complex composed of Sad1 and UNC-84 homology 1 (SUN-1) and ZYGote defective 12 (ZYG-12). Lamins contribute to oocyte nuclear stiffness and cooperate with other inner nuclear membrane proteins to distribute LINC complexes and protect nuclei from collapse. We speculate that a similar network may protect oocyte integrity during extended oocyte arrest in mammals.
Collapse
Affiliation(s)
- Chenshu Liu
- California Institute for Quantitative Biosciences (QB3) and Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, CA 94720, USA
- Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
| | - Rachel Rex
- Department of Mechanical Engineering, University of California Berkeley, Berkeley, CA 94720, USA
| | - Zoe Lung
- Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
| | - John S. Wang
- Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
| | - Fan Wu
- Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
| | - Hyung Jun Kim
- California Institute for Quantitative Biosciences (QB3) and Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, CA 94720, USA
| | - Liangyu Zhang
- California Institute for Quantitative Biosciences (QB3) and Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, CA 94720, USA
- Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
| | - Lydia L. Sohn
- Department of Mechanical Engineering, University of California Berkeley, Berkeley, CA 94720, USA
| | - Abby F. Dernburg
- California Institute for Quantitative Biosciences (QB3) and Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, CA 94720, USA
- Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
- Department of Biological Sciences and Engineering, Life Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| |
Collapse
|
12
|
Torres DE, Reckard AT, Klocko AD, Seidl MF. Nuclear genome organization in fungi: from gene folding to Rabl chromosomes. FEMS Microbiol Rev 2023; 47:fuad021. [PMID: 37197899 PMCID: PMC10246852 DOI: 10.1093/femsre/fuad021] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 04/28/2023] [Accepted: 05/08/2023] [Indexed: 05/19/2023] Open
Abstract
Comparative genomics has recently provided unprecedented insights into the biology and evolution of the fungal lineage. In the postgenomics era, a major research interest focuses now on detailing the functions of fungal genomes, i.e. how genomic information manifests into complex phenotypes. Emerging evidence across diverse eukaryotes has revealed that the organization of DNA within the nucleus is critically important. Here, we discuss the current knowledge on the fungal genome organization, from the association of chromosomes within the nucleus to topological structures at individual genes and the genetic factors required for this hierarchical organization. Chromosome conformation capture followed by high-throughput sequencing (Hi-C) has elucidated how fungal genomes are globally organized in Rabl configuration, in which centromere or telomere bundles are associated with opposite faces of the nuclear envelope. Further, fungal genomes are regionally organized into topologically associated domain-like (TAD-like) chromatin structures. We discuss how chromatin organization impacts the proper function of DNA-templated processes across the fungal genome. Nevertheless, this view is limited to a few fungal taxa given the paucity of fungal Hi-C experiments. We advocate for exploring genome organization across diverse fungal lineages to ensure the future understanding of the impact of nuclear organization on fungal genome function.
Collapse
Affiliation(s)
- David E Torres
- Theoretical Biology and Bioinformatics, Department of Biology, Utrecht University, Padualaan 8, 3584 CH Utrecht, The Netherlands
- Laboratory of Phytopathology, Wageningen University and Research,Droevendaalsesteeg 4, 6708 PB Wageningen, The Netherlands
| | - Andrew T Reckard
- Department of Chemistry and Biochemistry, University of Colorado Colorado Springs, 234 Centennial Hall, 1420 Austin Bluffs Pkwy, Colorado Springs, CO 80918 USA
| | - Andrew D Klocko
- Department of Chemistry and Biochemistry, University of Colorado Colorado Springs, 234 Centennial Hall, 1420 Austin Bluffs Pkwy, Colorado Springs, CO 80918 USA
| | - Michael F Seidl
- Theoretical Biology and Bioinformatics, Department of Biology, Utrecht University, Padualaan 8, 3584 CH Utrecht, The Netherlands
| |
Collapse
|
13
|
Fan Q, Li XM, Zhai C, Li B, Li ST, Dong MQ. Somatic nuclear blebbing in Caenorhabditis elegans is not a feature of organismal aging but a potential indicator of germline proliferation in early adulthood. G3 (BETHESDA, MD.) 2023; 13:jkad029. [PMID: 36735812 PMCID: PMC10085788 DOI: 10.1093/g3journal/jkad029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 01/26/2023] [Accepted: 01/27/2023] [Indexed: 02/05/2023]
Abstract
Abnormal nuclear morphology is suggested to be a hallmark of aging and one such abnormality is nuclear blebbing. However, little is known about whether and how nuclear blebbing participates in animal aging, and what regulates it. In this study, we show that the frequency of nuclear blebbing in the hypodermis increases during aging in wild-type C. elegans. These nuclear blebs are enveloped by the nuclear lamina, the inner and the outer nuclear membrane, and 42% of them contain chromatin. Although nuclear blebbing could lead to DNA loss if chromatin-containing blebs detach and fuse with lysosomes, we find by time-lapse imaging that nuclear blebs rarely detach, and the estimated lifetime of a nuclear bleb is 772 h or 32 days. The amount of DNA lost through nuclear blebbing is estimated to be about 0.1% of the total DNA loss by adult Day 11. Furthermore, the frequency of nuclear blebbing does not correlate with the rate of aging in C. elegans. Old age does not necessarily induce nuclear blebbing, neither does starvation, heat stress, or oxidative stress. Intriguingly, we find that proliferation of germ cells promotes nuclear blebbing.
Collapse
Affiliation(s)
- Qiang Fan
- Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
- National Institute of Biological Sciences (NIBS), Beijing 102206, China
- Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing 102206, China
| | - Xue-Mei Li
- National Institute of Biological Sciences (NIBS), Beijing 102206, China
- Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing 102206, China
| | - Chao Zhai
- National Institute of Biological Sciences (NIBS), Beijing 102206, China
- Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing 102206, China
| | - Bin Li
- National Institute of Biological Sciences (NIBS), Beijing 102206, China
- Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing 102206, China
| | - Shang-Tong Li
- National Institute of Biological Sciences (NIBS), Beijing 102206, China
- Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing 102206, China
| | - Meng-Qiu Dong
- Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
- National Institute of Biological Sciences (NIBS), Beijing 102206, China
- Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing 102206, China
| |
Collapse
|
14
|
Vadrot N, Ader F, Moulin M, Merlant M, Chapon F, Gandjbakhch E, Labombarda F, Maragnes P, Réant P, Rooryck C, Probst V, Donal E, Richard P, Ferreiro A, Buendia B. Abnormal Cellular Phenotypes Induced by Three TMPO/LAP2 Variants Identified in Men with Cardiomyopathies. Cells 2023; 12:337. [PMID: 36672271 PMCID: PMC9857342 DOI: 10.3390/cells12020337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 01/09/2023] [Accepted: 01/11/2023] [Indexed: 01/18/2023] Open
Abstract
A single missense variant of the TMPO/LAP2α gene, encoding LAP2 proteins, has been associated with cardiomyopathy in two brothers. To further evaluate its role in cardiac muscle, we included TMPO in our cardiomyopathy diagnostic gene panel. A screening of ~5000 patients revealed three novel rare TMPO heterozygous variants in six males diagnosed with hypertrophic or dilated cardiomypathy. We identified in different cellular models that (1) the frameshift variant LAP2α p.(Gly395Glufs*11) induced haploinsufficiency, impeding cell proliferation and/or producing a truncated protein mislocalized in the cytoplasm; (2) the C-ter missense variant LAP2α p.(Ala240Thr) led to a reduced proximity events between LAP2α and the nucleosome binding protein HMGN5; and (3) the LEM-domain missense variant p.(Leu124Phe) decreased both associations of LAP2α/β with the chromatin-associated protein BAF and inhibition of the E2F1 transcription factor activity which is known to be dependent on Rb, partner of LAP2α. Additionally, the LAP2α expression was lower in the left ventricles of male mice compared to females. In conclusion, our study reveals distinct altered properties of LAP2 induced by these TMPO/LAP2 variants, leading to altered cell proliferation, chromatin structure or gene expression-regulation pathways, and suggests a potential sex-dependent role of LAP2 in myocardial function and disease.
Collapse
Affiliation(s)
- Nathalie Vadrot
- Basic and Translational Myology Laboratory, Université Paris Cité, BFA, UMR 8251, CNRS, F-75013 Paris, France
| | - Flavie Ader
- APHP—Sorbonne Université, Unité Fonctionnelle de Cardiogénétique et Myogénétique Moléculaire, Service de Biochimie Métabolique, HU Pitié Salpêtrière—Charles Foix, F-75013 Paris, France
- INSERM, UMR_S 1166, Sorbonne Université, F-75005 Paris, France
- Faculté de Pharmacie Paris Descartes, Département 3, Université Paris Cité, F-75006 Paris, France
| | - Maryline Moulin
- Basic and Translational Myology Laboratory, Université Paris Cité, BFA, UMR 8251, CNRS, F-75013 Paris, France
| | - Marie Merlant
- Basic and Translational Myology Laboratory, Université Paris Cité, BFA, UMR 8251, CNRS, F-75013 Paris, France
| | | | - Estelle Gandjbakhch
- INSERM, UMR_S 1166, Sorbonne Université, F-75005 Paris, France
- Département de cardiologie, APHP—Sorbonne Université, HU Pitié Salpêtrière- Charles Foix, F-75610 Paris, France
| | - Fabien Labombarda
- Service de Cardiologie, CHU de Caen, Université de Caen Normandie, F-14000 Caen, France
| | - Pascale Maragnes
- Cardiologie pédiatrique, Service de pédiatrie, CHU de Caen, F-14000 Caen, France
| | - Patricia Réant
- Service de Cardiologie, Hôpital Haut Lévêque, CHU de Bordeaux, INSERM 1045, Université de Bordeaux, F-33000 Bordeaux, France
| | - Caroline Rooryck
- Service de Génétique Médicale, CHU Bordeaux, F-33000 Bordeaux, France
| | - Vincent Probst
- Centre de référence des maladies rythmiques cardiaques, CHU de Nantes, F-44000 Nantes, France
| | - Erwan Donal
- Centre Cardio-Pneumologique, CHU de Rennes Hôpital de Pontchaillou, F-35000 Rennes, France
| | - Pascale Richard
- APHP—Sorbonne Université, Unité Fonctionnelle de Cardiogénétique et Myogénétique Moléculaire, Service de Biochimie Métabolique, HU Pitié Salpêtrière—Charles Foix, F-75013 Paris, France
- INSERM, UMR_S 1166, Sorbonne Université, F-75005 Paris, France
| | - Ana Ferreiro
- Basic and Translational Myology Laboratory, Université Paris Cité, BFA, UMR 8251, CNRS, F-75013 Paris, France
- APHP, Centre de référence des Maladies Neuromusculaires, Institut de Myologie, Neuromyology Department, CHU Pitié Salpêtrière—Charles Foix, F-75013 Paris, France
| | - Brigitte Buendia
- Basic and Translational Myology Laboratory, Université Paris Cité, BFA, UMR 8251, CNRS, F-75013 Paris, France
| |
Collapse
|
15
|
Caravia XM, Ramirez-Martinez A, Gan P, Wang F, McAnally JR, Xu L, Bassel-Duby R, Liu N, Olson EN. Loss of function of the nuclear envelope protein LEMD2 causes DNA damage-dependent cardiomyopathy. J Clin Invest 2022; 132:e158897. [PMID: 36377660 PMCID: PMC9663152 DOI: 10.1172/jci158897] [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: 02/03/2022] [Accepted: 09/13/2022] [Indexed: 11/16/2022] Open
Abstract
Mutations in nuclear envelope proteins (NEPs) cause devastating genetic diseases, known as envelopathies, that primarily affect the heart and skeletal muscle. A mutation in the NEP LEM domain-containing protein 2 (LEMD2) causes severe cardiomyopathy in humans. However, the roles of LEMD2 in the heart and the pathological mechanisms responsible for its association with cardiac disease are unknown. We generated knockin (KI) mice carrying the human c.T38>G Lemd2 mutation, which causes a missense amino acid exchange (p.L13>R) in the LEM domain of the protein. These mice represent a preclinical model that phenocopies the human disease, as they developed severe dilated cardiomyopathy and cardiac fibrosis leading to premature death. At the cellular level, KI/KI cardiomyocytes exhibited disorganization of the transcriptionally silent heterochromatin associated with the nuclear envelope. Moreover, mice with cardiac-specific deletion of Lemd2 also died shortly after birth due to heart abnormalities. Cardiomyocytes lacking Lemd2 displayed nuclear envelope deformations and extensive DNA damage and apoptosis linked to p53 activation. Importantly, cardiomyocyte-specific Lemd2 gene therapy via adeno-associated virus rescued cardiac function in KI/KI mice. Together, our results reveal the essentiality of LEMD2 for genome stability and cardiac function and unveil its mechanistic association with human disease.
Collapse
Affiliation(s)
- Xurde M. Caravia
- Department of Molecular Biology, Hamon Center for Regenerative Science and Medicine
- Senator Paul D. Wellstone Muscular Dystrophy Specialized Research Center, and
| | - Andres Ramirez-Martinez
- Department of Molecular Biology, Hamon Center for Regenerative Science and Medicine
- Senator Paul D. Wellstone Muscular Dystrophy Specialized Research Center, and
| | - Peiheng Gan
- Department of Molecular Biology, Hamon Center for Regenerative Science and Medicine
- Senator Paul D. Wellstone Muscular Dystrophy Specialized Research Center, and
| | - Feng Wang
- Quantitative Biomedical Research Center, Department of Population and Data Sciences and Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - John R. McAnally
- Department of Molecular Biology, Hamon Center for Regenerative Science and Medicine
- Senator Paul D. Wellstone Muscular Dystrophy Specialized Research Center, and
| | - Lin Xu
- Quantitative Biomedical Research Center, Department of Population and Data Sciences and Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Rhonda Bassel-Duby
- Department of Molecular Biology, Hamon Center for Regenerative Science and Medicine
- Senator Paul D. Wellstone Muscular Dystrophy Specialized Research Center, and
| | - Ning Liu
- Department of Molecular Biology, Hamon Center for Regenerative Science and Medicine
- Senator Paul D. Wellstone Muscular Dystrophy Specialized Research Center, and
| | - Eric N. Olson
- Department of Molecular Biology, Hamon Center for Regenerative Science and Medicine
- Senator Paul D. Wellstone Muscular Dystrophy Specialized Research Center, and
| |
Collapse
|
16
|
Schirmer EC, Latonen L, Tollis S. Nuclear size rectification: A potential new therapeutic approach to reduce metastasis in cancer. Front Cell Dev Biol 2022; 10:1022723. [PMID: 36299481 PMCID: PMC9589484 DOI: 10.3389/fcell.2022.1022723] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 09/12/2022] [Indexed: 03/07/2024] Open
Abstract
Research on metastasis has recently regained considerable interest with the hope that single cell technologies might reveal the most critical changes that support tumor spread. However, it is possible that part of the answer has been visible through the microscope for close to 200 years. Changes in nuclear size characteristically occur in many cancer types when the cells metastasize. This was initially discarded as contributing to the metastatic spread because, depending on tumor types, both increases and decreases in nuclear size could correlate with increased metastasis. However, recent work on nuclear mechanics and the connectivity between chromatin, the nucleoskeleton, and the cytoskeleton indicate that changes in this connectivity can have profound impacts on cell mobility and invasiveness. Critically, a recent study found that reversing tumor type-dependent nuclear size changes correlated with reduced cell migration and invasion. Accordingly, it seems appropriate to now revisit possible contributory roles of nuclear size changes to metastasis.
Collapse
Affiliation(s)
- Eric C. Schirmer
- Institute of Cell Biology, University of Edinburgh, Edinburgh, United Kingdom
| | - Leena Latonen
- Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland
- Foundation for the Finnish Cancer Institute, Helsinki, Finland
| | - Sylvain Tollis
- Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland
| |
Collapse
|
17
|
Inesta-Vaquera F, Weiland F, Henderson CJ, Wolf CR. In vivo stress reporters as early biomarkers of the cellular changes associated with progeria. J Cell Mol Med 2022; 26:5463-5472. [PMID: 36201626 PMCID: PMC9639039 DOI: 10.1111/jcmm.17574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 09/15/2022] [Accepted: 09/20/2022] [Indexed: 11/29/2022] Open
Abstract
Age‐related diseases account for a high proportion of the total global burden of disease. Despite recent advances in understanding their molecular basis, there is a lack of suitable early biomarkers to test selected compounds and accelerate their translation to clinical trials. We have investigated the utility of in vivo stress reporter systems as surrogate early biomarkers of the degenerative disease progression. We hypothesized that cellular stress observed in models of human degenerative disease preceded overt cellular damage and at the same time will identify potential cytoprotective pathways. To test this hypothesis, we generated novel accelerated ageing (progeria) reporter mice by crossing the LmnaG609G mice into our oxidative stress/inflammation (Hmox1) and DNA damage (p21) stress reporter models. Histological analysis of reporter expression demonstrated a time‐dependent and tissue‐specific activation of the reporters in tissues directly associated with Progeria, including smooth muscle cells, the vasculature and gastrointestinal tract. Importantly, reporter expression was detected prior to any perceptible deleterious phenotype. Reporter expression can therefore be used as an early marker of progeria pathogenesis and to test therapeutic interventions. This work also demonstrates the potential to use stress reporter approaches to study and find new treatments for other degenerative diseases.
Collapse
Affiliation(s)
- Francisco Inesta-Vaquera
- Division of Systems Medicine, School of Medicine, University of Dundee, Jacqui Wood Cancer Centre, Ninewells Hospital, Dundee, UK
| | - Florian Weiland
- Department of Microbial and Molecular Systems (M2S), Centre for Food and Microbial Technology (CLMT), Laboratory of Enzyme, Fermentation and Brewing Technology (EFBT), Technology Campus Ghent, Ghent, Belgium
| | - Colin J Henderson
- Division of Systems Medicine, School of Medicine, University of Dundee, Jacqui Wood Cancer Centre, Ninewells Hospital, Dundee, UK
| | - Charles Roland Wolf
- Division of Systems Medicine, School of Medicine, University of Dundee, Jacqui Wood Cancer Centre, Ninewells Hospital, Dundee, UK
| |
Collapse
|
18
|
Şener Uslupehlivan E, Deveci R, Şahar U, İzzetoğlu S. Glycan analysis of Lamin A/C protein at G2/M and S phases of the cell cycle. Cell Biochem Biophys 2022; 80:689-698. [PMID: 36180658 DOI: 10.1007/s12013-022-01102-3] [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: 08/03/2022] [Accepted: 09/20/2022] [Indexed: 11/28/2022]
Abstract
During mitosis, phosphorylation and dephosphorylation of lamins triggers the nuclear envelope disassembly/assembly. However, it hasn't been known whether lamin proteins undergo any modification other than phosphorylation during the cell cycle. Glycosylation of lamin proteins is one of the less studied post-translational modification. Glycosylation and phosphorylation compete for the same positions and interplay between two modifications generate a post-translational code in the cell. Based on this, we hypothesized that glycosylation of lamin A/C protein may be important in the regulation of the structural organization of the nuclear lamina during interphase and mitosis. We analysed the glycan units of lamin A/C protein in lung carcinoma cells synchronized at G2/M and S phases via CapLC-ESI-MS/MS. Besides, the outermost glycan units were determined using lectin blotting and gold-conjugated antibody and lectin staining. TEM studies also allowed us to observe the localization of glycosylated lamin A/C protein. With this study, we determined that lamin A/C protein shows O-glycosylation at G2/M and S phases of the cell cycle. In addition to O-GlcNAcylation and O-GalNAcylation, lamin A/C is found to be contain Gal, Fuc, Man, and Sia sugars at G2/M and S phases for the first time. Having found the glycan units of the lamin A/C protein suggests that glycosylation might have a role in the nuclear organization during the cell cycle.
Collapse
Affiliation(s)
- Ecem Şener Uslupehlivan
- Faculty of Science, Department of Biology, Molecular Biology Section, Ege University, Izmir, Turkey
| | - Remziye Deveci
- Faculty of Science, Department of Biology, Molecular Biology Section, Ege University, Izmir, Turkey
| | - Umut Şahar
- Faculty of Science, Department of Biology, Molecular Biology Section, Ege University, Izmir, Turkey
| | - Savaş İzzetoğlu
- Faculty of Science, Department of Biology, Molecular Biology Section, Ege University, Izmir, Turkey.
| |
Collapse
|
19
|
Martín Caballero L, Capella M, Barrales RR, Dobrev N, van Emden T, Hirano Y, Suma Sreechakram VN, Fischer-Burkart S, Kinugasa Y, Nevers A, Rougemaille M, Sinning I, Fischer T, Hiraoka Y, Braun S. The inner nuclear membrane protein Lem2 coordinates RNA degradation at the nuclear periphery. Nat Struct Mol Biol 2022; 29:910-921. [PMID: 36123402 PMCID: PMC9507967 DOI: 10.1038/s41594-022-00831-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 08/02/2022] [Indexed: 11/21/2022]
Abstract
Transcriptionally silent chromatin often localizes to the nuclear periphery. However, whether the nuclear envelope (NE) is a site for post-transcriptional gene repression is not well understood. Here we demonstrate that Schizosaccharomycespombe Lem2, an NE protein, regulates nuclear-exosome-mediated RNA degradation. Lem2 deletion causes accumulation of RNA precursors and meiotic transcripts and de-localization of an engineered exosome substrate from the nuclear periphery. Lem2 does not directly bind RNA but instead interacts with the exosome-targeting MTREC complex and its human homolog PAXT to promote RNA recruitment. This pathway acts largely independently of nuclear bodies where exosome factors assemble. Nutrient availability modulates Lem2 regulation of meiotic transcripts, implying that this pathway is environmentally responsive. Our work reveals that multiple spatially distinct degradation pathways exist. Among these, Lem2 coordinates RNA surveillance of meiotic transcripts and non-coding RNAs by recruiting exosome co-factors to the nuclear periphery. The Braun lab shows that the conserved nuclear membrane protein Lem2 interacts with the MTREC complex of the nuclear-exosome pathway to promote recruitment and degradation of ncRNAs and meiotic transcripts at the nuclear periphery in Schizosaccharomycespombe.
Collapse
Affiliation(s)
- Lucía Martín Caballero
- BioMedical Center (BMC), Division of Physiological Chemistry, Faculty of Medicine, LMU Munich, Planegg-Martinsried, Germany.,International Max Planck Research School for Molecular and Cellular Life Sciences, Planegg-Martinsried, Germany
| | - Matías Capella
- BioMedical Center (BMC), Division of Physiological Chemistry, Faculty of Medicine, LMU Munich, Planegg-Martinsried, Germany.,Instituto de Agrobiotecnología del Litoral, CONICET, Universidad Nacional del Litoral, Santa Fe, Argentina
| | - Ramón Ramos Barrales
- BioMedical Center (BMC), Division of Physiological Chemistry, Faculty of Medicine, LMU Munich, Planegg-Martinsried, Germany.,Centro Andaluz de Biología del Desarrollo (CABD), Universidad Pablo de Olavide-Consejo Superior de Investigaciones Científicas-Junta de Andalucía, Seville, Spain
| | - Nikolay Dobrev
- Heidelberg University Biochemistry Center (BZH), Heidelberg, Germany.,European Molecular Biology Laboratory, Hamburg Unit, Hamburg, Germany
| | - Thomas van Emden
- BioMedical Center (BMC), Division of Physiological Chemistry, Faculty of Medicine, LMU Munich, Planegg-Martinsried, Germany.,International Max Planck Research School for Molecular and Cellular Life Sciences, Planegg-Martinsried, Germany
| | - Yasuhiro Hirano
- Graduate School of Frontier Biosciences, Osaka University, Suita, Japan
| | - Vishnu N Suma Sreechakram
- BioMedical Center (BMC), Division of Physiological Chemistry, Faculty of Medicine, LMU Munich, Planegg-Martinsried, Germany.,Institute for Genetics, Justus-Liebig-University Giessen, Giessen, Germany
| | - Sabine Fischer-Burkart
- BioMedical Center (BMC), Division of Physiological Chemistry, Faculty of Medicine, LMU Munich, Planegg-Martinsried, Germany
| | - Yasuha Kinugasa
- Graduate School of Frontier Biosciences, Osaka University, Suita, Japan.,Regulation for intractable Infectious Diseases, National Institutes of Biomedical Innovation, Health and Nutrition, Ibaraki, Japan
| | - Alicia Nevers
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), Gif-sur-Yvette, France.,University Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France
| | - Mathieu Rougemaille
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), Gif-sur-Yvette, France
| | - Irmgard Sinning
- Heidelberg University Biochemistry Center (BZH), Heidelberg, Germany
| | - Tamás Fischer
- Heidelberg University Biochemistry Center (BZH), Heidelberg, Germany.,The John Curtin School of Medical Research, The Australian National University, Canberra, Australian Capital Territory, Australia
| | - Yasushi Hiraoka
- Graduate School of Frontier Biosciences, Osaka University, Suita, Japan
| | - Sigurd Braun
- BioMedical Center (BMC), Division of Physiological Chemistry, Faculty of Medicine, LMU Munich, Planegg-Martinsried, Germany. .,International Max Planck Research School for Molecular and Cellular Life Sciences, Planegg-Martinsried, Germany. .,Institute for Genetics, Justus-Liebig-University Giessen, Giessen, Germany.
| |
Collapse
|
20
|
Prissette M, Fury W, Koss M, Racioppi C, Fedorova D, Dragileva E, Clarke G, Pohl T, Dugan J, Ahrens D, Chiu J, Hunt C, Siao CJ, Young T, Bhowmick A, Rogulin V, Desclaux M, Hayden EY, Podgorski M, Gao M, Macdonald LE, Frendewey D, Yancopoulos GD, Zambrowicz B. Disruption of nuclear envelope integrity as a possible initiating event in tauopathies. Cell Rep 2022; 40:111249. [PMID: 36001963 DOI: 10.1016/j.celrep.2022.111249] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 06/23/2022] [Accepted: 07/29/2022] [Indexed: 11/03/2022] Open
Abstract
The microtubule-associated protein tau is an abundant component of neurons of the central nervous system. In Alzheimer's disease and other neurodegenerative tauopathies, tau is found hyperphosphorylated and aggregated in neurofibrillary tangles. To obtain a better understanding of the cellular perturbations that initiate tau pathogenesis, we performed a CRISPR-Cas9 screen for genetic modifiers that enhance tau aggregation. This initial screen yielded three genes, BANF1, ANKLE2, and PPP2CA, whose inactivation promotes the accumulation of tau in a phosphorylated and insoluble form. In a complementary screen, we identified three additional genes, LEMD2, LEMD3, and CHMP7, that, when overexpressed, provide protection against tau aggregation. The proteins encoded by the identified genes are mechanistically linked and recognized for their roles in the maintenance and repair of the nuclear envelope. These results implicate the disruption of nuclear envelope integrity as a possible initiating event in tauopathies and reveal targets for therapeutic intervention.
Collapse
Affiliation(s)
| | - Wen Fury
- Regeneron Pharmaceuticals, Tarrytown, NY, USA
| | | | | | | | | | | | - Taylor Pohl
- Regeneron Pharmaceuticals, Tarrytown, NY, USA
| | - John Dugan
- Regeneron Pharmaceuticals, Tarrytown, NY, USA
| | | | - Joyce Chiu
- Regeneron Pharmaceuticals, Tarrytown, NY, USA
| | | | | | - Tara Young
- Regeneron Pharmaceuticals, Tarrytown, NY, USA
| | | | | | | | | | | | - Min Gao
- Regeneron Pharmaceuticals, Tarrytown, NY, USA
| | | | | | | | | |
Collapse
|
21
|
He T, Zhang Y, Li X, Liu C, Zhu G, Yin X, Zhang Z, Zhao K, Wang Z, Zhao P, Wang K. Collective analysis of the expression and prognosis for LEM-domain proteins in prostate cancer. World J Surg Oncol 2022; 20:174. [PMID: 35650630 PMCID: PMC9161513 DOI: 10.1186/s12957-022-02640-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Accepted: 05/19/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Mammalian LEM-domain proteins (LEMs) are encoded by seven genes, including LAP2, EMD, LEMD1, LEMD2, LEMD3, ANKLE1, and ANKLE2. Though some LEMs were involved in various tumor progression, the expression and prognostic values of LEMs in prostate adenocarcinoma (PRAD) have yet to be analyzed. METHODS Herein, we investigated the expression, survival data, and immune infiltration levels of LEMs in PRAD patients from ATCG, TIMER, LinkedOmics, and TISIDB databases. We also further validated the mRNA and protein expression levels of ANKLE1, EMD, and LEMD2 in human prostate tumor specimens by qPCR, WB, and IHC. RESULTS We found that all LEM expressions, except for that of LAP2, were markedly altered in PRAD compared to the normal samples. Among all LEMs, only the expressions of ANKLE1, EMD, and LEMD2 were correlated with advanced tumor stage and survival prognosis in PRAD. Consistent with the predicted computational results, the mRNA and protein expression levels of these genes were markedly increased in the PRAD group. We then found that ANKLE1, EMD, and LEMD2 expressions were markedly correlated with immune cell infiltration levels. High ANKLE1, EMD, and LEMD2 expressions predicted a worse prognosis in PRAD based on immune cells. DNA methylation or/and copy number variations may contribute to the abnormal upregulation of ANKLE1, EMD, and LEMD2 in PRAD. CONCLUSIONS Taken together, this study implied that ANKLE1, EMD, and LEMD2 were promising prognosis predictors and potential immunotherapy targets for PRAD patients.
Collapse
Affiliation(s)
- Tianzhen He
- Institute of Special Environmental Medicine, Nantong University, Nantong, 226019, China.
| | - Yulian Zhang
- Department of Gynecology, The Affiliated Hospital of Qingdao University, Qingdao University, No. 16 Jiangsu Road, Shinan District, Qingdao, 266000, Shandong Province, China
| | - Xueyu Li
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao University, No. 16 Jiangsu Road, Shinan District, Qingdao, 266000, Shandong Province, China
| | - Caihong Liu
- Western Administrative Office Center, Qingdao West Coast New District Health Bureau, No. 166 Shuangzhu Road, Huangdao District, Qingdao, 266000, Shandong Province, China
| | - Guanqun Zhu
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao University, No. 16 Jiangsu Road, Shinan District, Qingdao, 266000, Shandong Province, China
| | - Xinbao Yin
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao University, No. 16 Jiangsu Road, Shinan District, Qingdao, 266000, Shandong Province, China
| | - Zongliang Zhang
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao University, No. 16 Jiangsu Road, Shinan District, Qingdao, 266000, Shandong Province, China
| | - Kai Zhao
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao University, No. 16 Jiangsu Road, Shinan District, Qingdao, 266000, Shandong Province, China
| | - Zhenlin Wang
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao University, No. 16 Jiangsu Road, Shinan District, Qingdao, 266000, Shandong Province, China
| | - Peng Zhao
- Faculty of Sport Science and Coaching, Universiti Pendidikan Sultan Idris, 35900, Tanjong Malim, Perak Darul Ridzuan, Malaysia.
- Athletics Department, Duke Kunshan University, Kunshan, 215316, Jiangsu Province, China.
| | - Ke Wang
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao University, No. 16 Jiangsu Road, Shinan District, Qingdao, 266000, Shandong Province, China.
| |
Collapse
|
22
|
Maccaroni K, La Torre M, Burla R, Saggio I. Phase Separation in the Nucleus and at the Nuclear Periphery during Post-Mitotic Nuclear Envelope Reformation. Cells 2022; 11:1749. [PMID: 35681444 PMCID: PMC9179440 DOI: 10.3390/cells11111749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 05/24/2022] [Accepted: 05/24/2022] [Indexed: 02/04/2023] Open
Abstract
Membrane-enclosed organelle compartmentalization is not the only way by which cell processes are spatially organized. Phase separation is emerging as a new driver in the organization of membrane-less compartments and biological processes. Liquid-liquid phase separation has been indicated as a new way to control the kinetics of molecular reactions and is based on weak multivalent interactions affecting the stoichiometry of the molecules involved. In the nucleus, liquid-liquid phase separation may represent an ancestral means of controlling genomic activity by forming discrete chromatin regions, regulating transcriptional activity, contributing to the assembly of DNA damage response foci, and controlling the organization of chromosomes. Liquid-liquid phase separation also contributes to chromatin function through its role in the reorganization of the nuclear periphery in the post-mitotic phase. Herein, we describe the basic principles regulating liquid-liquid phase separation, analyze examples of phase separation occurring in the nucleus, and dedicate attention to the implication of liquid-liquid phase separation in the reorganization of the nuclear periphery by the endosomal sorting complexes required for transport (ESCRT) machinery. Although some caution is warranted, current scientific knowledge allows for the hypothesis that many factors and processes in the cell are yet to be discovered which are functionally associated with phase separation.
Collapse
Affiliation(s)
- Klizia Maccaroni
- Department of Biology and Biotechnology, Sapienza University, 00185 Rome, Italy; (K.M.); (M.L.T.); (R.B.)
| | - Mattia La Torre
- Department of Biology and Biotechnology, Sapienza University, 00185 Rome, Italy; (K.M.); (M.L.T.); (R.B.)
| | - Romina Burla
- Department of Biology and Biotechnology, Sapienza University, 00185 Rome, Italy; (K.M.); (M.L.T.); (R.B.)
- CNR Institute of Molecular Biology and Pathology, 00185 Rome, Italy
| | - Isabella Saggio
- Department of Biology and Biotechnology, Sapienza University, 00185 Rome, Italy; (K.M.); (M.L.T.); (R.B.)
- CNR Institute of Molecular Biology and Pathology, 00185 Rome, Italy
- Institute of Structural Biology, Nanyang Technological University, Singapore 639798, Singapore
- School of Biological Sciences, Nanyang Technological University, Singapore 639798, Singapore
| |
Collapse
|
23
|
Tollis S, Rizzotto A, Pham NT, Koivukoski S, Sivakumar A, Shave S, Wildenhain J, Zuleger N, Keys JT, Culley J, Zheng Y, Lammerding J, Carragher NO, Brunton VG, Latonen L, Auer M, Tyers M, Schirmer EC. Chemical Interrogation of Nuclear Size Identifies Compounds with Cancer Cell Line-Specific Effects on Migration and Invasion. ACS Chem Biol 2022; 17:680-700. [PMID: 35199530 PMCID: PMC8938924 DOI: 10.1021/acschembio.2c00004] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
![]()
Background: Lower survival rates for many cancer
types correlate with changes in nuclear size/scaling in a tumor-type/tissue-specific
manner. Hypothesizing that such changes might confer an advantage
to tumor cells, we aimed at the identification of commercially available
compounds to guide further mechanistic studies. We therefore screened
for Food and Drug Administration (FDA)/European Medicines Agency (EMA)-approved
compounds that reverse the direction of characteristic tumor nuclear
size changes in PC3, HCT116, and H1299 cell lines reflecting, respectively,
prostate adenocarcinoma, colonic adenocarcinoma, and small-cell squamous
lung cancer. Results: We found distinct, largely
nonoverlapping sets of compounds that rectify nuclear size changes
for each tumor cell line. Several classes of compounds including,
e.g., serotonin uptake inhibitors, cyclo-oxygenase inhibitors, β-adrenergic
receptor agonists, and Na+/K+ ATPase inhibitors,
displayed coherent nuclear size phenotypes focused on a particular
cell line or across cell lines and treatment conditions. Several compounds
from classes far afield from current chemotherapy regimens were also
identified. Seven nuclear size-rectifying compounds selected for further
investigation all inhibited cell migration and/or invasion. Conclusions: Our study provides (a) proof of concept that
nuclear size might be a valuable target to reduce cell migration/invasion
in cancer treatment and (b) the most thorough collection of tool compounds
to date reversing nuclear size changes specific to individual cancer-type
cell lines. Although these compounds still need to be tested in primary
cancer cells, the cell line-specific nuclear size and migration/invasion
responses to particular drug classes suggest that cancer type-specific
nuclear size rectifiers may help reduce metastatic spread.
Collapse
Affiliation(s)
- Sylvain Tollis
- Institute of Biomedicine, University of Eastern Finland, Kuopio 70210, Finland
- Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, Québec H3T 1J4, Canada
| | - Andrea Rizzotto
- The Institute of Cell Biology, University of Edinburgh, Kings Buildings, Michael Swann Buildings, Max Born Crescent, Edinburgh EH9 3BF, U.K
| | - Nhan T. Pham
- Institute of Quantitative Biology, Biochemistry and Biotechnology, University of Edinburgh, Edinburgh EH9 3BF, U.K
| | - Sonja Koivukoski
- Institute of Biomedicine, University of Eastern Finland, Kuopio 70210, Finland
| | - Aishwarya Sivakumar
- The Institute of Cell Biology, University of Edinburgh, Kings Buildings, Michael Swann Buildings, Max Born Crescent, Edinburgh EH9 3BF, U.K
| | - Steven Shave
- Institute of Quantitative Biology, Biochemistry and Biotechnology, University of Edinburgh, Edinburgh EH9 3BF, U.K
| | - Jan Wildenhain
- Institute of Quantitative Biology, Biochemistry and Biotechnology, University of Edinburgh, Edinburgh EH9 3BF, U.K
| | - Nikolaj Zuleger
- The Institute of Cell Biology, University of Edinburgh, Kings Buildings, Michael Swann Buildings, Max Born Crescent, Edinburgh EH9 3BF, U.K
| | - Jeremy T. Keys
- Nancy E. and Peter C. Meinig School of Biomedical Engineering & Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, New York 14853, United States
| | - Jayne Culley
- Edinburgh Cancer Research UK Centre, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh EH4 2XR, U.K
| | - Yijing Zheng
- Institute of Quantitative Biology, Biochemistry and Biotechnology, University of Edinburgh, Edinburgh EH9 3BF, U.K
| | - Jan Lammerding
- Nancy E. and Peter C. Meinig School of Biomedical Engineering & Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, New York 14853, United States
| | - Neil O. Carragher
- Edinburgh Cancer Research UK Centre, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh EH4 2XR, U.K
| | - Valerie G. Brunton
- Edinburgh Cancer Research UK Centre, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh EH4 2XR, U.K
| | - Leena Latonen
- Institute of Biomedicine, University of Eastern Finland, Kuopio 70210, Finland
| | - Manfred Auer
- Institute of Quantitative Biology, Biochemistry and Biotechnology, University of Edinburgh, Edinburgh EH9 3BF, U.K
| | - Mike Tyers
- Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, Québec H3T 1J4, Canada
| | - Eric C. Schirmer
- The Institute of Cell Biology, University of Edinburgh, Kings Buildings, Michael Swann Buildings, Max Born Crescent, Edinburgh EH9 3BF, U.K
| |
Collapse
|
24
|
Mechanisms of A-Type Lamin Targeting to Nuclear Ruptures Are Disrupted in LMNA- and BANF1-Associated Progerias. Cells 2022; 11:cells11050865. [PMID: 35269487 PMCID: PMC8909658 DOI: 10.3390/cells11050865] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 02/23/2022] [Accepted: 02/28/2022] [Indexed: 02/04/2023] Open
Abstract
Mutations in the genes LMNA and BANF1 can lead to accelerated aging syndromes called progeria. The protein products of these genes, A-type lamins and BAF, respectively, are nuclear envelope (NE) proteins that interact and participate in various cellular processes, including nuclear envelope rupture and repair. BAF localizes to sites of nuclear rupture and recruits NE-repair machinery, including the LEM-domain proteins, ESCRT-III complex, A-type lamins, and membranes. Here, we show that it is a mobile, nucleoplasmic population of A-type lamins that is rapidly recruited to ruptures in a BAF-dependent manner via BAF’s association with the Ig-like β fold domain of A-type lamins. These initially mobile lamins become progressively stabilized at the site of rupture. Farnesylated prelamin A and lamin B1 fail to localize to nuclear ruptures, unless that farnesylation is inhibited. Progeria-associated LMNA mutations inhibit the recruitment affected A-type lamin to nuclear ruptures, due to either permanent farnesylation or inhibition of BAF binding. A progeria-associated BAF mutant targets to nuclear ruptures but is unable to recruit A-type lamins. Together, these data reveal the mechanisms that determine how lamins respond to nuclear ruptures and how progeric mutations of LMNA and BANF1 impair recruitment of A-type lamins to nuclear ruptures.
Collapse
|
25
|
Abstract
Lamins interact with a host of nuclear membrane proteins, transcription factors, chromatin regulators, signaling molecules, splicing factors, and even chromatin itself to form a nuclear subcompartment, the nuclear lamina, that is involved in a variety of cellular processes such as the governance of nuclear integrity, nuclear positioning, mitosis, DNA repair, DNA replication, splicing, signaling, mechanotransduction and -sensation, transcriptional regulation, and genome organization. Lamins are the primary scaffold for this nuclear subcompartment, but interactions with lamin-associated peptides in the inner nuclear membrane are self-reinforcing and mutually required. Lamins also interact, directly and indirectly, with peripheral heterochromatin domains called lamina-associated domains (LADs) and help to regulate dynamic 3D genome organization and expression of developmentally regulated genes.
Collapse
Affiliation(s)
- Xianrong Wong
- Laboratory of Developmental and Regenerative Biology, Skin Research Institute of Singapore, Agency for Science, Technology and Research (A∗STAR), Singapore 138648
| | - Ashley J Melendez-Perez
- Department of Biological Chemistry and Center for Epigenetics, Johns Hopkins University of Medicine, Baltimore, Maryland 21205, USA
| | - Karen L Reddy
- Department of Biological Chemistry and Center for Epigenetics, Johns Hopkins University of Medicine, Baltimore, Maryland 21205, USA
- Sidney Kimmel Cancer Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, USA
| |
Collapse
|
26
|
Abstract
The nuclear envelope is composed of the nuclear membranes, nuclear lamina, and nuclear pore complexes. Laminopathies are diseases caused by mutations in genes encoding protein components of the lamina and these other nuclear envelope substructures. Mutations in the single gene encoding lamin A and C, which are expressed in most differentiated somatic cells, cause diseases affecting striated muscle, adipose tissue, peripheral nerve, and multiple systems with features of accelerated aging. Mutations in genes encoding other nuclear envelope proteins also cause an array of diseases that selectively affect different tissues or organs. In some instances, the molecular and cellular consequences of laminopathy-causing mutations are known. However, even when these are understood, mechanisms explaining specific tissue or organ pathology remain enigmatic. Current mechanistic hypotheses focus on how alterations in the nuclear envelope may affect gene expression, including via the regulation of signaling pathways, or cellular mechanics, including responses to mechanical stress.
Collapse
Affiliation(s)
- Ji-Yeon Shin
- Department of Medicine and Department of Pathology and Cell Biology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Howard J. Worman
- Department of Medicine and Department of Pathology and Cell Biology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| |
Collapse
|
27
|
Padilla-Mejia NE, Makarov AA, Barlow LD, Butterfield ER, Field MC. Evolution and diversification of the nuclear envelope. Nucleus 2021; 12:21-41. [PMID: 33435791 PMCID: PMC7889174 DOI: 10.1080/19491034.2021.1874135] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 12/08/2020] [Accepted: 12/11/2020] [Indexed: 02/06/2023] Open
Abstract
Eukaryotic cells arose ~1.5 billion years ago, with the endomembrane system a central feature, facilitating evolution of intracellular compartments. Endomembranes include the nuclear envelope (NE) dividing the cytoplasm and nucleoplasm. The NE possesses universal features: a double lipid bilayer membrane, nuclear pore complexes (NPCs), and continuity with the endoplasmic reticulum, indicating common evolutionary origin. However, levels of specialization between lineages remains unclear, despite distinct mechanisms underpinning various nuclear activities. Several distinct modes of molecular evolution facilitate organellar diversification and to understand which apply to the NE, we exploited proteomic datasets of purified nuclear envelopes from model systems for comparative analysis. We find enrichment of core nuclear functions amongst the widely conserved proteins to be less numerous than lineage-specific cohorts, but enriched in core nuclear functions. This, together with consideration of additional evidence, suggests that, despite a common origin, the NE has evolved as a highly diverse organelle with significant lineage-specific functionality.
Collapse
Affiliation(s)
- Norma E. Padilla-Mejia
- Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Dundee, UK
| | - Alexandr A. Makarov
- Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Dundee, UK
| | - Lael D. Barlow
- Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Dundee, UK
| | - Erin R. Butterfield
- Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Dundee, UK
| | - Mark C. Field
- Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Dundee, UK
- Institute of Parasitology, Biology Centre, Czech Academy of Sciences, České, Czech Republic
| |
Collapse
|
28
|
Yang X, Liu G, Wang Q, Gao X, Xia T, Zhao C, Dou H, Zhang H. Comparative transcriptome provides insights into the selection adaptation between wild and farmed foxes. Ecol Evol 2021; 11:13475-13486. [PMID: 34646484 PMCID: PMC8495804 DOI: 10.1002/ece3.8071] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 06/30/2021] [Accepted: 08/17/2021] [Indexed: 11/09/2022] Open
Abstract
The silver fox and blue fox are economically important fur species and were domesticated by humans from their wild counterparts, the arctic fox and red fox, respectively. Farmed foxes show obvious differences from their wild counterparts, including differences in physiology, body size, energy metabolism, and immunity. However, the molecular mechanisms underlying these differences are presently unclear. In this study, we built transcriptome libraries from multiple pooled tissues for each species of farmed fox, used RNA-seq to obtain a comprehensive dataset, and performed selection analysis and sequence-level analyses of orthologous genes to identify the genes that may be influenced by human domestication. More than 153.3, 248.0, 81.6, and 65.8 million clean reads were obtained and assembled into a total of 118,577, 401,520, 79,900, and 186,988 unigenes with an average length range from 521 to 667 bp for AF, BF, RF, and SF, respectively. Selective pressure analysis showed that 11 and 14 positively selected genes were identified, respectively, in the two groups (AF vs. BF and RF vs. SF). Several of these genes were associated with natural immunity (CFI and LRRFIP1), protein synthesis (GOLGA4, CEP19 and SLC35A2), and DNA damage repair (MDC1). Further functional enrichment analyses demonstrated that two positively selected genes (ACO1 and ACAD10) were involved in metabolic process (GO:0008152, p-value = .032), representing a significant enrichment. Sequence analysis of 117 orthologous genes shared by the two groups showed that the LEMD2, RRBP1, and IGBP1 genes might be affected by artificial selection in farmed foxes, with mutation sites located within sequences that are otherwise highly conserved across most mammals. Our results provide a valuable transcriptomic resource for future genetic studies and improvement in the assisted breeding of foxes and other farmed animals.
Collapse
Affiliation(s)
- Xiufeng Yang
- College of Life ScienceQufu Normal UniversityQufuChina
| | | | - Qi Wang
- Hulunbuir Academy of Inland Lakes in Northern Cold & Arid AreasHulunbuirChina
| | - Xiaodong Gao
- College of Life ScienceQufu Normal UniversityQufuChina
| | - Tian Xia
- College of Life ScienceQufu Normal UniversityQufuChina
| | - Chao Zhao
- College of Life ScienceQufu Normal UniversityQufuChina
| | - Huashan Dou
- Hulunbuir Academy of Inland Lakes in Northern Cold & Arid AreasHulunbuirChina
| | - Honghai Zhang
- College of Life ScienceQufu Normal UniversityQufuChina
| |
Collapse
|
29
|
Nuclear Dynamics and Chromatin Structure: Implications for Pancreatic Cancer. Cells 2021; 10:cells10102624. [PMID: 34685604 PMCID: PMC8534098 DOI: 10.3390/cells10102624] [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: 07/24/2021] [Revised: 09/20/2021] [Accepted: 09/27/2021] [Indexed: 12/14/2022] Open
Abstract
Changes in nuclear shape have been extensively associated with the dynamics and functionality of cancer cells. In most normal cells, nuclei have a regular ellipsoid shape and minimal variation in nuclear size; however, an irregular nuclear contour and abnormal nuclear size is often observed in cancer, including pancreatic cancer. Furthermore, alterations in nuclear morphology have become the 'gold standard' for tumor staging and grading. Beyond the utility of altered nuclear morphology as a diagnostic tool in cancer, the implications of altered nuclear structure for the biology and behavior of cancer cells are profound as changes in nuclear morphology could impact cellular responses to physical strain, adaptation during migration, chromatin organization, and gene expression. Here, we aim to highlight and discuss the factors that regulate nuclear dynamics and their implications for pancreatic cancer biology.
Collapse
|
30
|
Gerull B, Brodehl A. Insights Into Genetics and Pathophysiology of Arrhythmogenic Cardiomyopathy. Curr Heart Fail Rep 2021; 18:378-390. [PMID: 34478111 PMCID: PMC8616880 DOI: 10.1007/s11897-021-00532-z] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/09/2021] [Indexed: 02/07/2023]
Abstract
Purpose of Review Arrhythmogenic cardiomyopathy (ACM) is a genetic disease characterized by life-threatening ventricular arrhythmias and sudden cardiac death (SCD) in apparently healthy young adults. Mutations in genes encoding for cellular junctions can be found in about half of the patients. However, disease onset and severity, risk of arrhythmias, and outcome are highly variable and drug-targeted treatment is currently unavailable. Recent Findings This review focuses on advances in clinical risk stratification, genetic etiology, and pathophysiological concepts. The desmosome is the central part of the disease, but other intercalated disc and associated structural proteins not only broaden the genetic spectrum but also provide novel molecular and cellular insights into the pathogenesis of ACM. Signaling pathways and the role of inflammation will be discussed and targets for novel therapeutic approaches outlined. Summary Genetic discoveries and experimental-driven preclinical research contributed significantly to the understanding of ACM towards mutation- and pathway-specific personalized medicine.
Collapse
Affiliation(s)
- Brenda Gerull
- Comprehensive Heart Failure Center (CHFC), Department of Medicine I, University Clinic Würzburg, Am Schwarzenberg 15, 97078, Würzburg, Germany.
| | - Andreas Brodehl
- Heart and Diabetes Center NRW, Erich and Hanna Klessmann Institute, University Hospital of the Ruhr-University Bochum, Georgstrasse 11, 32545, Bad Oeynhausen, Germany
| |
Collapse
|
31
|
Pawar S, Kutay U. The Diverse Cellular Functions of Inner Nuclear Membrane Proteins. Cold Spring Harb Perspect Biol 2021; 13:a040477. [PMID: 33753404 PMCID: PMC8411953 DOI: 10.1101/cshperspect.a040477] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The nuclear compartment is delimited by a specialized expanded sheet of the endoplasmic reticulum (ER) known as the nuclear envelope (NE). Compared to the outer nuclear membrane and the contiguous peripheral ER, the inner nuclear membrane (INM) houses a unique set of transmembrane proteins that serve a staggering range of functions. Many of these functions reflect the exceptional position of INM proteins at the membrane-chromatin interface. Recent research revealed that numerous INM proteins perform crucial roles in chromatin organization, regulation of gene expression, genome stability, and mediation of signaling pathways into the nucleus. Other INM proteins establish mechanical links between chromatin and the cytoskeleton, help NE remodeling, or contribute to the surveillance of NE integrity and homeostasis. As INM proteins continue to gain prominence, we review these advancements and give an overview on the functional versatility of the INM proteome.
Collapse
Affiliation(s)
- Sumit Pawar
- Institute of Biochemistry, Department of Biology, ETH Zurich, 8093 Zurich, Switzerland
| | - Ulrike Kutay
- Institute of Biochemistry, Department of Biology, ETH Zurich, 8093 Zurich, Switzerland
| |
Collapse
|
32
|
Chen NY, Kim PH, Tu Y, Yang Y, Heizer PJ, Young SG, Fong LG. Increased expression of LAP2β eliminates nuclear membrane ruptures in nuclear lamin-deficient neurons and fibroblasts. Proc Natl Acad Sci U S A 2021; 118:e2107770118. [PMID: 34161290 PMCID: PMC8237679 DOI: 10.1073/pnas.2107770118] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Defects or deficiencies in nuclear lamins cause pathology in many cell types, and recent studies have implicated nuclear membrane (NM) ruptures as a cause of cell toxicity. We previously observed NM ruptures and progressive cell death in the developing brain of lamin B1-deficient mouse embryos. We also observed frequent NM ruptures and DNA damage in nuclear lamin-deficient fibroblasts. Factors modulating susceptibility to NM ruptures remain unclear, but we noted low levels of LAP2β, a chromatin-binding inner NM protein, in fibroblasts with NM ruptures. Here, we explored the apparent link between LAP2β and NM ruptures in nuclear lamin-deficient neurons and fibroblasts, and we tested whether manipulating LAP2β expression levels would alter NM rupture frequency. In cortical plate neurons of lamin B1-deficient embryos, we observed a strong correlation between low LAP2β levels and NM ruptures. We also found low LAP2β levels and frequent NM ruptures in neurons of cultured Lmnb1-/- neurospheres. Reducing LAP2β expression in Lmnb1-/- neurons with an siRNA markedly increased the NM rupture frequency (without affecting NM rupture duration), whereas increased LAP2β expression eliminated NM ruptures and reduced DNA damage. Consistent findings were observed in nuclear lamin-deficient fibroblasts. Reduced LAP2β expression increased NM ruptures, whereas increased LAP2β expression virtually abolished NM ruptures. Increased LAP2β expression nearly abolished NM ruptures in cells subjected to mechanical stress (an intervention that increases NM ruptures). Our studies showed that increasing LAP2β expression bolsters NM integrity in nuclear lamin-deficient cells and markedly reduces NM rupture frequency.
Collapse
Affiliation(s)
- Natalie Y Chen
- Department of Medicine, University of California, Los Angeles, CA 90095
| | - Paul H Kim
- Department of Medicine, University of California, Los Angeles, CA 90095
| | - Yiping Tu
- Department of Medicine, University of California, Los Angeles, CA 90095
| | - Ye Yang
- Department of Medicine, University of California, Los Angeles, CA 90095
| | - Patrick J Heizer
- Department of Medicine, University of California, Los Angeles, CA 90095
| | - Stephen G Young
- Department of Medicine, University of California, Los Angeles, CA 90095;
- Department of Human Genetics, University of California, Los Angeles, CA 90095
- Molecular Biology Institute, University of California, Los Angeles, CA 90095
| | - Loren G Fong
- Department of Medicine, University of California, Los Angeles, CA 90095;
| |
Collapse
|
33
|
Feurle P, Abentung A, Cera I, Wahl N, Ablinger C, Bucher M, Stefan E, Sprenger S, Teis D, Fischer A, Laighneach A, Whitton L, Morris DW, Apostolova G, Dechant G. SATB2-LEMD2 interaction links nuclear shape plasticity to regulation of cognition-related genes. EMBO J 2021; 40:e103701. [PMID: 33319920 PMCID: PMC7849313 DOI: 10.15252/embj.2019103701] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 10/22/2020] [Accepted: 11/06/2020] [Indexed: 01/22/2023] Open
Abstract
SATB2 is a schizophrenia risk gene and is genetically associated with human intelligence. How it affects cognition at molecular level is currently unknown. Here, we show that interactions between SATB2, a chromosomal scaffolding protein, and the inner nuclear membrane protein LEMD2 orchestrate the response of pyramidal neurons to neuronal activation. Exposure to novel environment in vivo causes changes in nuclear shape of CA1 hippocampal neurons via a SATB2-dependent mechanism. The activity-driven plasticity of the nuclear envelope requires not only SATB2, but also its protein interactor LEMD2 and the ESCRT-III/VPS4 membrane-remodeling complex. Furthermore, LEMD2 depletion in cortical neurons, similar to SATB2 ablation, affects neuronal activity-dependent regulation of multiple rapid and delayed primary response genes. In human genetic data, LEMD2-regulated genes are enriched for de novo mutations reported in intellectual disability and schizophrenia and are, like SATB2-regulated genes, enriched for common variants associated with schizophrenia and cognitive function. Hence, interactions between SATB2 and the inner nuclear membrane protein LEMD2 influence gene expression programs in pyramidal neurons that are linked to cognitive ability and psychiatric disorder etiology.
Collapse
Affiliation(s)
- Patrick Feurle
- Institute for NeuroscienceMedical University of InnsbruckInnsbruckAustria
| | - Andreas Abentung
- Institute for NeuroscienceMedical University of InnsbruckInnsbruckAustria
| | - Isabella Cera
- Institute for NeuroscienceMedical University of InnsbruckInnsbruckAustria
| | - Nico Wahl
- Institute for NeuroscienceMedical University of InnsbruckInnsbruckAustria
| | - Cornelia Ablinger
- Institute for NeuroscienceMedical University of InnsbruckInnsbruckAustria
| | - Michael Bucher
- Institute of Biochemistry and Center for Molecular BiosciencesUniversity of InnsbruckInnsbruckAustria
| | - Eduard Stefan
- Institute of Biochemistry and Center for Molecular BiosciencesUniversity of InnsbruckInnsbruckAustria
| | - Simon Sprenger
- Institute for Cell BiologyMedical University of InnsbruckInnsbruckAustria
| | - David Teis
- Institute for Cell BiologyMedical University of InnsbruckInnsbruckAustria
| | - Andre Fischer
- Department of Systems Medicine and EpigeneticsGerman Center for Neurodegenerative Diseases (DZNE)GoettingenGermany
- Department of Psychiatry and PsychotherapyUniversity Medical CenterGoettingenGermany
| | - Aodán Laighneach
- Neuroimaging, Cognition & Genomics (NICOG) CentreSchool of Psychology and Discipline of BiochemistryNational University of Ireland GalwayGalwayIreland
| | - Laura Whitton
- Neuroimaging, Cognition & Genomics (NICOG) CentreSchool of Psychology and Discipline of BiochemistryNational University of Ireland GalwayGalwayIreland
| | - Derek W Morris
- Neuroimaging, Cognition & Genomics (NICOG) CentreSchool of Psychology and Discipline of BiochemistryNational University of Ireland GalwayGalwayIreland
| | - Galina Apostolova
- Institute for NeuroscienceMedical University of InnsbruckInnsbruckAustria
| | - Georg Dechant
- Institute for NeuroscienceMedical University of InnsbruckInnsbruckAustria
| |
Collapse
|
34
|
The nuclear envelope protein Net39 is essential for muscle nuclear integrity and chromatin organization. Nat Commun 2021; 12:690. [PMID: 33514739 PMCID: PMC7846557 DOI: 10.1038/s41467-021-20987-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Accepted: 12/24/2020] [Indexed: 01/14/2023] Open
Abstract
Lamins and transmembrane proteins within the nuclear envelope regulate nuclear structure and chromatin organization. Nuclear envelope transmembrane protein 39 (Net39) is a muscle nuclear envelope protein whose functions in vivo have not been explored. We show that mice lacking Net39 succumb to severe myopathy and juvenile lethality, with concomitant disruption in nuclear integrity, chromatin accessibility, gene expression, and metabolism. These abnormalities resemble those of Emery-Dreifuss muscular dystrophy (EDMD), caused by mutations in A-type lamins (LMNA) and other genes, like Emerin (EMD). We observe that Net39 is downregulated in EDMD patients, implicating Net39 in the pathogenesis of this disorder. Our findings highlight the role of Net39 at the nuclear envelope in maintaining muscle chromatin organization, gene expression and function, and its potential contribution to the molecular etiology of EDMD.
Collapse
|
35
|
Nuclear Envelope Proteins Modulating the Heterochromatin Formation and Functions in Fission Yeast. Cells 2020; 9:cells9081908. [PMID: 32824370 PMCID: PMC7464478 DOI: 10.3390/cells9081908] [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: 07/30/2020] [Revised: 08/14/2020] [Accepted: 08/15/2020] [Indexed: 12/16/2022] Open
Abstract
The nuclear envelope (NE) consists of the inner and outer nuclear membranes (INM and ONM), and the nuclear pore complex (NPC), which penetrates the double membrane. ONM continues with the endoplasmic reticulum (ER). INM and NPC can interact with chromatin to regulate the genetic activities of the chromosome. Studies in the fission yeast Schizosaccharomyces pombe have contributed to understanding the molecular mechanisms underlying heterochromatin formation by the RNAi-mediated and histone deacetylase machineries. Recent studies have demonstrated that NE proteins modulate heterochromatin formation and functions through interactions with heterochromatic regions, including the pericentromeric and the sub-telomeric regions. In this review, we first introduce the molecular mechanisms underlying the heterochromatin formation and functions in fission yeast, and then summarize the NE proteins that play a role in anchoring heterochromatic regions and in modulating heterochromatin formation and functions, highlighting roles for a conserved INM protein, Lem2.
Collapse
|
36
|
Unrestrained ESCRT-III drives micronuclear catastrophe and chromosome fragmentation. Nat Cell Biol 2020; 22:856-867. [PMID: 32601372 DOI: 10.1038/s41556-020-0537-5] [Citation(s) in RCA: 85] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Accepted: 05/24/2020] [Indexed: 12/11/2022]
Abstract
The ESCRT-III membrane fission machinery maintains the integrity of the nuclear envelope. Although primary nuclei resealing takes minutes, micronuclear envelope ruptures seem to be irreversible. Instead, micronuclear ruptures result in catastrophic membrane collapse and are associated with chromosome fragmentation and chromothripsis, complex chromosome rearrangements thought to be a major driving force in cancer development. Here we use a combination of live microscopy and electron tomography, as well as computer simulations, to uncover the mechanism underlying micronuclear collapse. We show that, due to their small size, micronuclei inherently lack the capacity of primary nuclei to restrict the accumulation of CHMP7-LEMD2, a compartmentalization sensor that detects loss of nuclear integrity. This causes unrestrained ESCRT-III accumulation, which drives extensive membrane deformation, DNA damage and chromosome fragmentation. Thus, the nuclear-integrity surveillance machinery is a double-edged sword, as its sensitivity ensures rapid repair at primary nuclei while causing unrestrained activity at ruptured micronuclei, with catastrophic consequences for genome stability.
Collapse
|
37
|
von Appen A, LaJoie D, Johnson IE, Trnka MJ, Pick SM, Burlingame AL, Ullman KS, Frost A. LEM2 phase separation promotes ESCRT-mediated nuclear envelope reformation. Nature 2020; 582:115-118. [PMID: 32494070 PMCID: PMC7321842 DOI: 10.1038/s41586-020-2232-x] [Citation(s) in RCA: 108] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Accepted: 02/26/2020] [Indexed: 01/01/2023]
Abstract
During cell division, remodelling of the nuclear envelope enables chromosome segregation by the mitotic spindle1. The reformation of sealed nuclei requires ESCRTs (endosomal sorting complexes required for transport) and LEM2, a transmembrane ESCRT adaptor2-4. Here we show how the ability of LEM2 to condense on microtubules governs the activation of ESCRTs and coordinated spindle disassembly. The LEM motif of LEM2 binds BAF, conferring on LEM2 an affinity for chromatin5,6, while an adjacent low-complexity domain (LCD) promotes LEM2 phase separation. A proline-arginine-rich sequence within the LCD binds to microtubules and targets condensation of LEM2 to spindle microtubules that traverse the nascent nuclear envelope. Furthermore, the winged-helix domain of LEM2 activates the ESCRT-II/ESCRT-III hybrid protein CHMP7 to form co-oligomeric rings. Disruption of these events in human cells prevented the recruitment of downstream ESCRTs, compromised spindle disassembly, and led to defects in nuclear integrity and DNA damage. We propose that during nuclear reassembly LEM2 condenses into a liquid-like phase and coassembles with CHMP7 to form a macromolecular O-ring seal at the confluence between membranes, chromatin and the spindle. The properties of LEM2 described here, and the homologous architectures of related inner nuclear membrane proteins7,8, suggest that phase separation may contribute to other critical envelope functions, including interphase repair8-13 and chromatin organization14-17.
Collapse
Affiliation(s)
- Alexander von Appen
- Department of Biochemistry and Biophysics, University of California, San Francisco, CA, USA
| | - Dollie LaJoie
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Isabel E Johnson
- Department of Biochemistry and Biophysics, University of California, San Francisco, CA, USA
| | - Michael J Trnka
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA
| | - Sarah M Pick
- Faculty of Chemistry and Pharmacy, University of Freiburg, Freiburg, Germany
| | - Alma L Burlingame
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA
| | - Katharine S Ullman
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA.
| | - Adam Frost
- Department of Biochemistry and Biophysics, University of California, San Francisco, CA, USA.
- Chan Zuckerberg Biohub, San Francisco, CA, USA.
- Quantitative Biosciences Institute, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
38
|
Moser B, Basílio J, Gotzmann J, Brachner A, Foisner R. Comparative Interactome Analysis of Emerin, MAN1 and LEM2 Reveals a Unique Role for LEM2 in Nucleotide Excision Repair. Cells 2020; 9:E463. [PMID: 32085595 PMCID: PMC7072835 DOI: 10.3390/cells9020463] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 02/06/2020] [Accepted: 02/11/2020] [Indexed: 12/19/2022] Open
Abstract
LAP2-Emerin-MAN1 (LEM) domain-containing proteins represent an abundant group of inner nuclear membrane proteins involved in diverse nuclear functions, but their functional redundancies remain unclear. Here, using the biotinylation-dependent proximity approach, we report proteome-wide comparative interactome analysis of the two structurally related LEM proteins MAN1 (LEMD3) and LEM2 (LEMD2), and the more distantly related emerin (EMD). While over 60% of the relatively small group of MAN1 and emerin interactors were also found in the LEM2 interactome, the latter included a large number of candidates (>85%) unique for LEM2. The interacting partners unique for emerin support and provide further insight into the previously reported role of emerin in centrosome positioning, and the MAN1-specific interactors suggest a role of MAN1 in ribonucleoprotein complex assembly. Interestingly, the LEM2-specific interactome contained several proteins of the nucleotide excision repair pathway. Accordingly, LEM2-depleted cells, but not MAN1- and emerin-depleted cells, showed impaired proliferation following ultraviolet-C (UV-C) irradiation and prolonged accumulation of γH2AX, similar to cells deficient in the nucleotide excision repair protein DNA damage-binding protein 1 (DDB1). These findings indicate impaired DNA damage repair in LEM2-depleted cells. Overall, this interactome study identifies new potential interaction partners of emerin, MAN1 and particularly LEM2, and describes a novel potential involvement of LEM2 in nucleotide excision repair at the nuclear periphery.
Collapse
Affiliation(s)
- Bernhard Moser
- Max Perutz Labs, Center of Medical Biochemistry, Medical University of Vienna, Vienna Biocenter (VBC), 1030 Vienna, Austria; (B.M.); (J.G.)
- Department of Vascular Biology and Thrombosis Research, Center for Physiology and Pharmacology, Medical University of Vienna, 1090 Vienna, Austria
| | - José Basílio
- Department of Vascular Biology and Thrombosis Research, Center for Physiology and Pharmacology, Medical University of Vienna, 1090 Vienna, Austria
| | - Josef Gotzmann
- Max Perutz Labs, Center of Medical Biochemistry, Medical University of Vienna, Vienna Biocenter (VBC), 1030 Vienna, Austria; (B.M.); (J.G.)
| | - Andreas Brachner
- Max Perutz Labs, Center of Medical Biochemistry, Medical University of Vienna, Vienna Biocenter (VBC), 1030 Vienna, Austria; (B.M.); (J.G.)
| | - Roland Foisner
- Max Perutz Labs, Center of Medical Biochemistry, Medical University of Vienna, Vienna Biocenter (VBC), 1030 Vienna, Austria; (B.M.); (J.G.)
| |
Collapse
|
39
|
Tarsani E, Kranis A, Maniatis G, Avendano S, Hager-Theodorides AL, Kominakis A. Discovery and characterization of functional modules associated with body weight in broilers. Sci Rep 2019; 9:9125. [PMID: 31235723 PMCID: PMC6591351 DOI: 10.1038/s41598-019-45520-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Accepted: 06/04/2019] [Indexed: 12/31/2022] Open
Abstract
Aim of the present study was to investigate whether body weight (BW) in broilers is associated with functional modular genes. To this end, first a GWAS for BW was conducted using 6,598 broilers and the high density SNP array. The next step was to search for positional candidate genes and QTLs within strong LD genomic regions around the significant SNPs. Using all positional candidate genes, a network was then constructed and community structure analysis was performed. Finally, functional enrichment analysis was applied to infer the functional relevance of modular genes. A total number of 645 positional candidate genes were identified in strong LD genomic regions around 11 genome-wide significant markers. 428 of the positional candidate genes were located within growth related QTLs. Community structure analysis detected 5 modules while functional enrichment analysis showed that 52 modular genes participated in developmental processes such as skeletal system development. An additional number of 14 modular genes (GABRG1, NGF, APOBEC2, STAT5B, STAT3, SMAD4, MED1, CACNB1, SLAIN2, LEMD2, ZC3H18, TMEM132D, FRYL and SGCB) were also identified as related to body weight. Taken together, current results suggested a total number of 66 genes as most plausible functional candidates for the trait examined.
Collapse
Affiliation(s)
- Eirini Tarsani
- Department of Animal Science and Aquaculture, Agricultural University of Athens, Iera Odos 75, 11855, Athens, Greece.
| | - Andreas Kranis
- Aviagen Ltd., Newbridge, Midlothian, EH28 8SZ, UK.,The Roslin Institute, University of Edinburgh, EH25 9RG, Midlothian, United Kingdom
| | | | | | - Ariadne L Hager-Theodorides
- Department of Animal Science and Aquaculture, Agricultural University of Athens, Iera Odos 75, 11855, Athens, Greece
| | - Antonios Kominakis
- Department of Animal Science and Aquaculture, Agricultural University of Athens, Iera Odos 75, 11855, Athens, Greece
| |
Collapse
|
40
|
Kinugasa Y, Hirano Y, Sawai M, Ohno Y, Shindo T, Asakawa H, Chikashige Y, Shibata S, Kihara A, Haraguchi T, Hiraoka Y. The very-long-chain fatty acid elongase Elo2 rescues lethal defects associated with loss of the nuclear barrier function in fission yeast cells. J Cell Sci 2019; 132:jcs.229021. [PMID: 30975915 DOI: 10.1242/jcs.229021] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 04/03/2019] [Indexed: 12/21/2022] Open
Abstract
In eukaryotic cells, chromosomes are confined to the nucleus, which is compartmentalized by the nuclear membranes; these are continuous with the endoplasmic reticulum membranes. Maintaining the homeostasis of these membranes is an important cellular activity performed by lipid metabolic enzymes. However, how lipid metabolic enzymes affect nuclear membrane functions remains to be elucidated. We found that the very-long-chain fatty acid elongase Elo2 is located in the nuclear membrane and prevents lethal defects associated with nuclear membrane ruptures in mutants of the nuclear membrane proteins Lem2 and Bqt4 in the fission yeast Schizosaccharomyces pombe. Lipid composition analysis shows that t20:0/24:0 phytoceramide (a conjugate of C20:0 phytosphingosine and C24:0 fatty acid) is a major ceramide species in S. pombe The quantity of this ceramide is reduced in the absence of Lem2, and restored by increased expression of Elo2. Furthermore, loss of S. pombe Elo2 can be rescued by its human orthologs. These results suggest that the conserved very-long-chain fatty acid elongase producing the ceramide component is essential for nuclear membrane integrity and cell viability in eukaryotes.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Yasuha Kinugasa
- Graduate School of Frontier Biosciences, Osaka University, Suita 565-0871, Japan
| | - Yasuhiro Hirano
- Graduate School of Frontier Biosciences, Osaka University, Suita 565-0871, Japan
| | - Megumi Sawai
- Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo 060-0812, Japan
| | - Yusuke Ohno
- Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo 060-0812, Japan
| | - Tomoko Shindo
- Electron Microscope Laboratory, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Haruhiko Asakawa
- Graduate School of Frontier Biosciences, Osaka University, Suita 565-0871, Japan
| | - Yuji Chikashige
- Advanced ICT Research Institute Kobe, National Institute of Information and Communications Technology, Kobe 651-2492, Japan
| | - Shinsuke Shibata
- Electron Microscope Laboratory, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Akio Kihara
- Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo 060-0812, Japan
| | - Tokuko Haraguchi
- Graduate School of Frontier Biosciences, Osaka University, Suita 565-0871, Japan.,Advanced ICT Research Institute Kobe, National Institute of Information and Communications Technology, Kobe 651-2492, Japan
| | - Yasushi Hiraoka
- Graduate School of Frontier Biosciences, Osaka University, Suita 565-0871, Japan .,Advanced ICT Research Institute Kobe, National Institute of Information and Communications Technology, Kobe 651-2492, Japan
| |
Collapse
|
41
|
Abdelfatah N, Chen R, Duff HJ, Seifer CM, Buffo I, Huculak C, Clarke S, Clegg R, Jassal DS, Gordon PMK, Ober C, Frosk P, Gerull B. Characterization of a Unique Form of Arrhythmic Cardiomyopathy Caused by Recessive Mutation in LEMD2. JACC Basic Transl Sci 2019; 4:204-221. [PMID: 31061923 PMCID: PMC6488817 DOI: 10.1016/j.jacbts.2018.12.001] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Revised: 11/02/2018] [Accepted: 12/03/2018] [Indexed: 02/08/2023]
Abstract
Nuclear envelope proteins have been shown to play an important role in the pathogenesis of inherited dilated cardiomyopathy. Here, we present a remarkable cardiac phenotype caused by a homozygous LEMD2 mutation in patients of the Hutterite population with juvenile cataract. Mutation carriers develop arrhythmic cardiomyopathy with mild impairment of left ventricular systolic function but severe ventricular arrhythmias leading to sudden cardiac death. Affected cardiac tissue from a deceased patient and fibroblasts exhibit elongated nuclei with abnormal condensed heterochromatin at the periphery. The patient fibroblasts demonstrate cellular senescence and reduced proliferation capacity, which may suggest an involvement of LEM domain containing protein 2 in chromatin remodeling processes and premature aging.
Collapse
Key Words
- ACM, arrhythmogenic cardiomyopathy
- BANF, barrier to autointegration factor
- CMR, cardiac magnetic resonance
- DAPI, 4′,6′-diamidino-2-phenylindole
- DCM, dilated cardiomyopathy
- DNA, deoxyribonucleic acid
- EMD, emerin
- ICD, implantable cardioverter-defibrillator
- LEMD2
- LEMD2, LEM domain containing protein 2
- LGE, late gadolinium enhancement
- LMNA, lamin A/C
- LV, left ventricular
- NE, nuclear envelope
- P, passage
- PBS, phosphate-buffered saline
- SAHF, senescence-associated heterochromatin foci
- SNV, single nucleotide variant
- chromatin remodeling
- dilated cardiomyopathy
- eGFP, enhanced green fluorescent protein
- inner nuclear membrane
- sudden death
Collapse
Affiliation(s)
- Nelly Abdelfatah
- Department of Cardiac Sciences, Libin Cardiovascular Institute of Alberta, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Ruping Chen
- Comprehensive Heart Failure Center, University Hospital Würzburg, Würzburg, Germany
| | - Henry J Duff
- Department of Cardiac Sciences, Libin Cardiovascular Institute of Alberta, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Colette M Seifer
- Section of Cardiology, Department of Internal Medicine, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Ilan Buffo
- Variety Children's Heart Centre, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Cathleen Huculak
- Department of Medical Genetics, Alberta Health Services, Calgary, Alberta, Canada
| | - Stephanie Clarke
- Department of Biochemistry and Medical Genetics, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Robin Clegg
- Department of Pediatrics, University of Calgary, Calgary, Alberta, Canada
| | - Davinder S Jassal
- Section of Cardiology, Department of Internal Medicine, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Paul M K Gordon
- Cumming School of Medicine Centre for Health Genomics and Informatics, University of Calgary, Calgary, Alberta, Canada
| | - Carole Ober
- Department of Human Genetics, The University of Chicago, Chicago, Illinois
| | | | - Patrick Frosk
- Department of Biochemistry and Medical Genetics, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada.,Department of Pediatrics and Child Health, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Brenda Gerull
- Department of Cardiac Sciences, Libin Cardiovascular Institute of Alberta, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.,Comprehensive Heart Failure Center, University Hospital Würzburg, Würzburg, Germany.,Department of Internal Medicine I, University Hospital Würzburg, Würzburg, Germany
| |
Collapse
|
42
|
The Discovery of a LEMD2-Associated Nuclear Envelopathy with Early Progeroid Appearance Suggests Advanced Applications for AI-Driven Facial Phenotyping. Am J Hum Genet 2019; 104:749-757. [PMID: 30905398 DOI: 10.1016/j.ajhg.2019.02.021] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Accepted: 02/16/2019] [Indexed: 12/16/2022] Open
Abstract
Over a relatively short period of time, the clinical geneticist's "toolbox" has been expanded by machine-learning algorithms for image analysis, which can be applied to the task of syndrome identification on the basis of facial photographs, but these technologies harbor potential beyond the recognition of established phenotypes. Here, we comprehensively characterized two individuals with a hitherto unknown genetic disorder caused by the same de novo mutation in LEMD2 (c.1436C>T;p.Ser479Phe), the gene which encodes the nuclear envelope protein LEM domain-containing protein 2 (LEMD2). Despite different ages and ethnic backgrounds, both individuals share a progeria-like facial phenotype and a distinct combination of physical and neurologic anomalies, such as growth retardation; hypoplastic jaws crowded with multiple supernumerary, yet unerupted, teeth; and cerebellar intention tremor. Immunofluorescence analyses of patient fibroblasts revealed mutation-induced disturbance of nuclear architecture, recapitulating previously published data in LEMD2-deficient cell lines, and additional experiments suggested mislocalization of mutant LEMD2 protein within the nuclear lamina. Computational analysis of facial features with two different deep neural networks showed phenotypic proximity to other nuclear envelopathies. One of the algorithms, when trained to recognize syndromic similarity (rather than specific syndromes) in an unsupervised approach, clustered both individuals closely together, providing hypothesis-free hints for a common genetic etiology. We show that a recurrent de novo mutation in LEMD2 causes a nuclear envelopathy whose prognosis in adolescence is relatively good in comparison to that of classical Hutchinson-Gilford progeria syndrome, and we suggest that the application of artificial intelligence to the analysis of patient images can facilitate the discovery of new genetic disorders.
Collapse
|
43
|
Iwamoto M, Fukuda Y, Osakada H, Mori C, Hiraoka Y, Haraguchi T. Identification of the evolutionarily conserved nuclear envelope proteins Lem2 and MicLem2 in Tetrahymena thermophila. Gene 2019; 721S:100006. [PMID: 32550543 PMCID: PMC7285967 DOI: 10.1016/j.gene.2019.100006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 12/18/2018] [Accepted: 01/11/2019] [Indexed: 11/26/2022]
Abstract
Lem2 family proteins, i.e. the LAP2-Emerin-MAN1 (LEM) domain-containing nuclear envelope proteins, are well-conserved from yeasts to humans, both of which belong to the Opisthokonta supergroup. However, whether their homologs are present in other eukaryotic phylogenies remains unclear. In this study, we identified two Lem2 homolog proteins, which we named as Lem2 and MicLem2, in a ciliate Tetrahymena thermophila belonging to the SAR supergroup. Lem2 was localized to the nuclear envelope of the macronucleus (MAC) and micronucleus (MIC), while MicLem2 was exclusively localized to the nuclear envelope of the MIC. Immunoelectron microscopy revealed that Lem2 in T. thermophila was localized to both the inner and outer nuclear envelopes of the MAC and MIC, while MicLem2 was mostly localized to the nuclear pores of the MIC. Molecular domain analysis using GFP-fused protein showed that the N-terminal and luminal domains, including the transmembrane segments, are responsible for nuclear envelope localization. During sexual reproduction, enrichment of Lem2 occurred in the nuclear envelopes of the MAC and MIC to be degraded, while MicLem2 was enriched in the nuclear envelope of the MIC that escaped degradation. These findings suggest the unique characteristics of Tetrahymena Lem2 proteins. Our findings provide insight into the evolutionary divergence of nuclear envelope proteins. Conserved nuclear envelope proteins Lem2 and MicLem2 are identified in Tetrahymena. Lem2 is localized to the nuclear envelope of the macronucleus and the micronucleus. MicLem2 is localized to the nuclear pore complex of the micronucleus. In sexual reproduction, Lem2 is enriched to the nuclei assigned to degradation. MicLem2 is enriched to the micronuclei that are escaped from degradation.
Collapse
Key Words
- BAF, barrier-to-autointegration factor
- DAPI, 4′,6‑diamidino‑2‑phenylindole
- DDW, double distilled water
- EDTA, ethylenediaminetetraacetic acid
- ER, endoplasmic reticulum
- GA, glutaraldehyde
- HeH domain
- HeH, helix-extension-helix
- LAP2, lamina associated polypeptide 2
- LEM domain
- LEM, LAP2-Emerin-MAN1
- MAC, macronucleus
- MIC, micronucleus
- MSC domain
- MSC, Man1-Src1p-C-terminal
- Man1
- Man1-Src1p-C-terminal domain
- NE, nuclear envelope
- NLS, nuclear localization signal
- NPC, nuclear pore complex
- Nuclear dimorphism
- Nuclear envelope
- ONM and INM, outer and inner nuclear membranes
- PB, phosphate buffer
- PBS, phosphate buffered saline
- Protist
- RRM, RNA recognition motif
- TM, transmembrane
Collapse
Affiliation(s)
- Masaaki Iwamoto
- Advanced ICT Research Institute Kobe, National Institute of Information and Communications Technology, Kobe 651-2492, Japan
| | - Yasuhiro Fukuda
- Graduate School of Agricultural Science, Tohoku University, Osaki, 989-6711, Japan
| | - Hiroko Osakada
- Advanced ICT Research Institute Kobe, National Institute of Information and Communications Technology, Kobe 651-2492, Japan
| | - Chie Mori
- Advanced ICT Research Institute Kobe, National Institute of Information and Communications Technology, Kobe 651-2492, Japan
| | - Yasushi Hiraoka
- Advanced ICT Research Institute Kobe, National Institute of Information and Communications Technology, Kobe 651-2492, Japan.,Graduate School of Frontier Biosciences, Osaka University, Suita 565-0871, Japan
| | - Tokuko Haraguchi
- Advanced ICT Research Institute Kobe, National Institute of Information and Communications Technology, Kobe 651-2492, Japan.,Graduate School of Frontier Biosciences, Osaka University, Suita 565-0871, Japan
| |
Collapse
|
44
|
Paci M, Elkhatib R, Longepied G, Bourgeois P, Ray PF, Levy N, Mitchell MJ, Metzler-Guillemain C. The involvement of the nuclear lamina in human and rodent spermiogenesis: a systematic review. Basic Clin Androl 2018; 28:7. [PMID: 29946470 PMCID: PMC6008938 DOI: 10.1186/s12610-018-0072-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Accepted: 04/19/2018] [Indexed: 12/11/2022] Open
Abstract
The nuclear lamina (NL) is a filamentous protein meshwork, composed essentially of lamins, situated between the inner nuclear membrane and the chromatin. The NL is a component of the nuclear envelope, interacts with a wide range of proteins and is required for normal nuclear structure and physiological development. During spermiogenesis the spermatid nucleus is elongated, and dramatically reduced in size with protamines replacing histones to produce a highly compacted chromatin. There is mounting evidence from studies in human and rodent, that the NL plays an important role in mammalian spermatid differentiation during spermiogenesis. In this review, we summarize and discuss the data available in the literature regarding the involvement of lamins and their direct or indirect partners in normal and abnormal human spermiogenesis.
Collapse
Affiliation(s)
- Marine Paci
- 1Aix Marseille Univ, Inserm, MMG, U1251, Marseille Medical Genetics, 13385 Marseille, France.,APHM Hôpital La Conception, Pôle femmes-Parents-enfants, Centre Clinico-Biologique d'Assistance Médicale à la Procréation-CECOS, 13385 Marseille Cedex 5, France
| | - Razan Elkhatib
- 1Aix Marseille Univ, Inserm, MMG, U1251, Marseille Medical Genetics, 13385 Marseille, France
| | - Guy Longepied
- 1Aix Marseille Univ, Inserm, MMG, U1251, Marseille Medical Genetics, 13385 Marseille, France
| | - Patrice Bourgeois
- 1Aix Marseille Univ, Inserm, MMG, U1251, Marseille Medical Genetics, 13385 Marseille, France
| | - Pierre F Ray
- 3Genetic Epigenetic and Therapies of Infertility, Institute for Advanced Biosciences, Inserm U1209, CNRS UMR 5309, Université Grenoble Alpes, CHU Grenoble Alpes, F-38000 Grenoble, France
| | - Nicolas Levy
- 1Aix Marseille Univ, Inserm, MMG, U1251, Marseille Medical Genetics, 13385 Marseille, France
| | - Michael J Mitchell
- 1Aix Marseille Univ, Inserm, MMG, U1251, Marseille Medical Genetics, 13385 Marseille, France
| | - Catherine Metzler-Guillemain
- 1Aix Marseille Univ, Inserm, MMG, U1251, Marseille Medical Genetics, 13385 Marseille, France.,APHM Hôpital La Conception, Pôle femmes-Parents-enfants, Centre Clinico-Biologique d'Assistance Médicale à la Procréation-CECOS, 13385 Marseille Cedex 5, France
| |
Collapse
|
45
|
Linker of nucleoskeleton and cytoskeleton complex proteins in cardiomyopathy. Biophys Rev 2018; 10:1033-1051. [PMID: 29869195 PMCID: PMC6082319 DOI: 10.1007/s12551-018-0431-6] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Accepted: 05/24/2018] [Indexed: 12/21/2022] Open
Abstract
The linker of nucleoskeleton and cytoskeleton (LINC) complex couples the nuclear lamina to the cytoskeleton. The LINC complex and its associated proteins play diverse roles in cells, ranging from genome organization, nuclear morphology, gene expression, to mechanical stability. The importance of a functional LINC complex is highlighted by the large number of mutations in genes encoding LINC complex proteins that lead to skeletal and cardiac myopathies. In this review, the structure, function, and interactions between components of the LINC complex will be described. Mutations that are known to cause cardiomyopathy in patients will be discussed alongside their respective mouse models. Furthermore, future challenges for the field and emerging technologies to investigate LINC complex function will be discussed.
Collapse
|
46
|
Hirano Y, Kinugasa Y, Asakawa H, Chikashige Y, Obuse C, Haraguchi T, Hiraoka Y. Lem2 is retained at the nuclear envelope through its interaction with Bqt4 in fission yeast. Genes Cells 2018; 23:122-135. [PMID: 29292846 DOI: 10.1111/gtc.12557] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Accepted: 12/04/2017] [Indexed: 12/31/2022]
Abstract
Inner nuclear membrane (INM) proteins are thought to play important roles in modulating nuclear organization and function through their interactions with chromatin. However, these INM proteins share redundant functions in metazoans that pose difficulties for functional studies. The fission yeast Schizosaccharomyces pombe exhibits a relatively small number of INM proteins, and molecular genetic tools are available to separate their redundant functions. In S. pombe, it has been reported that among potentially redundant INM proteins, Lem2 displays a unique genetic interaction with another INM protein, Bqt4, which is involved in anchoring telomeres to the nuclear envelope. Double mutations in the lem2 and bqt4 genes confer synthetic lethality during vegetative growth. Here, we show that Lem2 is retained at the nuclear envelope through its interaction with Bqt4, as the loss of Bqt4 results in the exclusive accumulation of Lem2 to the spindle pole body (SPB). An N-terminal nucleoplasmic region of Lem2 bears affinity to both Bqt4 and the SPB in a competitive manner. In contrast, the synthetic lethality of the lem2 bqt4 double mutant is suppressed by the C-terminal region of Lem2. These results indicate that the N-terminal and C-terminal domains of Lem2 show independent functions with respect to Bqt4.
Collapse
Affiliation(s)
- Yasuhiro Hirano
- Graduate School of Frontier Biosciences, Osaka University, Suita, Japan
| | - Yasuha Kinugasa
- Graduate School of Frontier Biosciences, Osaka University, Suita, Japan
| | - Haruhiko Asakawa
- Graduate School of Frontier Biosciences, Osaka University, Suita, Japan
| | - Yuji Chikashige
- Advanced ICT Research Institute Kobe, National Institute of Information and Communications Technology, Kobe, Japan
| | - Chikashi Obuse
- Department of Biological Sciences, Graduate School of Science, Osaka University, Toyonaka, Osaka, Japan.,Graduate School of Life Science, Hokkaido University, Sapporo, Japan
| | - Tokuko Haraguchi
- Graduate School of Frontier Biosciences, Osaka University, Suita, Japan.,Advanced ICT Research Institute Kobe, National Institute of Information and Communications Technology, Kobe, Japan
| | - Yasushi Hiraoka
- Graduate School of Frontier Biosciences, Osaka University, Suita, Japan.,Advanced ICT Research Institute Kobe, National Institute of Information and Communications Technology, Kobe, Japan
| |
Collapse
|
47
|
Elkhatib RA, Paci M, Boissier R, Longepied G, Auguste Y, Achard V, Bourgeois P, Levy N, Branger N, Mitchell MJ, Metzler-Guillemain C. LEM-domain proteins are lost during human spermiogenesis but BAF and BAF-L persist. Reproduction 2017; 154:387-401. [PMID: 28684548 DOI: 10.1530/rep-17-0358] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Revised: 06/10/2017] [Accepted: 07/06/2017] [Indexed: 12/22/2022]
Abstract
During spermiogenesis the spermatid nucleus is elongated, and dramatically reduced in size with protamines replacing histones to produce a highly compacted chromatin. After fertilisation, this process is reversed in the oocyte to form the male pronucleus. Emerging evidence, including the coordinated loss of the nuclear lamina (NL) and the histones, supports the involvement of the NL in spermatid nuclear remodelling, but how the NL links to the chromatin is not known. In somatic cells, interactions between the NL and the chromatin have been demonstrated: LEM-domain proteins and LBR interact with the NL and respectively, the chromatin proteins BAF and HP1. We therefore sought to characterise the lamina-chromatin interface during spermiogenesis, by investigating the localisation of six LEM-domain proteins, two BAF proteins and LBR, in human spermatids and spermatozoa. Using RT-PCR, IF and western blotting, we show that six of the proteins tested are present in spermatids: LEMD1, LEMD2 (a short isoform), ANKLE2, LAP2β, BAF and BAF-L, and three absent: Emerin, LBR and LEMD3. The full-length LEMD2 isoform, required for nuclear integrity in somatic cells, is absent. In spermatids, no protein localised to the nuclear periphery, but five were nucleoplasmic, receding towards the posterior nuclear pole as spermatids matured. Our study therefore establishes that the lamina-chromatin interface in human spermatids is radically distinct from that defined in somatic cells. In ejaculated spermatozoa, we detected only BAF and BAF-L, suggesting that they might contribute to the shaping of the spermatozoon nucleus and, after fertilisation, its transition to the male pronucleus.
Collapse
Affiliation(s)
| | - Marine Paci
- Aix Marseille UnivINSERM, GMGF, UMR_S 910, Marseille, France
- APHM Hôpital La ConceptionGynépôle, Laboratoire de Biologie de la Reproduction-CECOS, Marseille Cedex 5, France
| | - Romain Boissier
- APHM Hôpital La ConceptionService d'Urologie, Marseille Cedex 5, France
| | - Guy Longepied
- Aix Marseille UnivINSERM, GMGF, UMR_S 910, Marseille, France
| | - Yasmina Auguste
- Aix Marseille UnivINSERM, GMGF, UMR_S 910, Marseille, France
| | - Vincent Achard
- APHM Hôpital La ConceptionGynépôle, Laboratoire de Biologie de la Reproduction-CECOS, Marseille Cedex 5, France
- Aix-Marseille UnivUniv Avignon, CNRS, IRD, IMBE, UMR7263, Marseille France
| | | | - Nicolas Levy
- Aix Marseille UnivINSERM, GMGF, UMR_S 910, Marseille, France
| | - Nicolas Branger
- APHM Hôpital La ConceptionService d'Urologie, Marseille Cedex 5, France
| | | | - Catherine Metzler-Guillemain
- Aix Marseille UnivINSERM, GMGF, UMR_S 910, Marseille, France
- APHM Hôpital La ConceptionGynépôle, Laboratoire de Biologie de la Reproduction-CECOS, Marseille Cedex 5, France
| |
Collapse
|
48
|
Gagliardi A, Besio R, Carnemolla C, Landi C, Armini A, Aglan M, Otaify G, Temtamy SA, Forlino A, Bini L, Bianchi L. Cytoskeleton and nuclear lamina affection in recessive osteogenesis imperfecta: A functional proteomics perspective. J Proteomics 2017; 167:46-59. [PMID: 28802583 PMCID: PMC5584732 DOI: 10.1016/j.jprot.2017.08.007] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Revised: 08/02/2017] [Accepted: 08/07/2017] [Indexed: 02/07/2023]
Abstract
Osteogenesis imperfecta (OI) is a collagen-related disorder associated to dominant, recessive or X-linked transmission, mainly caused by mutations in type I collagen genes or in genes involved in type I collagen metabolism. Among the recessive forms, OI types VII, VIII, and IX are due to mutations in CRTAP, P3H1, and PPIB genes, respectively. They code for the three components of the endoplasmic reticulum complex that catalyzes 3-hydroxylation of type I collagen α1Pro986. Under-hydroxylation of this residue leads to collagen structural abnormalities and results in moderate to lethal OI phenotype, despite the exact molecular mechanisms are still not completely clear. To shed light on these recessive forms, primary fibroblasts from OI patients with mutations in CRTAP (n=3), P3H1 (n=3), PPIB (n=1) genes and from controls (n=4) were investigated by a functional proteomic approach. Cytoskeleton and nucleoskeleton asset, protein fate, and metabolism were delineated as mainly affected. While western blot experiments confirmed altered expression of lamin A/C and cofilin-1, immunofluorescence analysis using antibody against lamin A/C and phalloidin showed an aberrant organization of nucleus and cytoskeleton. This is the first report describing an altered organization of intracellular structural proteins in recessive OI and pointing them as possible novel target for OI treatment. SIGNIFICANCE OI is a prototype for skeletal dysplasias. It is a highly heterogeneous collagen-related disorder with dominant, recessive and X-linked transmission. There is no definitive cure for this disease, thus a better understanding of the molecular basis of its pathophysiology is expected to contribute in identifying potential targets to develop new treatments. Based on this concept, we performed a functional proteomic study to delineate affected molecular pathways in primary fibroblasts from recessive OI patients, carrying mutations in CRTAP (OI type VII), P3H1 (OI type VIII), and PPIB (OI type IX) genes. Our analyses demonstrated the occurrence of an altered cytoskeleton and, for the first time in OI, of nuclear lamina organization. Hence, cytoskeleton and nucleoskeleton components may be considered as novel drug targets for clinical management of the disease. Finally, according to our analyses, OI emerged to share similar deregulated pathways and molecular aberrances, as previously described, with other rare disorders caused by different genetic defects. Those aberrances may provide common pharmacological targets to support classical clinical approach in treating different diseases.
Collapse
Affiliation(s)
- Assunta Gagliardi
- Functional Proteomics Laboratory, Department of Life Sciences, University of Siena, Siena, Italy; CIBIO, University of Trento, Trento, Italy
| | - Roberta Besio
- Department of Molecular Medicine, Biochemistry Unit, University of Pavia, Pavia, Italy
| | - Chiara Carnemolla
- Functional Proteomics Laboratory, Department of Life Sciences, University of Siena, Siena, Italy
| | - Claudia Landi
- Functional Proteomics Laboratory, Department of Life Sciences, University of Siena, Siena, Italy
| | - Alessandro Armini
- Functional Proteomics Laboratory, Department of Life Sciences, University of Siena, Siena, Italy
| | - Mona Aglan
- Department of Clinical Genetics, Human Genetics & Genome Research Division, Center of Excellence for Human Genetics, National Research Centre, Cairo, Egypt
| | - Ghada Otaify
- Department of Clinical Genetics, Human Genetics & Genome Research Division, Center of Excellence for Human Genetics, National Research Centre, Cairo, Egypt
| | - Samia A Temtamy
- Department of Clinical Genetics, Human Genetics & Genome Research Division, Center of Excellence for Human Genetics, National Research Centre, Cairo, Egypt
| | - Antonella Forlino
- Department of Molecular Medicine, Biochemistry Unit, University of Pavia, Pavia, Italy
| | - Luca Bini
- Functional Proteomics Laboratory, Department of Life Sciences, University of Siena, Siena, Italy
| | - Laura Bianchi
- Functional Proteomics Laboratory, Department of Life Sciences, University of Siena, Siena, Italy.
| |
Collapse
|
49
|
Bermeo S, Al-Saedi A, Kassem M, Vidal C, Duque G. The Role of the Nuclear Envelope Protein MAN1 in Mesenchymal Stem Cell Differentiation. J Cell Biochem 2017; 118:4425-4435. [PMID: 28449239 DOI: 10.1002/jcb.26096] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Accepted: 04/26/2017] [Indexed: 01/09/2023]
Abstract
Mutations in MAN1, a protein of the nuclear envelope, cause bone phenotypes characterized by hyperostosis. The mechanism of this pro-osteogenic phenotype remains unknown. We increased and decreased MAN1 expression in mesenchymal stem cells (MSC) upon which standard osteogenic and adipogenic differentiation were performed. MAN1 knockdown increased osteogenesis and mineralization. In contrast, osteogenesis remained stable upon MAN1 overexpression. Regarding a mechanism, we found that low levels of MAN1 facilitated the nuclear accumulation of regulatory smads and smads-related complexes, with a concurrently high expression of nuclear β-Catenin. In addition, we found adipogenesis to be decreased in both conditions, although predominantly affected by MAN1 overexpression. Finally, lamin A, a protein of the nuclear envelope that regulates MSC differentiation, was unaffected by changes in MAN1. In conclusion, our studies demonstrated that lower levels of MAN1 in differentiating MSC are associated with higher osteogenesis and lower adipogenesis. High levels of MAN1 only affected adipogenesis. These effects could have an important role in the understanding of the role of the proteins of the nuclear envelope in bone formation. J. Cell. Biochem. 118: 4425-4435, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Sandra Bermeo
- Sydney Medical School Nepean, The University of Sydney, Penrith, NSW, Australia.,Facultad de Ciencias Básicas y Biomédicas, Universidad Simón Bolívar, Barranquilla, Colombia
| | - Ahmed Al-Saedi
- Australian Institute for Musculoskeletal Science (AIMSS), The University of Melbourne and Western Health, Melbourne, VIC, Australia.,Department of Medicine-Western Health, Melbourne Medical School, The University of Melbourne, Melbourne, VIC, Australia
| | - Moustapha Kassem
- Department of Endocrinology and Metabolism, Odense University Hospital & University of Southern Denmark, J.B. Odense C, Denmark
| | - Christopher Vidal
- Sydney Medical School Nepean, The University of Sydney, Penrith, NSW, Australia
| | - Gustavo Duque
- Australian Institute for Musculoskeletal Science (AIMSS), The University of Melbourne and Western Health, Melbourne, VIC, Australia.,Department of Medicine-Western Health, Melbourne Medical School, The University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
50
|
Abstract
Nuclear lamins are components of the peripheral lamina that define the mechanical properties of nuclei and tether heterochromatin to the periphery. A-type lamins localize also to the nuclear interior, but the regulation and specific functions of this nucleoplasmic lamin pool are poorly understood. In this Commentary, we summarize known pathways that are potentially involved in the localization and dynamic behavior of intranuclear lamins, including their post-translational modifications and interactions with nucleoplasmic proteins, such as lamina-associated polypeptide 2α (LAP2α; encoded by TMPO). In addition, new data suggest that lamins in the nuclear interior have an important role in chromatin regulation and gene expression through dynamic binding to both hetero- and euchromatic genomic regions and promoter subdomains, thereby affecting epigenetic pathways and chromatin accessibility. Nucleoplasmic lamins also have a role in spatial chromatin organization and may be involved in mechanosignaling. In view of this newly emerging concept, we propose that the previously reported cellular phenotypes in lamin-linked diseases are, at least in part, rooted in an impaired regulation and/or function of the nucleoplasmic lamin A/C pool.
Collapse
Affiliation(s)
- Nana Naetar
- Center of Medical Biochemistry, Max F. Perutz Laboratories (MFPL), Medical University of Vienna, Vienna Biocenter (VBC), Dr.-Bohr-Gasse 9, Vienna A-1030, Austria
| | - Simona Ferraioli
- Center of Medical Biochemistry, Max F. Perutz Laboratories (MFPL), Medical University of Vienna, Vienna Biocenter (VBC), Dr.-Bohr-Gasse 9, Vienna A-1030, Austria
| | - Roland Foisner
- Center of Medical Biochemistry, Max F. Perutz Laboratories (MFPL), Medical University of Vienna, Vienna Biocenter (VBC), Dr.-Bohr-Gasse 9, Vienna A-1030, Austria
| |
Collapse
|