1
|
Nair A, Khanna J, Kler J, Ragesh R, Sengupta K. Nuclear envelope and chromatin choreography direct cellular differentiation. Nucleus 2025; 16:2449520. [PMID: 39943681 PMCID: PMC11834525 DOI: 10.1080/19491034.2024.2449520] [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: 10/01/2024] [Revised: 12/28/2024] [Accepted: 12/30/2024] [Indexed: 02/20/2025] Open
Abstract
The nuclear envelope plays an indispensable role in the spatiotemporal organization of chromatin and transcriptional regulation during the intricate process of cell differentiation. This review outlines the distinct regulatory networks between nuclear envelope proteins, transcription factors and epigenetic modifications in controlling the expression of cell lineage-specific genes during differentiation. Nuclear lamina with its associated nuclear envelope proteins organize heterochromatin via Lamina-Associated Domains (LADs), proximal to the nuclear periphery. Since nuclear lamina is mechanosensitive, we critically examine the impact of extracellular forces on differentiation outcomes. The nuclear envelope is spanned by nuclear pore complexes which, in addition to their central role in transport, are associated with chromatin organization. Furthermore, mutations in the nuclear envelope proteins disrupt differentiation, resulting in developmental disorders. Investigating the underlying nuclear envelope controlled regulatory mechanisms of chromatin remodelling during lineage commitment will accelerate our fundamental understanding of developmental biology and regenerative medicine.
Collapse
Affiliation(s)
- Anjitha Nair
- Chromosome Biology Lab (CBL), Indian Institute of Science Education and Research (IISER) Pune, Maharashtra, India
| | - Jayati Khanna
- Chromosome Biology Lab (CBL), Indian Institute of Science Education and Research (IISER) Pune, Maharashtra, India
| | - Jashan Kler
- Chromosome Biology Lab (CBL), Indian Institute of Science Education and Research (IISER) Pune, Maharashtra, India
| | - Rohith Ragesh
- Chromosome Biology Lab (CBL), Indian Institute of Science Education and Research (IISER) Pune, Maharashtra, India
| | - Kundan Sengupta
- Chromosome Biology Lab (CBL), Indian Institute of Science Education and Research (IISER) Pune, Maharashtra, India
| |
Collapse
|
2
|
Huerta-Padilla V, Marrero-Rodríguez D, Taniguchi-Ponciano K, López AE, Candanedo-González F, Salcedo E, Valdivia-Flores A, Rodriguez-Esquivel M, Virgilio LG, López-Romero R, Nambo-Lucio MDJ, Meza-Toledo SE, Bandala C, Meraz MA, Salcedo M. Thymopoietin- α, - β, and - γ Isoforms Increased Expression in Cervical Cancer Cells. THE CANADIAN JOURNAL OF INFECTIOUS DISEASES & MEDICAL MICROBIOLOGY = JOURNAL CANADIEN DES MALADIES INFECTIEUSES ET DE LA MICROBIOLOGIE MEDICALE 2025; 2025:1668482. [PMID: 40242184 PMCID: PMC12003041 DOI: 10.1155/cjid/1668482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 11/20/2024] [Indexed: 04/18/2025]
Abstract
Cervical cancer (CC) is a public health concern related to the human papillomavirus (HPV) persistent infection. Minichromosome maintenance 2 (MCM2) has been postulated as a surrogate marker for HPV infection. Thymopoietin (TMPO) is a nuclear protein regulated by E2F such as MCM2 or p16. TMPO can give rise to six different isoforms. Herein, both the mRNA and protein levels of TMPO isoforms were analyzed in cervical cells. TMPO expression was selected and analyzed through in silico in several databases from the healthy cervix and cervical lesions. TMPO RNA expression was evaluated in cervical samples and cell lines by RT-PCR and protein expression by Western-blot and immunohistochemistry assays. TMPO and MCM2 immunostaining were evaluated in cervical smears. The clinical-pathological correlation analysis was performed using Kruskal-Wallis or Χ 2 tests. TMPO is overexpressed in 74% of CC cells and all CC cell lines. Moreover, negative immunostaining was observed in normal cervical tissue, compared to strong expression for cervical lesions. Interestingly, TMPO-α, -β, -δ, -ε, and -γ are expressed in all cervical cells and tissues, but a differential expression for α, -β, and -γ isoforms among the cervical cells was observed as overexpressed when HPV is present. Also, the immunostaining of both MCM2 and TMPO was quite similar, but TMPO expression was more sensitive and specific than MCM2 protein. The present study has revealed that TMPO protein expression could be a potential molecular marker for cervical transformed cells, highlighting the TMPO-α, -β, and -γ isoforms as a promising molecular marker of HPV infection.
Collapse
Affiliation(s)
- Víctor Huerta-Padilla
- Oncology Genomics Biomedical Research Unit, Gynecology Pediatrics Hospital 3A, North Unity OOAD, Mexican Institute of Social Security, Mexico City, Mexico
- Departamento de Bioquimica, Laboratorio de Quimioterapia Experimental, Escuela Nacional de Ciencias Biológicas, Instituto Politecnico Nacional, Mexico City, Mexico
| | - Daniel Marrero-Rodríguez
- Endocrine Diseases Research Unit, Specialties Hospital, National Medical Center SXXI, Mexican Institute of Social Security, Mexico City, Mexico
| | - Keiko Taniguchi-Ponciano
- Endocrine Diseases Research Unit, Specialties Hospital, National Medical Center SXXI, Mexican Institute of Social Security, Mexico City, Mexico
| | - Ariana E. López
- Oncology Genomics Biomedical Research Unit, Gynecology Pediatrics Hospital 3A, North Unity OOAD, Mexican Institute of Social Security, Mexico City, Mexico
| | - Fernando Candanedo-González
- Anatomo-Pathology Service, Oncology Hospital, National Medical Center SXXI, Mexican Institute of Social Security, Mexico City, Mexico
| | - Emmanuel Salcedo
- Oncology Genomics Biomedical Research Unit, Gynecology Pediatrics Hospital 3A, North Unity OOAD, Mexican Institute of Social Security, Mexico City, Mexico
| | | | - Miriam Rodriguez-Esquivel
- Oncology Genomics Biomedical Research Unit, Gynecology Pediatrics Hospital 3A, North Unity OOAD, Mexican Institute of Social Security, Mexico City, Mexico
| | - Laura Gómez Virgilio
- Centro de Investigación y de Estudios Avanzados, Molecular Biomedicine Department, Instituto Politecnico Nacional, Mexico City, Mexico
| | - Ricardo López-Romero
- Oncology Genomics Biomedical Research Unit, Gynecology Pediatrics Hospital 3A, North Unity OOAD, Mexican Institute of Social Security, Mexico City, Mexico
| | | | - Sergio E. Meza-Toledo
- Departamento de Bioquimica, Laboratorio de Quimioterapia Experimental, Escuela Nacional de Ciencias Biológicas, Instituto Politecnico Nacional, Mexico City, Mexico
| | - Cindy Bandala
- Departamento de Medicina Traslacional aplicada a Neurociencias, Escuela Superior de Medicina, Instituto Politecnico Nacional, Mexico City, Mexico
| | - Marco A. Meraz
- Centro de Investigación y de Estudios Avanzados, Molecular Biomedicine Department, Instituto Politecnico Nacional, Mexico City, Mexico
| | - Mauricio Salcedo
- Oncology Genomics Biomedical Research Unit, Gynecology Pediatrics Hospital 3A, North Unity OOAD, Mexican Institute of Social Security, Mexico City, Mexico
| |
Collapse
|
3
|
Chen W, Byun J, Kang HC, Lee HS, Lee JY, Kwon YJ, Cho YY. Karyoptosis as a novel type of UVB-induced regulated cell death. Free Radic Res 2024; 58:796-810. [PMID: 39625813 DOI: 10.1080/10715762.2024.2433986] [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: 08/01/2024] [Revised: 11/14/2024] [Accepted: 11/20/2024] [Indexed: 12/28/2024]
Abstract
Karyoptosis is a type of regulated cell death (RCD) characterized by explosive nuclear rupture caused by a loss of nuclear membrane integrity, resulting in the release of genomic DNA and other nuclear components into the cytosol and extracellular environment. The mechanism underlying karyoptosis involves a delicate balance between the following forces: the expansion force exerted by the tightly packed DNA in the nucleus, the resistance provided by the nuclear lamina at the inner nuclear membrane (INM), and the tensile force from the cytoskeleton that helps position the nucleus at the center of the cytoplasm, allowing it to remain maximally expanded. In addition, CREB3, a type II integral membrane protein with DNA-binding ability, tethers chromatin to the INM, providing a tightening force through chromatin interactions that prevent nuclear membrane rupture. UVB radiation can trigger this process, inducing CREB3-FL cleavage and producing CREB3-CF. Therefore, UVB acts as an intrinsic factor in the induction of karyoptosis. Importantly, biochemical analysis of RCD markers shows that karyoptosis is distinct from other forms of cell death, such as apoptosis, autophagy, necroptosis, and pyroptosis. This review explores the mechanisms involved in maintaining nuclear membrane integrity and the role of CREB3 in triggering karyoptosis and provides brief suggestions on the potential implications for targeting cancer cells.
Collapse
Affiliation(s)
- Weidong Chen
- BK21-Four, College of Pharmacy, The Catholic University of Korea, Bucheon-si, Gyeonggi-do, South Korea
| | - Jiin Byun
- BK21-Four, College of Pharmacy, The Catholic University of Korea, Bucheon-si, Gyeonggi-do, South Korea
| | - Han Chang Kang
- College of Pharmacy, The Catholic University of Korea, Bucheon-si, Gyeonggi-do, South Korea
| | - Hye Suk Lee
- BK21-Four, College of Pharmacy, The Catholic University of Korea, Bucheon-si, Gyeonggi-do, South Korea
| | - Joo Young Lee
- BK21-Four, College of Pharmacy, The Catholic University of Korea, Bucheon-si, Gyeonggi-do, South Korea
| | - Young Jik Kwon
- College of Pharmacy, The Catholic University of Korea, Bucheon-si, Gyeonggi-do, South Korea
- Department of Pharmaceutical Sciences, University of California, Irvine, CA, USA
| | - Yong-Yeon Cho
- BK21-Four, College of Pharmacy, The Catholic University of Korea, Bucheon-si, Gyeonggi-do, South Korea
| |
Collapse
|
4
|
Wang B, Kou H, Wang Y, Zhang Q, Jiang D, Wang J, Zhao Z, Zhou Y, Zhang M, Sui L, Zhao M, Liu Y, Liu Y, Shi L, Wang F. LAP2α orchestrates alternative lengthening of telomeres suppression through telomeric heterochromatin regulation with HDAC1: unveiling a potential therapeutic target. Cell Death Dis 2024; 15:761. [PMID: 39426946 PMCID: PMC11490576 DOI: 10.1038/s41419-024-07116-4] [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: 02/01/2024] [Revised: 09/23/2024] [Accepted: 09/26/2024] [Indexed: 10/21/2024]
Abstract
In response to the challenge of telomere attrition during DNA replication, cancer cells predominantly employ telomerase or, in 10-15% of cases, the alternative lengthening of telomeres (ALT). The intricate details of ALT, however, remain elusive. In this study, we unveil that the knockdown of lamina-associated polypeptide 2 alpha (LAP2α) in ALT cells results in telomere dysfunction, triggering a notable increase in ALT-associated hallmarks, including high frequencies of PML bodies (APBs), C-rich extrachromosomal circles (C-circles), and telomere sister chromatid exchange (T-SCE). Furthermore, LAP2α emerges as a crucial player in break-induced telomere replication for telomerase-positive cells following telomeric double-strand breaks. Mechanistically, our investigation suggests that LAP2α may influence the regulation of the heterochromatic state of telomeres, thereby affecting telomeric accessibility. In line with our findings, LAP2α expression is markedly reduced in ALT-positive osteosarcoma. And the use of methotrexate (MTX) can restore the heterochromatin state altered by LAP2α depletion. This is evidenced by a significant inhibition of tumor proliferation in ALT-positive patient-derived xenograft (PDX) mouse models. These results indicate the important role of LAP2α in regulating ALT activity and offer insights into the interplay between lamina-associated proteins and telomeres in maintaining telomere length. Importantly, our findings may help identify a more appropriate target population for the osteosarcoma therapeutic drug, MTX.
Collapse
Grants
- 32170762 National Natural Science Foundation of China (National Science Foundation of China)
- This work was supported by the grant from the National Natural Science Foundation of China (No. 32170762, 3217050514, 31771520, 31471293, 91649102, 92149302, 81772243, 81771135, 81970958, 82303619), Tianjin Health Research Project (No. 19YFZCSY00600), Science and Technology Project of Tianjin Municipal Health Committee (No. TJWJ2022XK018, TJWJ2022QN030) and the Natural Science Foundation of Tianjin City (No. 19JCJQJC63500)
Collapse
Affiliation(s)
- Bing Wang
- The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Department of Genetics, School of Basic Medical Science, Institute of Prosthodontics School and Hospital of Stomatology, General Hospital, Tianjin Medical University, 300070, Tianjin, P. R. China
- Department of Hematology, Tianjin First Central Hospital, 300192, Tianjin, P. R. China
| | - Haomeng Kou
- The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Department of Genetics, School of Basic Medical Science, Institute of Prosthodontics School and Hospital of Stomatology, General Hospital, Tianjin Medical University, 300070, Tianjin, P. R. China
| | - Yuwen Wang
- The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Department of Genetics, School of Basic Medical Science, Institute of Prosthodontics School and Hospital of Stomatology, General Hospital, Tianjin Medical University, 300070, Tianjin, P. R. China
| | - Qi Zhang
- Department of Clinical Laboratory, First Affiliated Hospital of Xi'an Jiaotong University, 710061, Xi'an, Shaanxi, P. R. China
| | - Duo Jiang
- The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Department of Genetics, School of Basic Medical Science, Institute of Prosthodontics School and Hospital of Stomatology, General Hospital, Tianjin Medical University, 300070, Tianjin, P. R. China
| | - Juan Wang
- The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Department of Genetics, School of Basic Medical Science, Institute of Prosthodontics School and Hospital of Stomatology, General Hospital, Tianjin Medical University, 300070, Tianjin, P. R. China
| | - Ziqin Zhao
- Department of Pathology, Tianjin Hospital, 300221, Tianjin, P. R. China
| | - Yao Zhou
- Department of Bioinformatics, The Province and Ministry Co-Sponsored Collaborative Innovation Center for Medical Epigenetics, School of Basic Medical Sciences, Tianjin Medical University, 300070, Tianjin, P. R. China
| | - Miaomiao Zhang
- Department of Pathology, Jining No.1 People's Hospital, 272000, Jining, Shandong, P. R. China
| | - Lei Sui
- Department of Prosthodontics, School and Hospital of Stomatology, Tianjin Medical University, 300070, Tianjin, P. R. China
| | - Mingfeng Zhao
- Department of Hematology, Tianjin First Central Hospital, 300192, Tianjin, P. R. China
| | - Yancheng Liu
- Department of Bone and Soft Tissue Oncology, Tianjin Hospital, 300221, Tianjin, P. R. China.
| | - Yang Liu
- Department of Radiobiology, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, 300192, Tianjin, P. R. China.
| | - Lei Shi
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, 300070, Tianjin, P. R. China.
| | - Feng Wang
- The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Department of Genetics, School of Basic Medical Science, Institute of Prosthodontics School and Hospital of Stomatology, General Hospital, Tianjin Medical University, 300070, Tianjin, P. R. China.
| |
Collapse
|
5
|
Popęda M, Kowalski K, Wenta T, Beznoussenko GV, Rychłowski M, Mironov A, Lavagnino Z, Barozzi S, Richert J, Bertolio R, Myszczyński K, Szade J, Bieńkowski M, Miszewski K, Matuszewski M, Żaczek AJ, Braga L, Del Sal G, Bednarz-Knoll N, Maiuri P, Nastały P. Emerin mislocalization during chromatin bridge resolution can drive prostate cancer cell invasiveness in a collagen-rich microenvironment. Exp Mol Med 2024; 56:2016-2032. [PMID: 39218980 PMCID: PMC11446916 DOI: 10.1038/s12276-024-01308-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 06/14/2024] [Accepted: 06/17/2024] [Indexed: 09/04/2024] Open
Abstract
Micronuclei (MN) can form through many mechanisms, including the breakage of aberrant cytokinetic chromatin bridges. The frequent observation of MN in tumors suggests that they might not merely be passive elements but could instead play active roles in tumor progression. Here, we propose a mechanism through which the presence of micronuclei could induce specific phenotypic and functional changes in cells and increase the invasive potential of cancer cells. Through the integration of diverse in vitro imaging and molecular techniques supported by clinical samples from patients with prostate cancer (PCa) defined as high-risk by the D'Amico classification, we demonstrate that the resolution of chromosome bridges can result in the accumulation of Emerin and the formation of Emerin-rich MN. These structures are negative for Lamin A/C and positive for the Lamin-B receptor and Sec61β. MN can act as a protein sinks and result in the pauperization of Emerin from the nuclear envelope. The Emerin mislocalization phenotype is associated with a molecular signature that is correlated with a poor prognosis in PCa patients and is enriched in metastatic samples. Emerin mislocalization corresponds with increases in the migratory and invasive potential of tumor cells, especially in a collagen-rich microenvironment. Our study demonstrates that the mislocalization of Emerin to MN results in increased cell invasiveness, thereby worsening patient prognosis.
Collapse
Affiliation(s)
- Marta Popęda
- Division of Translational Oncology, Intercollegiate Faculty of Biotechnology, University of Gdańsk and Medical University of Gdańsk, Gdańsk, Poland
- Department of Pathomorphology, Medical University of Gdańsk, Gdańsk, Poland
| | - Kamil Kowalski
- Division of Translational Oncology, Intercollegiate Faculty of Biotechnology, University of Gdańsk and Medical University of Gdańsk, Gdańsk, Poland
| | - Tomasz Wenta
- Department of General and Medical Biochemistry, Faculty of Biology, University of Gdansk, Gdansk, Poland
| | | | - Michał Rychłowski
- Laboratory of Virus Molecular Biology, Intercollegiate Faculty of Biotechnology, University of Gdansk and Medical University of Gdansk, Gdansk, Poland
| | | | - Zeno Lavagnino
- IFOM ETS-The AIRC Institute of Molecular Oncology, Milan, Italy
| | - Sara Barozzi
- IFOM ETS-The AIRC Institute of Molecular Oncology, Milan, Italy
| | - Julia Richert
- Division of Translational Oncology, Intercollegiate Faculty of Biotechnology, University of Gdańsk and Medical University of Gdańsk, Gdańsk, Poland
| | - Rebecca Bertolio
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Area Science Park-Padriciano, Trieste, Italy
| | - Kamil Myszczyński
- Centre of Biostatistics and Bioinformatics Analysis, Medical University of Gdansk, Gdansk, Poland
| | - Jolanta Szade
- Department of Pathomorphology, Medical University of Gdańsk, Gdańsk, Poland
| | - Michał Bieńkowski
- Department of Pathomorphology, Medical University of Gdańsk, Gdańsk, Poland
| | - Kevin Miszewski
- Department of Urology, Medical University of Gdańsk, Gdańsk, Poland
| | | | - Anna J Żaczek
- Division of Translational Oncology, Intercollegiate Faculty of Biotechnology, University of Gdańsk and Medical University of Gdańsk, Gdańsk, Poland
| | - Luca Braga
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Area Science Park-Padriciano, Trieste, Italy
| | - Giannino Del Sal
- IFOM ETS-The AIRC Institute of Molecular Oncology, Milan, Italy
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Area Science Park-Padriciano, Trieste, Italy
- Department of Life Sciences, University of Trieste, Trieste, Italy
| | - Natalia Bednarz-Knoll
- Division of Translational Oncology, Intercollegiate Faculty of Biotechnology, University of Gdańsk and Medical University of Gdańsk, Gdańsk, Poland
| | - Paolo Maiuri
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy
| | - Paulina Nastały
- Division of Translational Oncology, Intercollegiate Faculty of Biotechnology, University of Gdańsk and Medical University of Gdańsk, Gdańsk, Poland.
| |
Collapse
|
6
|
Aryee DNT, Fock V, Kapoor U, Radic-Sarikas B, Kovar H. Zooming in on Long Non-Coding RNAs in Ewing Sarcoma Pathogenesis. Cells 2022; 11:1267. [PMID: 35455947 PMCID: PMC9032025 DOI: 10.3390/cells11081267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 03/29/2022] [Accepted: 04/06/2022] [Indexed: 11/16/2022] Open
Abstract
Ewing sarcoma (ES) is a rare aggressive cancer of bone and soft tissue that is mainly characterized by a reciprocal chromosomal translocation. As a result, about 90% of cases express the EWS-FLI1 fusion protein that has been shown to function as an aberrant transcription factor driving sarcomagenesis. ES is the second most common malignant bone tumor in children and young adults. Current treatment modalities include dose-intensified chemo- and radiotherapy, as well as surgery. Despite these strategies, patients who present with metastasis or relapse still have dismal prognosis, warranting a better understanding of treatment resistant-disease biology in order to generate better prognostic and therapeutic tools. Since the genomes of ES tumors are relatively quiet and stable, exploring the contributions of epigenetic mechanisms in the initiation and progression of the disease becomes inevitable. The search for novel biomarkers and potential therapeutic targets of cancer metastasis and chemotherapeutic drug resistance is increasingly focusing on long non-coding RNAs (lncRNAs). Recent advances in genome analysis by high throughput sequencing have immensely expanded and advanced our knowledge of lncRNAs. They are non-protein coding RNA species with multiple biological functions that have been shown to be dysregulated in many diseases and are emerging as crucial players in cancer development. Understanding the various roles of lncRNAs in tumorigenesis and metastasis would determine eclectic avenues to establish therapeutic and diagnostic targets. In ES, some lncRNAs have been implicated in cell proliferation, migration and invasion, features that make them suitable as relevant biomarkers and therapeutic targets. In this review, we comprehensively discuss known lncRNAs implicated in ES that could serve as potential biomarkers and therapeutic targets of the disease. Though some current reviews have discussed non-coding RNAs in ES, to our knowledge, this is the first review focusing exclusively on ES-associated lncRNAs.
Collapse
Affiliation(s)
- Dave N T Aryee
- St. Anna Children's Cancer Research Institute, 1090 Vienna, Austria
- Department of Pediatrics, Medical University of Vienna, 1090 Vienna, Austria
| | - Valerie Fock
- St. Anna Children's Cancer Research Institute, 1090 Vienna, Austria
| | - Utkarsh Kapoor
- St. Anna Children's Cancer Research Institute, 1090 Vienna, Austria
| | - Branka Radic-Sarikas
- St. Anna Children's Cancer Research Institute, 1090 Vienna, Austria
- Department of Pediatric Surgery, Medical University of Vienna, 1090 Vienna, Austria
| | - Heinrich Kovar
- St. Anna Children's Cancer Research Institute, 1090 Vienna, Austria
- Department of Pediatrics, Medical University of Vienna, 1090 Vienna, Austria
| |
Collapse
|
7
|
Bao K, Zhang Q, Liu S, Song N, Guo Q, Liu L, Tian S, Hao J, Zhu Y, Zhang K, Ai D, Yang J, Yao Z, Foisner R, Shi L. LAP2α preserves genome integrity through assisting RPA deposition on damaged chromatin. Genome Biol 2022; 23:64. [PMID: 35227284 PMCID: PMC8883701 DOI: 10.1186/s13059-022-02638-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Accepted: 02/17/2022] [Indexed: 11/22/2022] Open
Abstract
BACKGROUND Single-stranded DNA (ssDNA) coated with replication protein A (RPA) acts as a key platform for the recruitment and exchange of genome maintenance factors in DNA damage response. Yet, how the formation of the ssDNA-RPA intermediate is regulated remains elusive. RESULTS Here, we report that the lamin-associated protein LAP2α is physically associated with RPA, and LAP2α preferentially facilitates RPA deposition on damaged chromatin via physical contacts between LAP2α and RPA1. Importantly, LAP2α-promoted RPA binding to ssDNA plays a critical role in protection of replication forks, activation of ATR, and repair of damaged DNA. We further demonstrate that the preference of LAP2α-promoted RPA loading on damaged chromatin depends on poly ADP-ribose polymerase PARP1, but not poly(ADP-ribosyl)ation. CONCLUSIONS Our study provides mechanistic insight into RPA deposition in response to DNA damage and reveals a genome protection role of LAP2α.
Collapse
Affiliation(s)
- Kaiwen Bao
- State Key Laboratory of Experimental Hematology, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Key Laboratory of Breast Cancer Prevention and Therapy (Ministry of Education), Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University General Hospital, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Qi Zhang
- Department of Clinical Laboratory, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Shuai Liu
- State Key Laboratory of Experimental Hematology, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Key Laboratory of Breast Cancer Prevention and Therapy (Ministry of Education), Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University General Hospital, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Nan Song
- State Key Laboratory of Experimental Hematology, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Key Laboratory of Breast Cancer Prevention and Therapy (Ministry of Education), Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University General Hospital, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Qiushi Guo
- State Key Laboratory of Experimental Hematology, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Key Laboratory of Breast Cancer Prevention and Therapy (Ministry of Education), Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University General Hospital, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Ling Liu
- State Key Laboratory of Experimental Hematology, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Key Laboratory of Breast Cancer Prevention and Therapy (Ministry of Education), Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University General Hospital, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Shanshan Tian
- State Key Laboratory of Experimental Hematology, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Key Laboratory of Breast Cancer Prevention and Therapy (Ministry of Education), Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University General Hospital, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Jihui Hao
- State Key Laboratory of Experimental Hematology, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Key Laboratory of Breast Cancer Prevention and Therapy (Ministry of Education), Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University General Hospital, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Yi Zhu
- State Key Laboratory of Experimental Hematology, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Key Laboratory of Breast Cancer Prevention and Therapy (Ministry of Education), Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University General Hospital, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Kai Zhang
- State Key Laboratory of Experimental Hematology, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Key Laboratory of Breast Cancer Prevention and Therapy (Ministry of Education), Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University General Hospital, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Ding Ai
- State Key Laboratory of Experimental Hematology, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Key Laboratory of Breast Cancer Prevention and Therapy (Ministry of Education), Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University General Hospital, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Jie Yang
- State Key Laboratory of Experimental Hematology, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Key Laboratory of Breast Cancer Prevention and Therapy (Ministry of Education), Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University General Hospital, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Zhi Yao
- State Key Laboratory of Experimental Hematology, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Key Laboratory of Breast Cancer Prevention and Therapy (Ministry of Education), Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University General Hospital, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Roland Foisner
- Max Perutz Laboratories, Center of Medical Biochemistry, Medical University of Vienna, Vienna Biocenter (VBC), Vienna, Austria
| | - Lei Shi
- State Key Laboratory of Experimental Hematology, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Key Laboratory of Breast Cancer Prevention and Therapy (Ministry of Education), Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University General Hospital, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China.
- Department of Biochemistry and Molecular Biology, Tianjin Medical University, 22 Qixiangtai Road, Tianjin, 300070, China.
| |
Collapse
|
8
|
Tang Y, Dong Q, Wang T, Gong L, Gu Y. PNET2 is a component of the plant nuclear lamina and is required for proper genome organization and activity. Dev Cell 2021; 57:19-31.e6. [PMID: 34822788 DOI: 10.1016/j.devcel.2021.11.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 09/24/2021] [Accepted: 10/29/2021] [Indexed: 01/01/2023]
Abstract
The interaction between chromatin and the nuclear lamina (NL) is intrinsically important to the establishment of three-dimensional chromatin architecture and spatiotemporal regulation of gene expression. However, critical regulators involved in this process are poorly understood in plants. Here, we report that Arabidopsis PNET2 and its two homologs are bona fide inner nuclear membrane proteins and integral components of the NL. PNET2s physically interact with the plant nucleoskeleton and engage nucleosome-enriched chromatin at the nuclear periphery. Loss of all three PNET2s leads to severely disrupted growth and development, concomitant activation of abiotic and biotic stress responses, and ultimate lethality in Arabidopsis. The pent2 triple mutant also displays drastic transcriptome changes accompanied by a globally altered chromatin architecture revealed by HiC analysis. Our study identified PNET2 as an inner nuclear membrane (INM) component of the NL, which associates with chromatin and play a critical role in orchestrating gene expression and chromatin organization in plants.
Collapse
Affiliation(s)
- Yu Tang
- Department of Plant and Microbial Biology, University of California, Berkeley, CA 94720, USA; Innovative Genomics Institute, University of California, Berkeley, CA 94720, USA
| | - Qianli Dong
- Key Laboratory of Molecular Epigenetics of the Ministry of Education (MOE), Northeast Normal University, Changchun 130024, China
| | - Tianya Wang
- Key Laboratory of Molecular Epigenetics of the Ministry of Education (MOE), Northeast Normal University, Changchun 130024, China
| | - Lei Gong
- Key Laboratory of Molecular Epigenetics of the Ministry of Education (MOE), Northeast Normal University, Changchun 130024, China
| | - Yangnan Gu
- Department of Plant and Microbial Biology, University of California, Berkeley, CA 94720, USA; Innovative Genomics Institute, University of California, Berkeley, CA 94720, USA.
| |
Collapse
|
9
|
Park SH, Kim SJ, Myung K, Lee KY. Characterization of subcellular localization of eukaryotic clamp loader/unloader and its regulatory mechanism. Sci Rep 2021; 11:21817. [PMID: 34751190 PMCID: PMC8575788 DOI: 10.1038/s41598-021-01336-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 10/13/2021] [Indexed: 11/27/2022] Open
Abstract
Proliferating cell nuclear antigen (PCNA) plays a critical role as a processivity clamp for eukaryotic DNA polymerases and a binding platform for many DNA replication and repair proteins. The enzymatic activities of PCNA loading and unloading have been studied extensively in vitro. However, the subcellular locations of PCNA loaders, replication complex C (RFC) and CTF18-RFC-like-complex (RLC), and PCNA unloader ATAD5-RLC remain elusive, and the role of their subunits RFC2-5 is unknown. Here we used protein fractionation to determine the subcellular localization of RFC and RLCs and affinity purification to find molecular requirements for the newly defined location. All RFC/RLC proteins were detected in the nuclease-resistant pellet fraction. RFC1 and ATAD5 were not detected in the non-ionic detergent-soluble and nuclease-susceptible chromatin fractions, independent of cell cycle or exogenous DNA damage. We found that small RFC proteins contribute to maintaining protein levels of the RFC/RLCs. RFC1, ATAD5, and RFC4 co-immunoprecipitated with lamina-associated polypeptide 2 (LAP2) α which regulates intranuclear lamin A/C. LAP2α knockout consistently reduced detection of RFC/RLCs in the pellet fraction, while marginally affecting total protein levels. Our findings strongly suggest that PCNA-mediated DNA transaction occurs through regulatory machinery associated with nuclear structures, such as the nuclear matrix.
Collapse
Affiliation(s)
- Su Hyung Park
- Center for Genomic Integrity, Institute for Basic Science, Ulsan, 44919, Korea
| | - Seong-Jung Kim
- Center for Genomic Integrity, Institute for Basic Science, Ulsan, 44919, Korea.,Department of Biological Sciences, Ulsan National Institute of Science and Technology, Ulsan, 44919, Korea
| | - Kyungjae Myung
- Center for Genomic Integrity, Institute for Basic Science, Ulsan, 44919, Korea.,Department of Biomedical Engineering, Ulsan National Institute of Science and Technology, Ulsan, 44919, Korea
| | - Kyoo-Young Lee
- Center for Genomic Integrity, Institute for Basic Science, Ulsan, 44919, Korea.
| |
Collapse
|
10
|
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
|
11
|
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
|
12
|
Hao W, Zhao H, Li Z, Li J, Guo J, Chen Q, Gao Y, Ren M, Zhao X, Yue W. Identification of potential markers for differentiating epithelial ovarian cancer from ovarian low malignant potential tumors through integrated bioinformatics analysis. J Ovarian Res 2021; 14:46. [PMID: 33726773 PMCID: PMC7968266 DOI: 10.1186/s13048-021-00794-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Accepted: 03/05/2021] [Indexed: 01/10/2023] Open
Abstract
Background Epithelial ovarian cancer (EOC), as a lethal malignancy in women, is often diagnosed as advanced stages. In contrast, intermediating between benign and malignant tumors, ovarian low malignant potential (LMP) tumors show a good prognosis. However, the differential diagnosis of the two diseases is not ideal, resulting in delays or unnecessary therapies. Therefore, unveiling the molecular differences between LMP and EOC may contribute to differential diagnosis and novel therapeutic and preventive policies development for EOC. Methods In this study, three microarray data (GSE9899, GSE57477 and GSE27651) were used to explore the differentially expressed genes (DEGs) between LMP and EOC samples. Then, 5 genes were screened by protein–protein interaction (PPI) network, receiver operating characteristic (ROC), survival and Pearson correlation analysis. Meanwhile, chemical-core gene network construction was performed to identify the potential drugs or risk factors for EOC based on 5 core genes. Finally, we also identified the potential function of the 5 genes for EOC through pathway analysis. Results Two hundred thirty-four DEGs were successfully screened, including 81 up-regulated genes and 153 down-regulated genes. Then, 5 core genes (CCNB1, KIF20A, ASPM, AURKA, and KIF23) were identified through PPI network analysis, ROC analysis, survival and Pearson correlation analysis, which show better diagnostic efficiency and higher prognostic value for EOC. Furthermore, NetworkAnalyst was used to identify top 15 chemicals that link with the 5 core genes. Among them, 11 chemicals were potential drugs and 4 chemicals were risk factors for EOC. Finally, we found that all 5 core genes mainly regulate EOC development via the cell cycle pathway by the bioinformatic analysis. Conclusion Based on an integrated bioinformatic analysis, we identified potential biomarkers, risk factors and drugs for EOC, which may help to provide new ideas for EOC diagnosis, condition appraisal, prevention and treatment in future. Supplementary Information The online version contains supplementary material available at 10.1186/s13048-021-00794-0.
Collapse
Affiliation(s)
- Wende Hao
- Central Laboratory, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, 100026, China
| | - Hongyu Zhao
- Central Laboratory, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, 100026, China
| | - Zhefeng Li
- Central Laboratory, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, 100026, China
| | - Jie Li
- Central Laboratory, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, 100026, China
| | - Jiahao Guo
- Central Laboratory, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, 100026, China
| | - Qi Chen
- Central Laboratory, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, 100026, China
| | - Yan Gao
- Central Laboratory, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, 100026, China
| | - Meng Ren
- Central Laboratory, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, 100026, China
| | - Xiaoting Zhao
- Central Laboratory, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, 100026, China.
| | - Wentao Yue
- Central Laboratory, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, 100026, China.
| |
Collapse
|
13
|
Patil S, Sengupta K. Role of A- and B-type lamins in nuclear structure-function relationships. Biol Cell 2021; 113:295-310. [PMID: 33638183 DOI: 10.1111/boc.202000160] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2020] [Revised: 02/06/2021] [Accepted: 02/08/2021] [Indexed: 12/15/2022]
Abstract
Nuclear lamins are type V intermediate filament proteins that form a filamentous meshwork beneath the inner nuclear membrane. Additionally, a sub-population of A- and B-type lamins localizes in the nuclear interior. The nuclear lamina protects the nucleus from mechanical stress and mediates nucleo-cytoskeletal coupling. Lamins form a scaffold that partially tethers chromatin at the nuclear envelope. The nuclear lamina also stabilises protein-protein interactions involved in gene regulation and DNA repair. The lamin-based protein sub-complexes are implicated in both nuclear and cytoskeletal organisation, the mechanical stability of the nucleus, genome organisation, transcriptional regulation, genome stability and cellular differentiation. Here, we review recent research on nuclear lamins and unique roles of A- and B-type lamins in modulating various nuclear processes and their impact on cell function.
Collapse
Affiliation(s)
- Shalaka Patil
- Biology, Chromosome Biology Lab (CBL), Indian Institute of Science Education and Research (IISER), Dr. Homi Bhabha Road, Pune, 411008, India
| | - Kundan Sengupta
- Biology, Chromosome Biology Lab (CBL), Indian Institute of Science Education and Research (IISER), Dr. Homi Bhabha Road, Pune, 411008, India
| |
Collapse
|
14
|
Lesseur C, Ferreiro-Iglesias A, McKay JD, Bossé Y, Johansson M, Gaborieau V, Landi MT, Christiani DC, Caporaso NC, Bojesen SE, Amos CI, Shete S, Liu G, Rennert G, Albanes D, Aldrich MC, Tardon A, Chen C, Triantafillos L, Field JK, Teare MD, Kiemeney LA, Diergaarde B, Ferris RL, Zienolddiny S, Lam S, Olshan AF, Weissler MC, Lacko M, Risch A, Bickeböller H, Ness AR, Thomas S, Le Marchand L, Schabath MB, Wünsch-Filho V, Tajara EH, Andrew AS, Clifford GM, Lazarus P, Grankvist K, Johansson M, Arnold S, Melander O, Brunnström H, Boccia S, Cadoni G, Timens W, Obeidat M, Xiao X, Houlston RS, Hung RJ, Brennan P. Genome-wide association meta-analysis identifies pleiotropic risk loci for aerodigestive squamous cell cancers. PLoS Genet 2021; 17:e1009254. [PMID: 33667223 PMCID: PMC7968735 DOI: 10.1371/journal.pgen.1009254] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 03/17/2021] [Accepted: 11/05/2020] [Indexed: 11/20/2022] Open
Abstract
Squamous cell carcinomas (SqCC) of the aerodigestive tract have similar etiological risk factors. Although genetic risk variants for individual cancers have been identified, an agnostic, genome-wide search for shared genetic susceptibility has not been performed. To identify novel and pleotropic SqCC risk variants, we performed a meta-analysis of GWAS data on lung SqCC (LuSqCC), oro/pharyngeal SqCC (OSqCC), laryngeal SqCC (LaSqCC) and esophageal SqCC (ESqCC) cancers, totaling 13,887 cases and 61,961 controls of European ancestry. We identified one novel genome-wide significant (Pmeta<5x10-8) aerodigestive SqCC susceptibility loci in the 2q33.1 region (rs56321285, TMEM273). Additionally, three previously unknown loci reached suggestive significance (Pmeta<5x10-7): 1q32.1 (rs12133735, near MDM4), 5q31.2 (rs13181561, TMEM173) and 19p13.11 (rs61494113, ABHD8). Multiple previously identified loci for aerodigestive SqCC also showed evidence of pleiotropy in at least another SqCC site, these include: 4q23 (ADH1B), 6p21.33 (STK19), 6p21.32 (HLA-DQB1), 9p21.33 (CDKN2B-AS1) and 13q13.1(BRCA2). Gene-based association and gene set enrichment identified a set of 48 SqCC-related genes rel to DNA damage and epigenetic regulation pathways. Our study highlights the importance of cross-cancer analyses to identify pleiotropic risk loci of histology-related cancers arising at distinct anatomical sites.
Collapse
Affiliation(s)
- Corina Lesseur
- Section of Genetics, Genetic Epidemiology Group, International Agency for Research on Cancer, World Health Organization, Lyon, France
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Aida Ferreiro-Iglesias
- Section of Genetics, Genetic Epidemiology Group, International Agency for Research on Cancer, World Health Organization, Lyon, France
| | - James D. McKay
- Section of Genetics, Genetic Cancer Susceptibility Group, International Agency for Research on Cancer, World Health Organization, Lyon, France
| | - Yohan Bossé
- Department of Molecular Medicine, Institut universitaire de cardiologie et de pneumologie de Québec, Laval University, Quebec City, Canada
| | - Mattias Johansson
- Section of Genetics, Genetic Epidemiology Group, International Agency for Research on Cancer, World Health Organization, Lyon, France
| | - Valerie Gaborieau
- Section of Genetics, Genetic Epidemiology Group, International Agency for Research on Cancer, World Health Organization, Lyon, France
| | - Maria Teresa Landi
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - David C. Christiani
- Department of Environmental Health, Harvard TH Chan School of Public Health, Massachusetts General Hospital, Boston, Massachusetts, United States of America
| | - Neil C. Caporaso
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Stig E. Bojesen
- Copenhagen General Population Study, Herlev and Gentofte Hospital, Copenhagen, Denmark
| | - Christopher I. Amos
- Department of Medicine, Baylor college of Medicine, Houston, Texas, United States of America
| | - Sanjay Shete
- Department of Biostatistics, The University of Texas M.D. Anderson Cancer Center, Houston, Texas, United States of America
| | - Geoffrey Liu
- Lunenfeld-Tanenbaum Research Institute of Sinai Health System, University of Toronto, Toronto, Canada
| | - Gadi Rennert
- Clalit National Cancer Control Center, Carmel Medical Center and Technion Faculty of Medicine, Haifa, Israel
| | - Demetrius Albanes
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Melinda C. Aldrich
- Department of Thoracic Surgery, Division of Epidemiology, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Adonina Tardon
- Faculty of Medicine, University of Oviedo and CIBERESP, Oviedo, Spain
| | - Chu Chen
- Department of Epidemiology, University of Washington School of Public Health and Community Medicine, Seattle, Washington, United States of America
| | | | - John K. Field
- Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Marion Dawn Teare
- School of Health and Related Research, University Of Sheffield, Sheffield, United Kingdom
| | | | - Brenda Diergaarde
- Department of Human Genetics, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Robert L. Ferris
- UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | | | - Stephen Lam
- British Columbia Cancer Agency, Vancouver, Canada
| | - Andrew F. Olshan
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Mark C. Weissler
- Department of Otolaryngology/Head and Neck Surgery, UNC Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Martin Lacko
- Department of Otorhinolaryngology, Head and Neck Surgery, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Angela Risch
- University of Salzburg, Department of Biosciences and Cancer Cluster Salzburg, Salzburg, Austria
- Division of Epigenomics, DKFZ – German Cancer Research Center, Heidelberg, Germany
- Translational Lung Research Center Heidelberg (TLRC-H), Member of the German Center for Lung Research (DZL), Heidelberg, Germany
| | - Heike Bickeböller
- Department of Genetic Epidemiology, University Medical Center, Georg-August-University Göttingen, Göttingen, Germany
| | - Andy R. Ness
- National Institute for Health Research (NIHR) Bristol Biomedical Research Centre, University Hospitals Bristol NHS Foundation Trust, Bristol, United Kingdom
- Bristol Dental School, University of Bristol, Bristol, United Kingdom
| | - Steve Thomas
- Bristol Dental School, University of Bristol, Bristol, United Kingdom
| | - Loic Le Marchand
- Epidemiology Program, University of Hawaii Cancer Center, University of Hawaii, Honolulu, Hawaii, United States of America
| | - Matthew B. Schabath
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, United States of America
| | | | - Eloiza H. Tajara
- Department of Molecular Biology, School of Medicine of São José do Rio Preto, São José do Rio Preto, Brazil
| | - Angeline S. Andrew
- Biomedical Data Science, Geisel School of Medicine at Dartmouth, Dartmouth College, Hanover, New Hampshire, United States of America
| | - Gary M. Clifford
- Infections Section, Infections and Cancer Epidemiology Group, International Agency for Research on Cancer, World Health Organization, Lyon, France
| | - Philip Lazarus
- Department of Pharmaceutical Sciences, College of Pharmacy, Washington State University, Spokane, Washington, United States of America
| | - Kjell Grankvist
- Department of Medical Biosciences, Umeå University, Umeå, Sweden
| | | | - Susanne Arnold
- Markey Cancer Center, University of Kentucky, Lexington, Kentucky, United States of America
| | - Olle Melander
- Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden
- Department of Internal Medicine, Skåne University Hospital, Malmö, Sweden
| | - Hans Brunnström
- Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Stefania Boccia
- Section of Hygiene, University Department of Life Sciences and Public Health, Università Cattolica del Sacro Cuore, Roma, Italia
- Department of Woman and Child Health and Public Health - Public Health Area, Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italia
| | - Gabriella Cadoni
- Dipartimento Patologia Testa Collo e Organi di Senso, Istituto di Clinica Otorinolaringoiatrica, Università Cattolica del Sacro Cuore, Roma, Italia
- Dipartimento di Scienze dell’Invecchiamento, Neurologiche, Ortopediche e della Testa-Collo, Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italia
| | - Wim Timens
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- GRIAC Research Institute, University of Groningen, Groningen, The Netherlands
| | - Ma’en Obeidat
- Centre for Heart Lung Innovation, St Paul’s Hospital, The University of British Columbia, Vancouver, Canada
| | - Xiangjun Xiao
- Department of Medicine, Baylor college of Medicine, Houston, Texas, United States of America
| | - Richard S. Houlston
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, United Kingdom
| | - Rayjean J. Hung
- Prosserman Centre for Population Health Research, Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, Canada
| | - Paul Brennan
- Section of Genetics, Genetic Epidemiology Group, International Agency for Research on Cancer, World Health Organization, Lyon, France
| |
Collapse
|
15
|
Bakshi D, Katoch A, Chakraborty S, Shah R, Sharma B, Bhat A, Verma S, Bhat GR, Nagpal A, Vaishnavi S, Goswami A, Kumar R. ANKLE1 as New Hotspot Mutation for Breast Cancer in Indian Population and Has a Role in DNA Damage and Repair in Mammalian Cells. Front Genet 2021; 11:609758. [PMID: 33584808 PMCID: PMC7873468 DOI: 10.3389/fgene.2020.609758] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Accepted: 11/30/2020] [Indexed: 12/12/2022] Open
Abstract
Breast cancer has replaced cervical cancer as being the most common and having the highest mortality among women in India. ANKLE gene is conserved among organisms during evolutionary succession and is a member of LEM family proteins in lower metazoans and is involved in critical functions in the nuclear architecture, gene expression and cell signaling. ANKLE1 is the human orthologous of LEM-3 and is involved in DNA damage response and DNA repair. Whole Exome Sequencing (WES) of paired breast cancer samples was performed and ANKLE1 was found to be a new possible hotspot for predisposition of breast cancer. The mass array genotyping for breast cancer variant rs2363956 further confirmed the ANKLE1 association with the studied population of breast cancer. To elucidate the role of ANKLE1 in DNA damage, it was knocked down in MCF-7 breast cancer cell line and the expression of γH2AX was assessed. ANKLE1 knockdown cells displayed elevated levels of γ-H2AX foci in response to the cisplatin induced replication stress. The localization pattern of ANKLE1 further emphasized the role of ANKLE1 in DNA repair process. We observed that ANKLE1 is required for maintaining genomic stability and plays a role in DNA damage and repair process. These findings provided a molecular basis for the suspected role of ANKLE1 in human breast cancer and suggested an important role of this gene in controlling breast cancer development among women in India.
Collapse
Affiliation(s)
| | - Archana Katoch
- Cancer Pharmacology Division, Indian Institute of Integrative Medicine (CSIR) Jammu, Jammu, India.,Academy of Scientific and Innovative Research (AcSIR), New Delhi, India
| | - Souneek Chakraborty
- Cancer Pharmacology Division, Indian Institute of Integrative Medicine (CSIR) Jammu, Jammu, India.,Academy of Scientific and Innovative Research (AcSIR), New Delhi, India
| | - Ruchi Shah
- Shri Mata Vaishno Devi University, Katra, India
| | | | - Amrita Bhat
- Shri Mata Vaishno Devi University, Katra, India
| | | | | | | | | | - Anindya Goswami
- Cancer Pharmacology Division, Indian Institute of Integrative Medicine (CSIR) Jammu, Jammu, India.,Academy of Scientific and Innovative Research (AcSIR), New Delhi, India
| | | |
Collapse
|
16
|
Dubik N, Mai S. Lamin A/C: Function in Normal and Tumor Cells. Cancers (Basel) 2020; 12:cancers12123688. [PMID: 33316938 PMCID: PMC7764147 DOI: 10.3390/cancers12123688] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 12/07/2020] [Accepted: 12/08/2020] [Indexed: 02/07/2023] Open
Abstract
Simple Summary The aim of this review is to summarize lamin A/C’s currently known functions in both normal and diseased cells. Lamin A/C is a nuclear protein with many functions in cells, such as maintaining a cell’s structural stability, cell motility, mechanosensing, chromosome organization, gene regulation, cell differentiation, DNA damage repair, and telomere protection. Mutations of the lamin A/C gene, incorrect processing of the protein, and lamin A/C deregulation can lead to various diseases and cancer. This review touches on diseases caused by mutation and incorrect processing of lamin A/C, called laminopathies. The effect of lamin A/C deregulation in cancer is also reviewed, and lamin A/C’s potential in helping to diagnose prostate cancers more accurately is discussed. Abstract This review is focused on lamin A/C, a nuclear protein with multiple functions in normal and diseased cells. Its functions, as known to date, are summarized. This summary includes its role in maintaining a cell’s structural stability, cell motility, mechanosensing, chromosome organization, gene regulation, cell differentiation, DNA damage repair, and telomere protection. As lamin A/C has a variety of critical roles within the cell, mutations of the lamin A/C gene and incorrect processing of the protein results in a wide variety of diseases, ranging from striated muscle disorders to accelerated aging diseases. These diseases, collectively termed laminopathies, are also touched upon. Finally, we review the existing evidence of lamin A/C’s deregulation in cancer. Lamin A/C deregulation leads to various traits, including genomic instability and increased tolerance to mechanical insult, which can lead to more aggressive cancer and poorer prognosis. As lamin A/C’s expression in specific cancers varies widely, currently known lamin A/C expression in various cancers is reviewed. Additionally, Lamin A/C’s potential as a biomarker in various cancers and as an aid in more accurately diagnosing intermediate Gleason score prostate cancers is also discussed.
Collapse
|
17
|
Mirza AN, Gonzalez F, Ha SK, Oro AE. The Sky's the LEMit: New insights into nuclear structure regulation of transcription factor activity. Curr Opin Cell Biol 2020; 68:173-180. [PMID: 33227657 DOI: 10.1016/j.ceb.2020.10.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 09/24/2020] [Accepted: 10/08/2020] [Indexed: 12/31/2022]
Abstract
The nucleoskeleton has been associated with partitioning the genome into active and inactive compartments that dictate local transcription factor (TF) activity. However, recent data indicate that the nucleoskeleton and TFs reciprocally influence each other in dynamic TF trafficking pathways through the functions of LEM proteins. While the conserved peripheral recruitment of TFs by LEM proteins has been viewed as a mechanism of repressing transcription, a diversity of release mechanisms from the lamina suggest this compartment serves as a refuge for nuclear TF accumulation for rapid mobilization and signal stability. Detailed mechanisms suggest that TFs toggle between nuclear lamina refuge and nuclear matrix lamin-LEM protein complexes at sites of active transcription. In this review we will highlight emerging LEM functions acting at the interface of chromatin and nucleoskeleton to create TF trafficking networks.
Collapse
Affiliation(s)
- Amar N Mirza
- Program in Epithelial Biology, Stanford University School of Medicine Stanford, CA, 94305, USA
| | - Fernanda Gonzalez
- Program in Epithelial Biology, Stanford University School of Medicine Stanford, CA, 94305, USA
| | - Sierra K Ha
- Program in Epithelial Biology, Stanford University School of Medicine Stanford, CA, 94305, USA
| | - Anthony E Oro
- Program in Epithelial Biology, Stanford University School of Medicine Stanford, CA, 94305, USA.
| |
Collapse
|
18
|
Tang Y, Zhang X, Ge W, Zhou Y. Knockdown of LAP2α inhibits osteogenic differentiation of human adipose-derived stem cells by activating NF-κB. Stem Cell Res Ther 2020; 11:263. [PMID: 32611381 PMCID: PMC7329510 DOI: 10.1186/s13287-020-01774-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 06/01/2020] [Accepted: 06/16/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Lamina-associated polypeptide 2α (LAP2α) is a nucleoplasmic protein that has been involved in the regulation of the cell cycle, gene transcription, and adult stem cell function. LAP2α down-regulation is linked to age-related osteoporosis and bone deformities; however, the underlying mechanisms remain obscure. The present study aimed to elucidate the function of LAP2α in the osteogenic differentiation of human adipose-derived stem cells (hASCs), which are attractive sources for bone tissue engineering. METHODS The expression of LAP2α during the osteogenic differentiation of hASCs was detected firstly. A loss of function investigation was then carried out to characterize the function of LAP2α in osteogenic differentiation of hASCs both in vitro and in vivo. Moreover, RNA-sequences, western blotting, and confocal analyses were performed to clarify the molecular mechanism of LAP2α-regulated osteogenesis. RESULTS We found that LAP2α expression was upregulated upon osteogenic induction. Both in vitro and in vivo experiments indicated that LAP2α knockdown resulted in impaired osteogenic differentiation of hASCs. Mechanistically, we revealed that LAP2α deficiency activated nuclear factor kappa B (NF-κB) signaling by controlling the cytoplasmic-nuclear translocation of p65. CONCLUSIONS Collectively, our findings revealed that LAP2α functions as an essential regulator for osteogenesis of hASCs by modulating NF-κB signaling, thus providing novel insights for mesenchymal stem cell-mediated bone tissue engineering.
Collapse
Affiliation(s)
- Yiman Tang
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, Beijing, China.,Fourth Clinical Division, Peking University School and Hospital of Stomatology, Beijing, China
| | - Xiao Zhang
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, Beijing, China.,National Clinical Research Center for Oral Diseases, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Beijing, China
| | - Wenshu Ge
- National Clinical Research Center for Oral Diseases, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Beijing, China. .,Department of General Dentistry II, Peking University School and Hospital of Stomatology, 22 Zhongguancun Avenue South, Haidian District, Beijing, China.
| | - Yongsheng Zhou
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, Beijing, China.,National Clinical Research Center for Oral Diseases, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Beijing, China
| |
Collapse
|
19
|
Fischer EG. Nuclear Morphology and the Biology of Cancer Cells. Acta Cytol 2020; 64:511-519. [PMID: 32570234 DOI: 10.1159/000508780] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 05/19/2020] [Indexed: 01/09/2023]
Abstract
BACKGROUND For more than a century, diagnostic pathologists have used morphologic abnormalities of the nucleus as essential diagnostic features to distinguish benign from malignant cells. These features include nuclear enlargement and increased nuclear-to-cytoplasmic ratio, nuclear membrane irregularities, hyperchromasia, and abnormal chromatin distribution. As our knowledge about the genetic and epigenetic abnormalities of cancer cells has increased in recent decades, the pathophysiologic mechanisms that underlie these morphologic abnormalities remain incompletely understood. SUMMARY This review attempts to summarize biologic abnormalities in malignant cells related to these morphologic changes. The molecular anatomy of the nuclear envelope in normal and malignant cells is discussed as well as regulation of nuclear size and shape, regulation of signal transduction pathways by molecules of the nuclear envelope, chromatin distribution, and the effects of HPV infection on dysplastic cells in the uterine cervix. Key Message: Causes of morphologic nuclear abnormalities in malignant cells are likely multifactorial. They probably include mutations, dysregulation of signal transduction pathways, abnormal gene expression patterns, alterations of nuclear envelope proteins and chromatin, and aneuploidy.
Collapse
Affiliation(s)
- Edgar G Fischer
- Division of Surgical Pathology and Cytopathology, Department of Pathology, University of New Mexico, Albuquerque, New Mexico, USA,
| |
Collapse
|
20
|
Hu H, Ji Q, Song M, Ren J, Liu Z, Wang Z, Liu X, Yan K, Hu J, Jing Y, Wang S, Zhang W, Liu GH, Qu J. ZKSCAN3 counteracts cellular senescence by stabilizing heterochromatin. Nucleic Acids Res 2020; 48:6001-6018. [PMID: 32427330 PMCID: PMC7293006 DOI: 10.1093/nar/gkaa425] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 04/13/2020] [Accepted: 05/08/2020] [Indexed: 02/07/2023] Open
Abstract
Zinc finger protein with KRAB and SCAN domains 3 (ZKSCAN3) has long been known as a master transcriptional repressor of autophagy. Here, we identify a novel role for ZKSCAN3 in alleviating senescence that is independent of its autophagy-related activity. Downregulation of ZKSCAN3 is observed in aged human mesenchymal stem cells (hMSCs) and depletion of ZKSCAN3 accelerates senescence of these cells. Mechanistically, ZKSCAN3 maintains heterochromatin stability via interaction with heterochromatin-associated proteins and nuclear lamina proteins. Further study shows that ZKSCAN3 deficiency results in the detachment of genomic lamina-associated domains (LADs) from the nuclear lamina, loss of heterochromatin, a more accessible chromatin status and consequently, aberrant transcription of repetitive sequences. Overexpression of ZKSCAN3 not only rescues premature senescence phenotypes in ZKSCAN3-deficient hMSCs but also rejuvenates physiologically and pathologically senescent hMSCs. Together, these data reveal for the first time that ZKSCAN3 functions as an epigenetic modulator to maintain heterochromatin organization and thereby attenuate cellular senescence. Our findings establish a new functional link among ZKSCAN3, epigenetic regulation, and stem cell aging.
Collapse
Affiliation(s)
- Huifang Hu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qianzhao Ji
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Moshi Song
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Institute for Stem cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jie Ren
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
- China National Center for Bioinformation, Beijing 100101, China
- Institute for Stem cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zunpeng Liu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zehua Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiaoqian Liu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Institute for Stem cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Kaowen Yan
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Institute for Stem cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jianli Hu
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yaobin Jing
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- School of Future Technology, University of Chinese Academy of Sciences, Beijing 100190, China
| | - Si Wang
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Advanced Innovation Center for Human Brain Protection, National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing 100053, China
- Institute for Stem cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Weiqi Zhang
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
- China National Center for Bioinformation, Beijing 100101, China
- Institute for Stem cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Guang-Hui Liu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Advanced Innovation Center for Human Brain Protection, National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing 100053, China
- Institute for Stem cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jing Qu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Institute for Stem cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
21
|
Overexpression and Clinicopathological Correlation of Long Noncoding RNA TMPO-AS1 in Colorectal Cancer Patients. J Gastrointest Cancer 2019; 51:952-956. [DOI: 10.1007/s12029-019-00333-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
|
22
|
Sun DP, Liew PL, Lin CC, Hung ST, Chen TC, Fang CL, Lin KY. Clinicopathologic and Prognostic Significance of Thymopoietin-α Overexpression in Gastric Cancer. J Cancer 2019; 10:5099-5107. [PMID: 31602262 PMCID: PMC6775605 DOI: 10.7150/jca.30738] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Accepted: 07/22/2019] [Indexed: 12/18/2022] Open
Abstract
As one of the deadliest and most common malignancies in the world, gastric cancer (GC) represents a serious health threat. Despite recent advances in the field, the prognosis of patients with metastatic GC remains poor. In this study, we aimed to investigate the clinical impact of the alpha subunit of the nuclear structural protein thymopoietin (TMPO-α) in GC. The expression of TMPO-α in seven gastric cell lines was detected by immunoblotting. The expression level of TMPO-α levels in gastric tissues collected from 145 GC patients was examined by immunohistochemistry. The correlations between TMPO-α expression level and clinicopathologic parameters, as well as the association of TMPO-α expression with overall survival, were assessed. Immunohistochemistry showed that the expression of TMPO-α was significantly higher in GC tissues and cells in comparison with non-tumor tissues and cells. Furthermore, the overexpression of TMPO-α in gastric tissues (56%) was positively associated with Lauren classification (P = 0.0159), nodal status (P = 0.0265), distant metastasis (P < 0.0001), stage (P = 0.0367), and degree of differentiation (P = 0.0009). Patients with high TMPO-α levels had a significantly poorer overall survival than those with low levels (P = 0.001). Multivariate Cox regression analysis also indicated that TMPO-α was an independent prognostic marker for GC (P = 0.045). In addition, studies conducted in GC cells indicated that knockdown of TMPO-α suppressed cell proliferation and invasion. These findings indicate that TMPO-α overexpression can predict clinicopathologic features and the outcome of patients with GC.
Collapse
Affiliation(s)
- Ding-Ping Sun
- Department of Surgery, Chi Mei Medical Center, Tainan, Taiwan.,Department of Food Science and Technology, Chia Nan University of Pharmacy and Science, Tainan, Taiwan
| | - Phui-Ly Liew
- Department of Pathology, Shuang Ho Hospital, Taipei Medical University, New Taipei, Taiwan.,Department of Pathology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chih-Chan Lin
- Department of Medical Research, Chi Mei Medical Center, Tainan, Taiwan
| | - Shih-Ting Hung
- Department of Medical Research, Chi Mei Medical Center, Tainan, Taiwan
| | - Tai-Chi Chen
- Department of Medical Research, Chi Mei Medical Center, Tainan, Taiwan
| | - Chia-Lang Fang
- Department of Pathology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Department of Pathology, Wang Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Kai-Yuan Lin
- Department of Medical Research, Chi Mei Medical Center, Tainan, Taiwan.,Department of Biotechnology, Chia Nan University of Pharmacy and Science, Tainan, Taiwan
| |
Collapse
|
23
|
Sidorenko E, Vartiainen MK. Nucleoskeletal regulation of transcription: Actin on MRTF. Exp Biol Med (Maywood) 2019; 244:1372-1381. [PMID: 31142145 DOI: 10.1177/1535370219854669] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Myocardin-related transcription factor A (MRTF-A) and serum response factor (SRF) form an essential transcriptional complex that regulates the expression of many cytoskeletal genes in response to dynamic changes in the actin cytoskeleton. The nucleoskeleton, a “dynamic network of networks,” consists of numerous proteins that contribute to nuclear shape and to its various functions, including gene expression. In this review, we will discuss recent work that has identified many nucleoskeletal proteins, such as nuclear lamina and lamina-associated proteins, nuclear actin, and the linker of the cytoskeleton and nucleoskeleton complex as important regulators of MRTF-A/SRF transcriptional activity, especially in the context of mechanical control of transcription. Impact statement Regulation of gene expression is a fundamental cellular process that ensures the appropriate response of a cell to its surroundings. Alongside biochemical signals, mechanical cues, such as substrate rigidity, have been recognized as key regulators of gene expression. Nucleoskeletal components play an important role in mechanoresponsive transcription, particularly in controlling the activity of MRTF-A/SRF transcription factors. This ensures that the cell can balance the internal and external mechanical forces by fine-tuning the expression of cytoskeletal genes.
Collapse
Affiliation(s)
- Ekaterina Sidorenko
- Institute of Biotechnology, Helsinki Institute of Life Science, University of Helsinki, Helsinki 00014, Finland
| | - Maria K Vartiainen
- Institute of Biotechnology, Helsinki Institute of Life Science, University of Helsinki, Helsinki 00014, Finland
| |
Collapse
|
24
|
Alvarado-Kristensson M, Rosselló CA. The Biology of the Nuclear Envelope and Its Implications in Cancer Biology. Int J Mol Sci 2019; 20:E2586. [PMID: 31137762 PMCID: PMC6566445 DOI: 10.3390/ijms20102586] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2019] [Revised: 05/07/2019] [Accepted: 05/25/2019] [Indexed: 12/18/2022] Open
Abstract
The formation of the nuclear envelope and the subsequent compartmentalization of the genome is a defining feature of eukaryotes. Traditionally, the nuclear envelope was purely viewed as a physical barrier to preserve genetic material in eukaryotic cells. However, in the last few decades, it has been revealed to be a critical cellular component in controlling gene expression and has been implicated in several human diseases. In cancer, the relevance of the cell nucleus was first reported in the mid-1800s when an altered nuclear morphology was observed in tumor cells. This review aims to give a current and comprehensive view of the role of the nuclear envelope on cancer first by recapitulating the changes of the nuclear envelope during cell division, second, by reviewing the role of the nuclear envelope in cell cycle regulation, signaling, and the regulation of the genome, and finally, by addressing the nuclear envelope link to cell migration and metastasis and its use in cancer prognosis.
Collapse
Affiliation(s)
- Maria Alvarado-Kristensson
- Molecular Pathology, Department of Translational Medicine, Lund University, Skåne University Hospital, 20502 Malmö, Sweden.
| | - Catalina Ana Rosselló
- Laboratory of Molecular Cell Biomedicine, University of the Balearic Islands, 07121 Palma de Mallorca, Spain.
- Lipopharma Therapeutics, Isaac Newton, 07121 Palma de Mallorca, Spain.
| |
Collapse
|
25
|
Liu C, Yu H, Shen X, Qiao J, Wu X, Chang J, Zhu X, Wang J, Shen X. Prognostic significance and biological function of Lamina-associated polypeptide 2 in non-small-cell lung cancer. Onco Targets Ther 2019; 12:3817-3827. [PMID: 31190881 PMCID: PMC6529027 DOI: 10.2147/ott.s179870] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Accepted: 04/26/2019] [Indexed: 01/24/2023] Open
Abstract
Purpose: Lamina-associated polypeptide 2 (LAP2; encoded by TMPO), is a nuclear protein that may affect chromatin regulation and gene expression through dynamically binding to nuclear lamin. TMPO (LAP2) plays dual roles of either suppressing or promoting proliferation of cells, depending on the status of the cell. It has been reported that TMPO is up-regulated in various cancer types. However, its function in lung cancer has not been studied yet. Materials and methods: A series of clinical microarray datasets for lung cancer were investigated to demonstrate the expression of TMPO. The transcription of TMPO gene in human lung cancer was analyzed using Oncomine platform (www.oncomine.org) according to the standardized procedures described previously. Four separate datasets (Hou Lung, Okayama Lung, Beer Lung, and Garber Lung) were analyzed. Results: Here, we show that TMPO is over-expressed in lung cancer tissues, and that a high level of TMPO indicates a poor prognosis in lung cancer patients. Knockdown of TMPO in lung cancer cells inhibits cell proliferation and induces apoptosis. Also, down-regulation of TMPO leads to an impaired metastatic ability of tumor cells. A nude mice tumor model show that knockdown of TMPO suppresses tumor formation in vivo. Conclusion: Collectively, this study suggests TMPO as an oncogene and a novel prognostic gene in lung cancer.
Collapse
Affiliation(s)
- Chang Liu
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, People's Republic of China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, People's Republic of China
| | - Hui Yu
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, People's Republic of China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, People's Republic of China
| | - Xuxia Shen
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, People's Republic of China.,Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai 200032, People's Republic of China
| | - Jie Qiao
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, People's Republic of China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, People's Republic of China
| | - Xianghua Wu
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, People's Republic of China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, People's Republic of China
| | - Jianhua Chang
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, People's Republic of China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, People's Republic of China
| | - Xunxia Zhu
- Department of Thoracic Surgery, The Affiliated Huadong Hospital of Fudan University, Shanghai 200040, People's Republic of China
| | - Jialei Wang
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, People's Republic of China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, People's Republic of China
| | - Xiaoyong Shen
- Department of Thoracic Surgery, The Affiliated Huadong Hospital of Fudan University, Shanghai 200040, People's Republic of China
| |
Collapse
|
26
|
Mirza AN, McKellar SA, Urman NM, Brown AS, Hollmig T, Aasi SZ, Oro AE. LAP2 Proteins Chaperone GLI1 Movement between the Lamina and Chromatin to Regulate Transcription. Cell 2019; 176:198-212.e15. [PMID: 30503211 PMCID: PMC6379078 DOI: 10.1016/j.cell.2018.10.054] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 07/13/2018] [Accepted: 10/26/2018] [Indexed: 12/20/2022]
Abstract
Understanding transcription factor navigation through the nucleus remains critical for developing targeted therapeutics. The GLI1 transcription factor must maintain maximal Hedgehog pathway output in basal cell carcinomas (BCCs), and we have previously shown that resistant BCCs increase GLI1 deacetylation through atypical protein kinase Cι/λ (aPKC) and HDAC1. Here we identify a lamina-associated polypeptide 2 (LAP2) isoform-dependent nuclear chaperoning system that regulates GLI1 movement between the nuclear lamina and nucleoplasm to achieve maximal activation. LAP2β forms a two-site interaction with the GLI1 zinc-finger domain and acetylation site, stabilizing an acetylation-dependent reserve on the inner nuclear membrane (INM). By contrast, the nucleoplasmic LAP2α competes with LAP2β for GLI1 while scaffolding HDAC1 to deacetylate the secondary binding site. aPKC functions to promote GLI1 association with LAP2α, promoting egress off the INM. GLI1 intranuclear trafficking by LAP2 isoforms represents a powerful signal amplifier in BCCs with implications for zinc finger-based signal transduction and therapeutics.
Collapse
Affiliation(s)
- Amar N Mirza
- Program in Epithelial Biology and Department of Dermatology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Siegen A McKellar
- Program in Epithelial Biology and Department of Dermatology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Nicole M Urman
- Program in Epithelial Biology and Department of Dermatology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Alexander S Brown
- Program in Epithelial Biology and Department of Dermatology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Tyler Hollmig
- Program in Epithelial Biology and Department of Dermatology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Sumaira Z Aasi
- Program in Epithelial Biology and Department of Dermatology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Anthony E Oro
- Program in Epithelial Biology and Department of Dermatology, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
27
|
Alhudiri IM, Nolan CC, Ellis IO, Elzagheid A, Rakha EA, Green AR, Chapman CJ. Expression of Lamin A/C in early-stage breast cancer and its prognostic value. Breast Cancer Res Treat 2019; 174:661-668. [DOI: 10.1007/s10549-018-05092-w] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Accepted: 12/07/2018] [Indexed: 10/27/2022]
|
28
|
Majerska J, Feretzaki M, Glousker G, Lingner J. Transformation-induced stress at telomeres is counteracted through changes in the telomeric proteome including SAMHD1. Life Sci Alliance 2018; 1:e201800121. [PMID: 30456372 PMCID: PMC6238619 DOI: 10.26508/lsa.201800121] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Revised: 07/04/2018] [Accepted: 07/05/2018] [Indexed: 12/13/2022] Open
Abstract
The authors apply telomeric chromatin analysis to identify factors that accumulate at telomeres during cellular transformation, promoting telomere replication and repair and counteracting oncogene-borne telomere replication stress. Telomeres play crucial roles during tumorigenesis, inducing cellular senescence upon telomere shortening and extensive chromosome instability during telomere crisis. However, it has not been investigated if and how cellular transformation and oncogenic stress alter telomeric chromatin composition and function. Here, we transform human fibroblasts by consecutive transduction with vectors expressing hTERT, the SV40 early region, and activated H-RasV12. Pairwise comparisons of the telomeric proteome during different stages of transformation reveal up-regulation of proteins involved in chromatin remodeling, DNA repair, and replication at chromosome ends. Depletion of several of these proteins induces telomere fragility, indicating their roles in replication of telomeric DNA. Depletion of SAMHD1, which has reported roles in DNA resection and homology-directed repair, leads to telomere breakage events in cells deprived of the shelterin component TRF1. Thus, our analysis identifies factors, which accumulate at telomeres during cellular transformation to promote telomere replication and repair, resisting oncogene-borne telomere replication stress.
Collapse
Affiliation(s)
- Jana Majerska
- School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland.,Swiss Institute for Experimental Cancer Research, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Marianna Feretzaki
- School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland.,Swiss Institute for Experimental Cancer Research, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Galina Glousker
- School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland.,Swiss Institute for Experimental Cancer Research, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Joachim Lingner
- School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland.,Swiss Institute for Experimental Cancer Research, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| |
Collapse
|
29
|
Gao A, Sun T, Ma G, Cao J, Hu Q, Chen L, Wang Y, Wang Q, Sun J, Wu R, Wu Q, Zhou J, Liu L, Hu J, Dong JT, Zhu Z. LEM4 confers tamoxifen resistance to breast cancer cells by activating cyclin D-CDK4/6-Rb and ERα pathway. Nat Commun 2018; 9:4180. [PMID: 30301939 PMCID: PMC6177406 DOI: 10.1038/s41467-018-06309-8] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Accepted: 07/02/2018] [Indexed: 02/07/2023] Open
Abstract
The elucidation of molecular events that confer tamoxifen resistance to estrogen receptor α (ER) positive breast cancer is of major scientific and therapeutic importance. Here, we report that LEM4 overexpression renders ER+ breast cancer cells resistant to tamoxifen by activating the cyclin D-CDK4/6 axis and the ERα signaling. We show that LEM4 overexpression accelerates tumor growth. Interaction with LEM4 stabilizes CDK4 and Rb, promotes Rb phosphorylation and the G1/S phase transition. LEM4 depletion or combined tamoxifen and PD0332991 treatment significantly reverses tamoxifen resistance. Furthermore, LEM4 interacts with and stabilizes both Aurora-A and ERα, promotes Aurora-A mediated phosphorylation of ERα-Ser167, leading to increase in ERα DNA-binding and transactivation activity. Elevated levels of LEM4 correlates with poorer relapse-free survival in patients with ER+ breast cancer undergoing endocrine therapy. Thus, LEM4 represents a prognostic marker and an attractive target for breast cancer therapeutics. Functional antagonism of LEM4 could overcome tamoxifen resistance.
Collapse
Affiliation(s)
- Ang Gao
- Department of Genetics and Cell Biology, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Tonghua Sun
- Department of Genetics and Cell Biology, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Gui Ma
- Department of Genetics and Cell Biology, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Jiangran Cao
- Department of Genetics and Cell Biology, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Qingxia Hu
- Department of Genetics and Cell Biology, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Ling Chen
- Department of Pathology, Tianjin Central Hospital of Gynecology and Obstetrics, Tianjin, 300100, China
| | - Yanxin Wang
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Qianying Wang
- Department of Genetics and Cell Biology, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Jiafu Sun
- Department of Genetics and Cell Biology, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Rui Wu
- Department of Genetics and Cell Biology, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Qiao Wu
- Department of Genetics and Cell Biology, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Jiaxi Zhou
- State Key Laboratory of Experimental Hematology, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, 288 Nanjing Road, Tianjin, 300020, China
| | - Lin Liu
- Department of Genetics and Cell Biology, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Junjie Hu
- Department of Genetics and Cell Biology, College of Life Sciences, Nankai University, Tianjin, 300071, China.
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China.
| | - Jin-Tang Dong
- Department of Genetics and Cell Biology, College of Life Sciences, Nankai University, Tianjin, 300071, China.
- Department of Hematology and Medical Oncology, School of Medicine, Winship Cancer Institute, Emory University, Atlanta, Georgia.
| | - Zhengmao Zhu
- Department of Genetics and Cell Biology, College of Life Sciences, Nankai University, Tianjin, 300071, China.
| |
Collapse
|
30
|
Lamin A/C might be involved in the EMT signalling pathway. Gene 2018; 663:51-64. [PMID: 29665450 DOI: 10.1016/j.gene.2018.04.040] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 04/13/2018] [Indexed: 12/13/2022]
Abstract
We have previously reported a heterogeneous expression pattern of the nuclear membrane protein lamin A/C in low- and high-Gleason score (GS) prostate cancer (PC) tissues, and we have now found that this change is not associated with LMNA mutations. This expression pattern appears to be similar to the process of epithelial to mesenchymal transition (EMT) or to that of mesenchymal to epithelial transition (MET). The role of lamin A/C in EMT or MET in PC remains unclear. Therefore, we first investigated the expression levels of and the associations between lamin A/C and several common EMT markers, such as E-cadherin, N-cadherin, β-catenin, snail, slug and vimentin in PC tissues with different GS values and in different cell lines with varying invasion abilities. Our results suggest that lamin A/C might constitute a type of epithelial marker that better signifies EMT and MET in PC tissue, since a decrease in lamin A/C expression in GS 4 + 5 cases is likely associated with the EMT process, while the re-expression of lamin A/C in GS 5 + 4 cases is likely linked with MET. The detailed GS better exhibited the changes in lamin A/C and the EMT markers examined. Lamin A/C overexpression or knockdown had an impact on EMT biomarkers in a cell model by direct regulation of β-catenin. Hence, we suggest that lamin A/C might serve as a reliable epithelial biomarker for the distinction of PC cell differentiation and might also be a fundamental factor in the occurrence of EMT or MET in PC.
Collapse
|
31
|
de Leeuw R, Gruenbaum Y, Medalia O. Nuclear Lamins: Thin Filaments with Major Functions. Trends Cell Biol 2017; 28:34-45. [PMID: 28893461 DOI: 10.1016/j.tcb.2017.08.004] [Citation(s) in RCA: 222] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Revised: 08/08/2017] [Accepted: 08/14/2017] [Indexed: 10/18/2022]
Abstract
The nuclear lamina is a nuclear peripheral meshwork that is mainly composed of nuclear lamins, although a small fraction of lamins also localizes throughout the nucleoplasm. Lamins are classified as type V intermediate filament (IF) proteins. Mutations in lamin genes cause at least 15 distinct human diseases, collectively termed laminopathies, including muscle, metabolic, and neuronal diseases, and can cause accelerated aging. Most of these mutations are in the LMNA gene encoding A-type lamins. A growing number of nuclear proteins are known to bind lamins and are implicated in both nuclear and cytoskeletal organization, mechanical stability, chromatin organization, signaling, gene regulation, genome stability, and cell differentiation. Recent studies reveal the organization of the lamin filament meshwork in somatic cells where they assemble as tetramers in cross-section of the filaments.
Collapse
Affiliation(s)
- Rebecca de Leeuw
- Department of Biochemistry, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Yosef Gruenbaum
- Department of Genetics, The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Ohad Medalia
- Department of Biochemistry, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland; Department of Life Sciences and the National Institute for Biotechnology in the Negev, Ben-Gurion University, Beer-Sheva 84105, Israel.
| |
Collapse
|
32
|
Nikolakaki E, Mylonis I, Giannakouros T. Lamin B Receptor: Interplay between Structure, Function and Localization. Cells 2017; 6:cells6030028. [PMID: 28858257 PMCID: PMC5617974 DOI: 10.3390/cells6030028] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Revised: 08/28/2017] [Accepted: 08/30/2017] [Indexed: 12/15/2022] Open
Abstract
Lamin B receptor (LBR) is an integral protein of the inner nuclear membrane, containing a hydrophilic N-terminal end protruding into the nucleoplasm, eight hydrophobic segments that span the membrane and a short, nucleoplasmic C-terminal tail. Two seemingly unrelated functions have been attributed to LBR. Its N-terminal domain tethers heterochromatin to the nuclear periphery, thus contributing to the shape of interphase nuclear architecture, while its transmembrane domains exhibit sterol reductase activity. Mutations within the transmembrane segments result in defects in cholesterol synthesis and are associated with diseases such as the Pelger–Huët anomaly and Greenberg skeletal dysplasia, whereas no such harmful mutations related to the anchoring properties of LBR have been reported so far. Recent evidence suggests a dynamic regulation of LBR expression levels, structural organization, localization and function, in response to various signals. The molecular mechanisms underlying this dynamic behavior have not yet been fully unraveled. Here, we provide an overview of the current knowledge of the interplay between the structure, function and localization of LBR, and hint at the interconnection of the two distinct functions of LBR.
Collapse
Affiliation(s)
- Eleni Nikolakaki
- Laboratory of Biochemistry, Department of Chemistry, Aristotelian University, Thessaloniki 54124, Greece.
| | - Ilias Mylonis
- Laboratory of Biochemistry, Faculty of Medicine, University of Thessaly, Panepistimiou 3 BIOPOLIS, Larissa 41500, Greece.
| | - Thomas Giannakouros
- Laboratory of Biochemistry, Department of Chemistry, Aristotelian University, Thessaloniki 54124, Greece.
| |
Collapse
|
33
|
Golubtsova NN, Filippov FN, Gunin AG. Lamin A and lamin-associated polypeptide 2 (LAP-2) in human skin fibroblasts in the process of aging. ADVANCES IN GERONTOLOGY 2017. [DOI: 10.1134/s2079057017010076] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
|
34
|
Zhang L, Wang G, Chen S, Ding J, Ju S, Cao H, Tian H. Depletion of thymopoietin inhibits proliferation and induces cell cycle arrest/apoptosis in glioblastoma cells. World J Surg Oncol 2016; 14:267. [PMID: 27756319 PMCID: PMC5069786 DOI: 10.1186/s12957-016-1018-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Accepted: 10/04/2016] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND Glioblastoma (GBM) is the most malignant nervous system tumor with an almost 100 % recurrence rate. Thymopoietin (TMPO) has been demonstrated to be upregulated in various tumors, including lung cancer, breast cancer, and so on, but its role in GBM has not been reported. This study was aimed to determine the role of TMPO in GBM. METHODS Publicly available Oncomine dataset analysis was used to explore the expression level of TMPO in GBM specimens. Then the expression of TMPO was knocked down in GBM cells using lentiviral system, and the knockdown efficacy was further validated by real-time quantitative PCR and western blot analysis. Furthermore, the effects of TMPO silencing on GBM cell proliferation and apoptosis were examined by MTT, colony formation, and flow cytometry analysis. Meanwhile, the expression of apoptotic markers caspase-3 and poly(ADP-ribose) polymerase (PARP) were investigated by western blot analysis. RESULTS This study observed that the expression of TMPO in GBM specimens was remarkably higher than that in normal brain specimens. Moreover, knockdown of TMPO could significantly inhibit cell proliferation and arrest cell cycle progression at the G2/M phase. It also found that TMPO knockdown promoted cell apoptosis by upregulation of the cleavage of caspase-3 and PARP protein levels which are the markers of apoptosis. CONCLUSIONS The results suggested TMPO might be a novel therapeutic target for GBM.
Collapse
Affiliation(s)
- Lin Zhang
- Department of Neurosurgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, School of Medicine, Shanghai Jiao Tong University, No. 600 Yishan Road, Xuhui District, Shanghai, 200233, China
| | - Gan Wang
- Department of Neurosurgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, School of Medicine, Shanghai Jiao Tong University, No. 600 Yishan Road, Xuhui District, Shanghai, 200233, China
| | - Shiwen Chen
- Department of Neurosurgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, School of Medicine, Shanghai Jiao Tong University, No. 600 Yishan Road, Xuhui District, Shanghai, 200233, China
| | - Jun Ding
- Department of Neurosurgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, School of Medicine, Shanghai Jiao Tong University, No. 600 Yishan Road, Xuhui District, Shanghai, 200233, China
| | - Shiming Ju
- Department of Neurosurgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, School of Medicine, Shanghai Jiao Tong University, No. 600 Yishan Road, Xuhui District, Shanghai, 200233, China
| | - Heli Cao
- Department of Neurosurgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, School of Medicine, Shanghai Jiao Tong University, No. 600 Yishan Road, Xuhui District, Shanghai, 200233, China
| | - Hengli Tian
- Department of Neurosurgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, School of Medicine, Shanghai Jiao Tong University, No. 600 Yishan Road, Xuhui District, Shanghai, 200233, China.
| |
Collapse
|
35
|
Zlopasa L, Brachner A, Foisner R. Nucleo-cytoplasmic shuttling of the endonuclease ankyrin repeats and LEM domain-containing protein 1 (Ankle1) is mediated by canonical nuclear export- and nuclear import signals. BMC Cell Biol 2016; 17:23. [PMID: 27245214 PMCID: PMC4888674 DOI: 10.1186/s12860-016-0102-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Accepted: 05/19/2016] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Ankyrin repeats and LEM domain containing protein 1 (Ankle1) belongs to the LEM protein family, whose members share a chromatin-interacting LEM motif. Unlike most other LEM proteins, Ankle1 is not an integral protein of the inner nuclear membrane but shuttles between the nucleus and the cytoplasm. It contains a GIY-YIG-type nuclease domain, but its function is unknown. The mammalian genome encodes only one other GIY-YIG domain protein, termed Slx1. Slx1 has been described as a resolvase that processes Holliday junctions during homologous recombination-mediated DNA double strand break repair. Resolvase activity is regulated in a spatial and temporal manner during the cell cycle. We hypothesized that Ankle1 may have a similar function and its nucleo-cytoplasmic shuttling may contribute to the regulation of Ankle1 activity. Hence, we aimed at identifying the domains mediating Ankle1 shuttling and investigating whether cellular localization is affected during DNA damage response. RESULTS Sequence analysis predicts the presence of two canonical nuclear import and export signals in Ankle1. Immunofluorescence microscopy of cells expressing wild-type and various mutated Ankle1-fusion proteins revealed a C-terminally located classical monopartite nuclear localization signal and a centrally located CRM1-dependent nuclear export signal that mediate nucleo-cytoplasmic shuttling of Ankle1. These sequences are also functional in heterologous proteins. The predominant localization of Ankle1 in the cytoplasm, however, does not change upon induction of several DNA damage response pathways throughout the cell cycle. CONCLUSIONS We identified the domains mediating nuclear import and export of Ankle1. Ankle1's cellular localization was not affected following DNA damage.
Collapse
Affiliation(s)
- Livija Zlopasa
- Max F. Perutz Laboratories (MFPL), Department of Medical Biochemistry, Medical University of Vienna, Vienna Biocenter (VBC), Vienna, Austria
| | - Andreas Brachner
- Max F. Perutz Laboratories (MFPL), Department of Medical Biochemistry, Medical University of Vienna, Vienna Biocenter (VBC), Vienna, Austria.
| | - Roland Foisner
- Max F. Perutz Laboratories (MFPL), Department of Medical Biochemistry, Medical University of Vienna, Vienna Biocenter (VBC), Vienna, Austria.
| |
Collapse
|
36
|
Vidak S, Kubben N, Dechat T, Foisner R. Proliferation of progeria cells is enhanced by lamina-associated polypeptide 2α (LAP2α) through expression of extracellular matrix proteins. Genes Dev 2016; 29:2022-36. [PMID: 26443848 PMCID: PMC4604344 DOI: 10.1101/gad.263939.115] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
A single heterozygous mutation of LMNA generates the lamin A/C variant progerin and causes Hutchinson-Gilford progeria syndrome (HGPS). Vidak et al. show that this mutation leads to loss of LAP2α and nucleoplasmic lamins A/C, impaired proliferation, and down-regulation of extracellular matrix components. Ectopic expression of LAP2α in cells expressing progerin restores proliferation and extracellular matrix expression but not the levels of nucleoplasmic lamins A/C. Lamina-associated polypeptide 2α (LAP2α) localizes throughout the nucleoplasm and interacts with the fraction of lamins A/C that is not associated with the peripheral nuclear lamina. The LAP2α–lamin A/C complex negatively affects cell proliferation. Lamins A/C are encoded by LMNA, a single heterozygous mutation of which causes Hutchinson-Gilford progeria syndrome (HGPS). This mutation generates the lamin A variant progerin, which we show here leads to loss of LAP2α and nucleoplasmic lamins A/C, impaired proliferation, and down-regulation of extracellular matrix components. Surprisingly, contrary to wild-type cells, ectopic expression of LAP2α in cells expressing progerin restores proliferation and extracellular matrix expression but not the levels of nucleoplasmic lamins A/C. We conclude that, in addition to its cell cycle-inhibiting function with lamins A/C, LAP2α can also regulate extracellular matrix components independently of lamins A/C, which may help explain the proliferation-promoting function of LAP2α in cells expressing progerin.
Collapse
Affiliation(s)
- Sandra Vidak
- Max F. Perutz Laboratories (MFPL), Department of Medical Biochemistry, Medical University of Vienna, Vienna Biocenter (VBC), A-1030 Vienna, Austria
| | - Nard Kubben
- National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Thomas Dechat
- Max F. Perutz Laboratories (MFPL), Department of Medical Biochemistry, Medical University of Vienna, Vienna Biocenter (VBC), A-1030 Vienna, Austria
| | - Roland Foisner
- Max F. Perutz Laboratories (MFPL), Department of Medical Biochemistry, Medical University of Vienna, Vienna Biocenter (VBC), A-1030 Vienna, Austria
| |
Collapse
|
37
|
Webster BM, Lusk CP. Border Safety: Quality Control at the Nuclear Envelope. Trends Cell Biol 2015; 26:29-39. [PMID: 26437591 DOI: 10.1016/j.tcb.2015.08.002] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Revised: 07/27/2015] [Accepted: 08/07/2015] [Indexed: 10/23/2022]
Abstract
The unique biochemical identity of the nuclear envelope confers its capacity to establish a barrier that protects the nuclear compartment and directly contributes to nuclear function. Recent work uncovered quality control mechanisms employing the endosomal sorting complexes required for transport (ESCRT) machinery and a new arm of endoplasmic reticulum-associated protein degradation (ERAD) to counteract the unfolding, damage, or misassembly of nuclear envelope proteins and ensure the integrity of the nuclear envelope membranes. Moreover, cells have the capacity to recognize and triage defective nuclear pore complexes to prevent their inheritance and preserve the longevity of progeny. These mechanisms serve to highlight the diverse strategies used by cells to maintain nuclear compartmentalization; we suggest they mitigate the progression and severity of diseases associated with nuclear envelope malfunction such as the laminopathies.
Collapse
|
38
|
Shimi T, Kittisopikul M, Tran J, Goldman AE, Adam SA, Zheng Y, Jaqaman K, Goldman RD. Structural organization of nuclear lamins A, C, B1, and B2 revealed by superresolution microscopy. Mol Biol Cell 2015; 26:4075-86. [PMID: 26310440 PMCID: PMC4710238 DOI: 10.1091/mbc.e15-07-0461] [Citation(s) in RCA: 188] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Accepted: 08/20/2015] [Indexed: 12/22/2022] Open
Abstract
Superresolution microscopy and computational image analysis demonstrate that the four nuclear lamin isoforms of mammalian cells are each organized into distinct meshwork structures sharing similar physical characteristics. Knockouts of single lamins alter the structure of the remaining lamins, suggesting interactions among the meshworks. The nuclear lamina is a key structural element of the metazoan nucleus. However, the structural organization of the major proteins composing the lamina is poorly defined. Using three-dimensional structured illumination microscopy and computational image analysis, we characterized the supramolecular structures of lamin A, C, B1, and B2 in mouse embryo fibroblast nuclei. Each isoform forms a distinct fiber meshwork, with comparable physical characteristics with respect to mesh edge length, mesh face area and shape, and edge connectivity to form faces. Some differences were found in face areas among isoforms due to variation in the edge lengths and number of edges per face, suggesting that each meshwork has somewhat unique assembly characteristics. In fibroblasts null for the expression of either lamins A/C or lamin B1, the remaining lamin meshworks are altered compared with the lamin meshworks in wild-type nuclei or nuclei lacking lamin B2. Nuclei lacking LA/C exhibit slightly enlarged meshwork faces and some shape changes, whereas LB1-deficient nuclei exhibit primarily a substantial increase in face area. These studies demonstrate that individual lamin isoforms assemble into complex networks within the nuclear lamina and that A- and B-type lamins have distinct roles in maintaining the organization of the nuclear lamina.
Collapse
Affiliation(s)
- Takeshi Shimi
- Department of Cell and Molecular Biology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| | - Mark Kittisopikul
- Department of Biophysics, UT Southwestern Medical Center, Dallas, TX 75390
| | - Joseph Tran
- Department of Embryology, Carnegie Institution for Science, Baltimore, MD 21218
| | - Anne E Goldman
- Department of Cell and Molecular Biology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| | - Stephen A Adam
- Department of Cell and Molecular Biology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| | - Yixian Zheng
- Department of Embryology, Carnegie Institution for Science, Baltimore, MD 21218
| | - Khuloud Jaqaman
- Department of Biophysics, UT Southwestern Medical Center, Dallas, TX 75390
| | - Robert D Goldman
- Department of Cell and Molecular Biology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| |
Collapse
|
39
|
Di Luca A, Henry M, Meleady P, O'Connor R. Label-free LC-MS analysis of HER2+ breast cancer cell line response to HER2 inhibitor treatment. ACTA ACUST UNITED AC 2015; 23:40. [PMID: 26238995 PMCID: PMC4524286 DOI: 10.1186/s40199-015-0120-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Accepted: 06/24/2015] [Indexed: 12/21/2022]
Abstract
BACKGROUND Human epidermal growth-factor receptor (HER)-2 is overexpressed in 25 % of breast-cancers and is associated with an aggressive form of the disease with significantly shortened disease free and overall survival. In recent years, the use of HER2-targeted therapies, monoclonal-antibodies and small molecule tyrosine-kinase inhibitors has significantly improved the clinical outcome for HER2-positive breast-cancer patients. However, only a fraction of HER2-amplified patients will respond to therapy and the use of these treatments is often limited by tumour drug insensitivity or resistance and drug toxicities. Currently there is no way to identify likely responders or rational combinations with the potential to improve HER2-focussed treatment outcome. METHODS In order to further understand the molecular mechanisms of treatment-response with HER2-inhibitors, we used a highly-optimised and reproducible quantitative label-free LC-MS strategy to characterize the proteomes of HER2-overexpressing breast-cancer cell-lines (SKBR3, BT474 and HCC1954) in response to drug-treatment with HER2-inhibitors (lapatinib, neratinib or afatinib). RESULTS Following 12 ours treatment with different HER2-inhibitors in the BT474 cell-line; compared to the untreated cells, 16 proteins changed significantly in abundance following lapatinib treatment (1 μM), 21 proteins changed significantly following neratinib treatment (150 nM) and 38 proteins changed significantly following afatinib treatment (150 nM). Whereas following 24 hours treatment with neratinib (200 nM) 46 proteins changed significantly in abundance in the HCC1954 cell-line and 23 proteins in the SKBR3 cell-line compared to the untreated cells. Analysing the data we found that, proteins like trifunctional-enzyme subunit-alpha, mitochondrial; heterogeneous nuclear ribonucleoprotein-R and lamina-associated polypeptide 2, isoform alpha were up-regulated whereas heat shock cognate 71 kDa protein was down-regulated in 3 or more comparisons. CONCLUSION This proteomic study highlights several proteins that are closely associated with early HER2-inhibitor response and will provide a valuable resource for further investigation of ways to improve efficacy of breast-cancer treatment.
Collapse
Affiliation(s)
- Alessio Di Luca
- National Institute for Cellular Biotechnology, DCU, Glasnevin, Dublin 9, Dublin, Ireland.
| | - Michael Henry
- National Institute for Cellular Biotechnology, DCU, Glasnevin, Dublin 9, Dublin, Ireland.
| | - Paula Meleady
- National Institute for Cellular Biotechnology, DCU, Glasnevin, Dublin 9, Dublin, Ireland.
| | - Robert O'Connor
- National Institute for Cellular Biotechnology, DCU, Glasnevin, Dublin 9, Dublin, Ireland. .,School of Nursing and Human Sciences, DCU, Glasnevin, Dublin 9, Dublin, Ireland.
| |
Collapse
|
40
|
Barton LJ, Soshnev AA, Geyer PK. Networking in the nucleus: a spotlight on LEM-domain proteins. Curr Opin Cell Biol 2015; 34:1-8. [PMID: 25863918 DOI: 10.1016/j.ceb.2015.03.005] [Citation(s) in RCA: 146] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Revised: 03/23/2015] [Accepted: 03/25/2015] [Indexed: 11/29/2022]
Abstract
Proteins resident in the inner nuclear membrane and underlying nuclear lamina form a network that regulates nuclear functions. This review highlights a prominent family of nuclear lamina proteins that carries the LAP2-emerin-MAN1-domain (LEM-D). LEM-D proteins share an ability to bind lamins and tether repressive chromatin at the nuclear periphery. The importance of this family is underscored by findings that loss of individual LEM-D proteins causes progressive, tissue-restricted diseases, known as laminopathies. Diverse functions of LEM-D proteins are linked to interactions with unique and overlapping partners including signal transduction effectors, transcription factors and architectural proteins. Recent investigations suggest that LEM-D proteins form hubs within the nuclear lamina that integrate external signals important for tissue homeostasis and maintenance of progenitor cell populations.
Collapse
Affiliation(s)
- Lacy J Barton
- Department of Biochemistry, University of Iowa, College of Medicine, Iowa City, IA 52242, USA; Skirball Institute, Department of Cell Biology, NYU School of Medicine, NYU Langone Medical Center, New York, NY 10016, USA
| | - Alexey A Soshnev
- Laboratory of Chromatin Biology and Epigenetics, The Rockefeller University, New York, NY, 10065, USA
| | - Pamela K Geyer
- Department of Biochemistry, University of Iowa, College of Medicine, Iowa City, IA 52242, USA.
| |
Collapse
|