1
|
Solomon AD, Gouttia OG, Wang L, Zhu S, Wang F, Li Y, Paydar M, Bessho T, Kwok BH, Peng A. γ-tubulin mediates DNA double-strand break repair. J Cell Sci 2025; 138:jcs262255. [PMID: 40135584 PMCID: PMC12050090 DOI: 10.1242/jcs.262255] [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: 05/04/2024] [Accepted: 12/20/2024] [Indexed: 03/27/2025] Open
Abstract
Double-strand breaks (DSBs) in DNA pose a critical threat to genomic integrity, potentially leading to the onset and progression of various diseases, including cancer. Cellular responses to such lesions entail sophisticated repair mechanisms primarily mediated by non-homologous end joining (NHEJ) and homologous recombination (HR). Interestingly, the efficient recruitment of repair proteins and completion of DSB repair likely involve complex, inter-organelle communication and coordination of cellular components. In this study, we report a role of γ-tubulin in DSB repair. γ-tubulin is a major microtubule nucleation factor governing microtubule dynamics. We show that γ-tubulin is recruited to the site of DNA damage and is required for efficient DSB repair via both NHEJ and HR. Suppression of γ-tubulin impedes DNA repair and exacerbates DNA damage accumulation. Furthermore, γ-tubulin mediates the mobilization and formation of DNA damage foci, which serve as repair centers, thereby facilitating the recruitment of HR and NHEJ repair proteins on damaged chromatin. Finally, pharmacological inhibition of γ-tubulin enhances the cytotoxic effect of DNA-damaging agents, consistent with the DNA repair function of γ-tubulin, and underscoring the potential of its therapeutic intervention in cancer therapy.
Collapse
Affiliation(s)
- Abhishikt David Solomon
- Division of Oral and Craniofacial Health Sciences, Adams School of Dentistry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Odjo G. Gouttia
- Division of Oral and Craniofacial Health Sciences, Adams School of Dentistry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Ling Wang
- Division of Oral and Craniofacial Health Sciences, Adams School of Dentistry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Songli Zhu
- Department of Oral Biology, University of Nebraska Medical Center, Lincoln, NE 68583, USA
| | - Feifei Wang
- Department of Oral Biology, University of Nebraska Medical Center, Lincoln, NE 68583, USA
| | - Yanqui Li
- Department of Oral Biology, University of Nebraska Medical Center, Lincoln, NE 68583, USA
| | - Mohammadjavad Paydar
- Institute for Research in Immunology and Cancer (IRIC), Département de médecine, Université de Montréal, Montréal H3C 3J7, Canada
| | - Tadayoshi Bessho
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Benjamin H. Kwok
- Department of Oral Biology, University of Nebraska Medical Center, Lincoln, NE 68583, USA
| | - Aimin Peng
- Division of Oral and Craniofacial Health Sciences, Adams School of Dentistry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
2
|
Schapfl MA, LoMastro GM, Braun VZ, Hirai M, Levine MS, Kiermaier E, Labi V, Holland AJ, Villunger A. Centrioles are frequently amplified in early B cell development but dispensable for humoral immunity. Nat Commun 2024; 15:8890. [PMID: 39406735 PMCID: PMC11480410 DOI: 10.1038/s41467-024-53222-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Accepted: 10/07/2024] [Indexed: 10/19/2024] Open
Abstract
Centrioles define centrosome structure and function. Deregulation of centriole numbers can cause developmental defects and cancer. The p53 tumor suppressor limits the growth of cells lacking or harboring additional centrosomes and can be engaged by the "mitotic surveillance" or the "PIDDosome pathway", respectively. Here, we show that early B cell progenitors frequently present extra centrioles, ensuing their high proliferative activity and related DNA damage. Extra centrioles are efficiently cleared during B cell maturation. In contrast, centriole loss upon Polo-like kinase 4 (Plk4) deletion causes apoptosis and arrests B cell development. This defect can be rescued by co-deletion of Usp28, a critical component of the mitotic surveillance pathway, that restores cell survival and maturation. Centriole-deficient mature B cells are proliferation competent and mount a humoral immune response. Our findings imply that progenitor B cells are intolerant to centriole loss but permissive to centriole amplification, a feature potentially facilitating their malignant transformation.
Collapse
Affiliation(s)
- Marina A Schapfl
- Institute for Developmental Immunology, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
| | - Gina M LoMastro
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Vincent Z Braun
- Institute for Developmental Immunology, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
| | - Maretoshi Hirai
- Department of Pharmacology, Kansai Medical University, Hirakata, Osaka, Japan
| | - Michelle S Levine
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Eva Kiermaier
- Life and Medical Sciences Institute, Immune and Tumor Biology, University of Bonn, Bonn, Germany
| | - Verena Labi
- Institute for Developmental Immunology, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
| | - Andrew J Holland
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Andreas Villunger
- Institute for Developmental Immunology, Biocenter, Medical University of Innsbruck, Innsbruck, Austria.
- The Research Center for Molecular Medicine (CeMM) of the Austrian Academy of Sciences, Vienna, Austria.
| |
Collapse
|
3
|
Zhang C, Wu BZ, Di Ciano-Oliveira C, Wu YF, Khavkine Binstock SS, Soria-Bretones I, Pham NA, Elia AJ, Chari R, Lam WL, Bray MR, Mak TW, Tsao MS, Cescon DW, Thu KL. Identification of KIFC1 as a putative vulnerability in lung cancers with centrosome amplification. Cancer Gene Ther 2024; 31:1559-1570. [PMID: 39179685 PMCID: PMC11489082 DOI: 10.1038/s41417-024-00824-1] [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: 01/20/2024] [Revised: 07/31/2024] [Accepted: 08/14/2024] [Indexed: 08/26/2024]
Abstract
Centrosome amplification (CA), an abnormal increase in the number of centrosomes in the cell, is a recurrent phenomenon in lung and other malignancies. Although CA promotes tumor development and progression by inducing genomic instability (GIN), it also induces mitotic stress that jeopardizes cellular integrity. CA leads to the formation of multipolar mitotic spindles that can cause lethal chromosome segregation errors. To sustain the benefits of CA by mitigating its consequences, malignant cells are dependent on adaptive mechanisms that represent therapeutic vulnerabilities. We aimed to discover genetic dependencies associated with CA in lung cancer. Combining a CRISPR/Cas9 functional genomics screen with tumor genomic analyses, we identified the motor protein KIFC1, also known as HSET, as a putative vulnerability specifically in lung adenocarcinoma (LUAD) with CA. KIFC1 expression was positively correlated with CA in LUAD and associated with worse patient outcomes, smoking history, and indicators of GIN. KIFC1 loss-of-function sensitized LUAD cells with high basal KIFC1 expression to potentiation of CA, which was associated with a diminished ability to cluster extra centrosomes into pseudo-bipolar mitotic spindles. Our work suggests that KIFC1 inhibition represents a novel approach for potentiating GIN to lethal levels in LUAD with CA by forcing cells to divide with multipolar spindles, rationalizing further studies to investigate its therapeutic potential.
Collapse
Affiliation(s)
- Christopher Zhang
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Unity Health Toronto, Toronto, Canada
| | - Benson Z Wu
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Unity Health Toronto, Toronto, Canada
| | - Caterina Di Ciano-Oliveira
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Unity Health Toronto, Toronto, Canada
| | - Yin Fang Wu
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Unity Health Toronto, Toronto, Canada
| | - Sharon S Khavkine Binstock
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Unity Health Toronto, Toronto, Canada
| | | | - Nhu-An Pham
- Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
| | - Andrew J Elia
- Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
| | - Raj Chari
- Laboratory Animal Sciences Program, Genome Modification Core, Frederick National Laboratory for Cancer Research, Frederick, USA
| | - Wan L Lam
- Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, Canada
| | - Mark R Bray
- Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
| | - Tak W Mak
- Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
- Department of Immunology, University of Toronto, Toronto, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Canada
| | - Ming-Sound Tsao
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
- Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Canada
| | - David W Cescon
- Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
- Institute of Medical Science, University of Toronto, Toronto, Canada
| | - Kelsie L Thu
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada.
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Unity Health Toronto, Toronto, Canada.
| |
Collapse
|
4
|
Stracker TH. Regulation of p53 by the mitotic surveillance/stopwatch pathway: implications in neurodevelopment and cancer. Front Cell Dev Biol 2024; 12:1451274. [PMID: 39398482 PMCID: PMC11466822 DOI: 10.3389/fcell.2024.1451274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 09/06/2024] [Indexed: 10/15/2024] Open
Abstract
The transcription factor p53 (encoded by TP53) plays diverse roles in human development and disease. While best known for its role in tumor suppression, p53 signaling also influences mammalian development by triggering cell fate decisions in response to a wide variety of stresses. After over 4 decades of study, a new pathway that triggers p53 activation in response to mitotic delays was recently identified. Termed the mitotic surveillance or mitotic stopwatch pathway, the USP28 and 53BP1 proteins activate p53 in response to delayed mitotic progression to control cell fate and promote genomic stability. In this Minireview, I discuss its identification, potential roles in neurodevelopmental disorders and cancer, as well as explore outstanding questions about its function, regulation and potential use as a biomarker for anti-mitotic therapies.
Collapse
Affiliation(s)
- Travis H. Stracker
- Center for Cancer Research, Radiation Oncology Branch, National Cancer Institute, Bethesda, MD, United States
| |
Collapse
|
5
|
Burlet D, Huber AL, Tissier A, Petrilli V. Crosstalk between inflammasome sensors and DNA damage response pathways. FEBS J 2024; 291:3978-3988. [PMID: 38273453 DOI: 10.1111/febs.17060] [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: 09/05/2023] [Revised: 12/04/2023] [Accepted: 01/10/2024] [Indexed: 01/27/2024]
Abstract
Eukaryotic cells encounter diverse threats jeopardizing their integrity, prompting the development of defense mechanisms against these stressors. Among these mechanisms, inflammasomes are well-known for their roles in coordinating the inflammatory response against infections. Extensive research has unveiled their multifaceted involvement in cellular processes beyond inflammation. Recent studies emphasize the intricate relationship between the inflammasome and the DNA damage response (DDR). They highlight how the DDR participates in inflammasome activation and the reciprocal impact of inflammasome on DDR and genome integrity preservation. Moreover, novel functions of inflammasome sensors in DDR pathways have emerged, broadening our understanding of their roles. Finally, this review delves into identifying common signals that drive the activation of inflammasome sensors alongside activation cues for the DNA damage response, offering potential insights into shared regulatory pathways between these critical cellular processes.
Collapse
Affiliation(s)
- Delphine Burlet
- INSERM U1052, Centre de Recherche en Cancérologie de Lyon, France
- CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, France
- Université de Lyon, Université Lyon 1, France
- Centre Léon Bérard, Lyon, France
| | - Anne-Laure Huber
- INSERM U1052, Centre de Recherche en Cancérologie de Lyon, France
- CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, France
- Université de Lyon, Université Lyon 1, France
- Centre Léon Bérard, Lyon, France
| | - Agnès Tissier
- INSERM U1052, Centre de Recherche en Cancérologie de Lyon, France
- CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, France
- Université de Lyon, Université Lyon 1, France
- Centre Léon Bérard, Lyon, France
| | - Virginie Petrilli
- INSERM U1052, Centre de Recherche en Cancérologie de Lyon, France
- CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, France
- Université de Lyon, Université Lyon 1, France
- Centre Léon Bérard, Lyon, France
| |
Collapse
|
6
|
Bai S, Martin-Sanchez F, Brough D, Lopez-Castejon G. Pyroptosis leads to loss of centrosomal integrity in macrophages. Cell Death Discov 2024; 10:354. [PMID: 39117604 PMCID: PMC11310477 DOI: 10.1038/s41420-024-02093-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 06/27/2024] [Accepted: 07/05/2024] [Indexed: 08/10/2024] Open
Abstract
NLRP3 forms a multiprotein inflammasome complex to initiate the inflammatory response when macrophages sense infection or tissue damage, which leads to caspase-1 activation, maturation and release of the inflammatory cytokines interleukin-1β (IL-1β) and IL-18 and Gasdermin-D (GSDMD) mediated pyroptosis. NLRP3 inflammasome activity must be controlled as unregulated and chronic inflammation underlies inflammatory and autoimmune diseases. Several findings uncovered that NLRP3 inflammasome activity is under the regulation of centrosome localized proteins such as NEK7 and HDAC6, however, whether the centrosome composition or structure is altered during the inflammasome activation is not known. Our data show that levels of the centrosomal scaffold protein pericentrin (PCNT) are reduced upon NLRP3 inflammasome activation via different activators in human and murine macrophages. PCNT loss occurs in the presence of membrane stabilizer punicalagin, suggesting this is not a consequence of membrane rupture. We found that PCNT loss is dependent on NLRP3 and active caspases as MCC950 and pan caspase inhibitor ZVAD prevent its degradation. Moreover, caspase-1 and GSDMD are both required for this NLRP3-mediated PCNT loss because absence of caspase-1 or GSDMD triggers an alternative regulation of PCNT via its cleavage by caspase-3 in response to nigericin stimulation. PCNT degradation occurs in response to nigericin, but also other NLRP3 activators including lysomotropic agent L-Leucyl-L-Leucine methyl ester (LLOMe) and hypotonicity but not AIM2 activation. Our work reveals that the NLRP3 inflammasome activation alters centrosome composition highlighting the need to further understand the role of this organelle during inflammatory responses.
Collapse
Affiliation(s)
- Siyi Bai
- Division of Infection, Immunity and Respiratory Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, M13 9PT, UK
- The Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, M13 9PT, UK
| | - Fatima Martin-Sanchez
- Division of Infection, Immunity and Respiratory Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, M13 9PT, UK
- The Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, M13 9PT, UK
- Department of Pharmacology, Faculty of Medicine, University of Murcia, Murcia, Spain
- Biomedical Research Institute of Murcia (IMIB-Pascual Parrilla), Faculty of Medicine, University of Murcia, 30120, Murcia, Spain
| | - David Brough
- The Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, M13 9PT, UK
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
- Geoffrey Jefferson Brain Research Centre, The Manchester Academic Health Science Centre, Northern Care Alliance NHS Group, University of Manchester, Manchester, UK
| | - Gloria Lopez-Castejon
- Division of Infection, Immunity and Respiratory Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, M13 9PT, UK.
- The Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, M13 9PT, UK.
| |
Collapse
|
7
|
Jovasevic V, Wood EM, Cicvaric A, Zhang H, Petrovic Z, Carboncino A, Parker KK, Bassett TE, Moltesen M, Yamawaki N, Login H, Kalucka J, Sananbenesi F, Zhang X, Fischer A, Radulovic J. Formation of memory assemblies through the DNA-sensing TLR9 pathway. Nature 2024; 628:145-153. [PMID: 38538785 PMCID: PMC10990941 DOI: 10.1038/s41586-024-07220-7] [Citation(s) in RCA: 34] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 02/21/2024] [Indexed: 04/05/2024]
Abstract
As hippocampal neurons respond to diverse types of information1, a subset assembles into microcircuits representing a memory2. Those neurons typically undergo energy-intensive molecular adaptations, occasionally resulting in transient DNA damage3-5. Here we found discrete clusters of excitatory hippocampal CA1 neurons with persistent double-stranded DNA (dsDNA) breaks, nuclear envelope ruptures and perinuclear release of histone and dsDNA fragments hours after learning. Following these early events, some neurons acquired an inflammatory phenotype involving activation of TLR9 signalling and accumulation of centrosomal DNA damage repair complexes6. Neuron-specific knockdown of Tlr9 impaired memory while blunting contextual fear conditioning-induced changes of gene expression in specific clusters of excitatory CA1 neurons. Notably, TLR9 had an essential role in centrosome function, including DNA damage repair, ciliogenesis and build-up of perineuronal nets. We demonstrate a novel cascade of learning-induced molecular events in discrete neuronal clusters undergoing dsDNA damage and TLR9-mediated repair, resulting in their recruitment to memory circuits. With compromised TLR9 function, this fundamental memory mechanism becomes a gateway to genomic instability and cognitive impairments implicated in accelerated senescence, psychiatric disorders and neurodegenerative disorders. Maintaining the integrity of TLR9 inflammatory signalling thus emerges as a promising preventive strategy for neurocognitive deficits.
Collapse
Affiliation(s)
- Vladimir Jovasevic
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Elizabeth M Wood
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Ana Cicvaric
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Hui Zhang
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Zorica Petrovic
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Anna Carboncino
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Kendra K Parker
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Thomas E Bassett
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Maria Moltesen
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- PROMEMO, Aarhus University, Aarhus, Denmark
- DANDRITE, Aarhus University, Aarhus, Denmark
| | - Naoki Yamawaki
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- PROMEMO, Aarhus University, Aarhus, Denmark
- DANDRITE, Aarhus University, Aarhus, Denmark
| | - Hande Login
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- PROMEMO, Aarhus University, Aarhus, Denmark
- DANDRITE, Aarhus University, Aarhus, Denmark
| | - Joanna Kalucka
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- PROMEMO, Aarhus University, Aarhus, Denmark
- DANDRITE, Aarhus University, Aarhus, Denmark
| | - Farahnaz Sananbenesi
- Department for Psychiatry and Psychotherapy, German Center for Neurodegenerative Diseases, University Medical Center, Göttingen, Germany
- Cluster of Excellence MBExC, University of Göttingen, Göttingen, Germany
| | - Xusheng Zhang
- Computational Genomics Core, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Andre Fischer
- Department for Psychiatry and Psychotherapy, German Center for Neurodegenerative Diseases, University Medical Center, Göttingen, Germany
- Cluster of Excellence MBExC, University of Göttingen, Göttingen, Germany
| | - Jelena Radulovic
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA.
- Department of Biomedicine, Aarhus University, Aarhus, Denmark.
- PROMEMO, Aarhus University, Aarhus, Denmark.
- DANDRITE, Aarhus University, Aarhus, Denmark.
- Department of Psychiatry and Behavioral Sciences, Psychiatry Research Institute Montefiore Einstein (PRIME), Albert Einstein College of Medicine, Bronx, NY, USA.
| |
Collapse
|
8
|
Rodríguez‐Real G, Domínguez‐Calvo A, Prados‐Carvajal R, Bayona‐Feliú A, Gomes‐Pereira S, Balestra FR, Huertas P. Centriolar subdistal appendages promote double-strand break repair through homologous recombination. EMBO Rep 2023; 24:e56724. [PMID: 37664992 PMCID: PMC10561181 DOI: 10.15252/embr.202256724] [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: 12/23/2022] [Revised: 07/18/2023] [Accepted: 08/19/2023] [Indexed: 09/05/2023] Open
Abstract
The centrosome is a cytoplasmic organelle with roles in microtubule organization that has also been proposed to act as a hub for cellular signaling. Some centrosomal components are required for full activation of the DNA damage response. However, whether the centrosome regulates specific DNA repair pathways is not known. Here, we show that centrosome presence is required to fully activate recombination, specifically to completely license its initial step, the so-called DNA end resection. Furthermore, we identify a centriolar structure, the subdistal appendages, and a specific factor, CEP170, as the critical centrosomal component involved in the regulation of recombination and resection. Cells lacking centrosomes or depleted for CEP170 are, consequently, hypersensitive to DNA damaging agents. Moreover, low levels of CEP170 in multiple cancer types correlate with an increase of the mutation burden associated with specific mutational signatures and a better prognosis, suggesting that changes in CEP170 can act as a mutation driver but could also be targeted to improve current oncological treatments.
Collapse
Affiliation(s)
- Guillermo Rodríguez‐Real
- Departamento de Genética, Facultad de BiologíaUniversidad de SevillaSevillaSpain
- Centro Andaluz de Biología Molecular y Medicina Regenerativa‐CABIMERUniversidad de Sevilla‐CSIC‐Universidad Pablo de OlavideSevillaSpain
| | - Andrés Domínguez‐Calvo
- Departamento de Genética, Facultad de BiologíaUniversidad de SevillaSevillaSpain
- Centro Andaluz de Biología Molecular y Medicina Regenerativa‐CABIMERUniversidad de Sevilla‐CSIC‐Universidad Pablo de OlavideSevillaSpain
| | - Rosario Prados‐Carvajal
- Departamento de Genética, Facultad de BiologíaUniversidad de SevillaSevillaSpain
- Centro Andaluz de Biología Molecular y Medicina Regenerativa‐CABIMERUniversidad de Sevilla‐CSIC‐Universidad Pablo de OlavideSevillaSpain
| | - Aleix Bayona‐Feliú
- Genome Data Science, Institute for Research in Biomedicine (IRB Barcelona)The Barcelona Institute of Science and Technology (BIST)BarcelonaSpain
| | - Sonia Gomes‐Pereira
- Department of Cell Biology, Sciences IIIUniversity of GenevaGenevaSwitzerland
| | - Fernando R Balestra
- Departamento de Genética, Facultad de BiologíaUniversidad de SevillaSevillaSpain
- Centro Andaluz de Biología Molecular y Medicina Regenerativa‐CABIMERUniversidad de Sevilla‐CSIC‐Universidad Pablo de OlavideSevillaSpain
| | - Pablo Huertas
- Departamento de Genética, Facultad de BiologíaUniversidad de SevillaSevillaSpain
- Centro Andaluz de Biología Molecular y Medicina Regenerativa‐CABIMERUniversidad de Sevilla‐CSIC‐Universidad Pablo de OlavideSevillaSpain
| |
Collapse
|
9
|
Song H, Kim EH, Hong J, Gwon D, Kim JW, Bae GU, Jang CY. Hornerin mediates phosphorylation of the polo-box domain in Plk1 by Chk1 to induce death in mitosis. Cell Death Differ 2023; 30:2151-2166. [PMID: 37596441 PMCID: PMC10482915 DOI: 10.1038/s41418-023-01208-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 05/25/2023] [Accepted: 06/13/2023] [Indexed: 08/20/2023] Open
Abstract
The centrosome assembles a bipolar spindle for faithful chromosome segregation during mitosis. To prevent the inheritance of DNA damage, the DNA damage response (DDR) triggers programmed spindle multipolarity and concomitant death in mitosis through a poorly understood mechanism. We identified hornerin, which forms a complex with checkpoint kinase 1 (Chk1) and polo-like kinase 1 (Plk1) to mediate phosphorylation at the polo-box domain (PBD) of Plk1, as the link between the DDR and death in mitosis. We demonstrate that hornerin mediates DDR-induced precocious centriole disengagement through a dichotomous mechanism that includes sequestration of Sgo1 and Plk1 in the cytoplasm through phosphorylation of the PBD in Plk1 by Chk1. Phosphorylation of the PBD in Plk1 abolishes the interaction with Sgo1 and phosphorylation-dependent Sgo1 translocation to the centrosome, leading to precocious centriole disengagement and spindle multipolarity. Mechanistically, hornerin traps phosphorylated Plk1 in the cytoplasm. Furthermore, PBD phosphorylation inactivates Plk1 and disrupts Cep192::Aurora A::Plk1 complex translocation to the centrosome and concurrent centrosome maturation. Remarkably, hornerin depletion leads to chemoresistance against DNA damaging agents by attenuating DDR-induced death in mitosis. These results reveal how the DDR eradicates mitotic cells harboring DNA damage to ensure genome integrity during cell division.
Collapse
Affiliation(s)
- Haiyu Song
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Sookmyung Women's University, Seoul, 04310, Republic of Korea
| | - Eun Ho Kim
- Department of Biochemistry, School of Medicine, Catholic University of Daegu, Daegu, 42472, Republic of Korea
| | - Jihee Hong
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Sookmyung Women's University, Seoul, 04310, Republic of Korea
| | - Dasom Gwon
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Sookmyung Women's University, Seoul, 04310, Republic of Korea
| | - Jee Won Kim
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Sookmyung Women's University, Seoul, 04310, Republic of Korea
| | - Gyu-Un Bae
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Sookmyung Women's University, Seoul, 04310, Republic of Korea.
| | - Chang-Young Jang
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Sookmyung Women's University, Seoul, 04310, Republic of Korea.
| |
Collapse
|
10
|
Bloomfield M, Cimini D. The fate of extra centrosomes in newly formed tetraploid cells: should I stay, or should I go? Front Cell Dev Biol 2023; 11:1210983. [PMID: 37576603 PMCID: PMC10413984 DOI: 10.3389/fcell.2023.1210983] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Accepted: 07/17/2023] [Indexed: 08/15/2023] Open
Abstract
An increase in centrosome number is commonly observed in cancer cells, but the role centrosome amplification plays along with how and when it occurs during cancer development is unclear. One mechanism for generating cancer cells with extra centrosomes is whole genome doubling (WGD), an event that occurs in over 30% of human cancers and is associated with poor survival. Newly formed tetraploid cells can acquire extra centrosomes during WGD, and a generally accepted model proposes that centrosome amplification in tetraploid cells promotes cancer progression by generating aneuploidy and chromosomal instability. Recent findings, however, indicate that newly formed tetraploid cells in vitro lose their extra centrosomes to prevent multipolar cell divisions. Rather than persistent centrosome amplification, this evidence raises the possibility that it may be advantageous for tetraploid cells to initially restore centrosome number homeostasis and for a fraction of the population to reacquire additional centrosomes in the later stages of cancer evolution. In this review, we explore the different evolutionary paths available to newly formed tetraploid cells, their effects on centrosome and chromosome number distribution in daughter cells, and their probabilities of long-term survival. We then discuss the mechanisms that may alter centrosome and chromosome numbers in tetraploid cells and their relevance to cancer progression following WGD.
Collapse
Affiliation(s)
- Mathew Bloomfield
- Department of Biological Sciences and Fralin Life Sciences Institute, Virginia Tech, Blacksburg, VA, United States
| | - Daniela Cimini
- Department of Biological Sciences and Fralin Life Sciences Institute, Virginia Tech, Blacksburg, VA, United States
| |
Collapse
|
11
|
Qi H, Kikuchi M, Yoshino Y, Fang Z, Ohashi K, Gotoh T, Ideta R, Ui A, Endo S, Otsuka K, Shindo N, Gonda K, Ishioka C, Miki Y, Iwabuchi T, Chiba N. BRCA1 transports the DNA damage signal for CDDP-induced centrosome amplification through the centrosomal Aurora A. Cancer Sci 2022; 113:4230-4243. [PMID: 36082621 PMCID: PMC9746055 DOI: 10.1111/cas.15573] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 08/25/2022] [Accepted: 08/26/2022] [Indexed: 12/15/2022] Open
Abstract
Breast cancer gene 1 (BRCA1) plays roles in DNA repair and centrosome regulation and is involved in DNA damage-induced centrosome amplification (DDICA). Here, the centrosomal localization of BRCA1 and the kinases involved in centrosome duplication were analyzed in each cell cycle phase after treatment with DNA crosslinker cisplatin (CDDP). CDDP treatment increased the centrosomal localization of BRCA1 in early S-G2 phase. BRCA1 contributed to the increased centrosomal localization of Aurora A in S phase and that of phosphorylated Polo-like kinase 1 (PLK1) in late S phase after CDDP treatment, resulting in centriole disengagement and overduplication. The increased centrosomal localization of BRCA1 and Aurora A induced by CDDP treatment involved the nuclear export of BRCA1 and BRCA1 phosphorylation by ataxia telangiectasia mutated (ATM). Patient-derived variants and mutations at phosphorylated residues of BRCA1 suppressed the interaction between BRCA1 and Aurora A, as well as the CDDP-induced increase in the centrosomal localization of BRCA1 and Aurora A. These results suggest that CDDP induces the phosphorylation of BRCA1 by ATM in the nucleus and its transport to the cytoplasm, thereby promoting the centrosomal localization Aurora A, which phosphorylates PLK1. The function of BRCA1 in the translocation of the DNA damage signal from the nucleus to the centrosome to induce centrosome amplification after CDDP treatment might support its role as a tumor suppressor.
Collapse
Affiliation(s)
- Huicheng Qi
- Department of Cancer Biology; Institute of Development, Aging and CancerTohoku UniversitySendaiJapan
- Department of Cancer BiologyTohoku University Graduate School of MedicineSendaiJapan
| | - Megumi Kikuchi
- Department of Cancer Biology; Institute of Development, Aging and CancerTohoku UniversitySendaiJapan
- Laboratory of Cancer Biology, Graduate School of Life SciencesTohoku UniversitySendaiJapan
| | - Yuki Yoshino
- Department of Cancer Biology; Institute of Development, Aging and CancerTohoku UniversitySendaiJapan
- Department of Cancer BiologyTohoku University Graduate School of MedicineSendaiJapan
- Laboratory of Cancer Biology, Graduate School of Life SciencesTohoku UniversitySendaiJapan
| | - Zhenzhou Fang
- Department of Cancer Biology; Institute of Development, Aging and CancerTohoku UniversitySendaiJapan
- Department of Cancer BiologyTohoku University Graduate School of MedicineSendaiJapan
| | - Kazune Ohashi
- Department of Cancer Biology; Institute of Development, Aging and CancerTohoku UniversitySendaiJapan
- Laboratory of Cancer Biology, Graduate School of Life SciencesTohoku UniversitySendaiJapan
| | - Takato Gotoh
- Department of Cancer Biology; Institute of Development, Aging and CancerTohoku UniversitySendaiJapan
- Laboratory of Cancer Biology, Graduate School of Life SciencesTohoku UniversitySendaiJapan
| | - Ryo Ideta
- Department of Cancer Biology; Institute of Development, Aging and CancerTohoku UniversitySendaiJapan
- Tohoku University School of MedicineSendaiJapan
| | - Ayako Ui
- Department of Molecular Oncology, Institute of Development, Aging and CancerTohoku UniversitySendaiJapan
| | - Shino Endo
- Department of Cancer Biology; Institute of Development, Aging and CancerTohoku UniversitySendaiJapan
- Department of Cancer BiologyTohoku University Graduate School of MedicineSendaiJapan
| | - Kei Otsuka
- Department of Cancer Biology; Institute of Development, Aging and CancerTohoku UniversitySendaiJapan
- Laboratory of Cancer Biology, Graduate School of Life SciencesTohoku UniversitySendaiJapan
| | - Norihisa Shindo
- Division of Molecular and Cellular OncologyMiyagi Cancer Center Research InstituteNatoriJapan
| | - Kohsuke Gonda
- Department of Medical PhysicsTohoku University Graduate School of MedicineSendaiJapan
| | - Chikashi Ishioka
- Department of Clinical OncologyTohoku University Graduate School of MedicineSendaiJapan
| | - Yoshio Miki
- Department of Molecular Genetics, Medical Research InstituteTokyo Medical and Dental UniversityTokyoJapan
| | - Tokuro Iwabuchi
- Faculty of Bioscience and BiotechnologyTokyo University of TechnologyTokyoJapan
| | - Natsuko Chiba
- Department of Cancer Biology; Institute of Development, Aging and CancerTohoku UniversitySendaiJapan
- Department of Cancer BiologyTohoku University Graduate School of MedicineSendaiJapan
- Laboratory of Cancer Biology, Graduate School of Life SciencesTohoku UniversitySendaiJapan
| |
Collapse
|
12
|
Lu JY, Simon M, Zhao Y, Ablaeva J, Corson N, Choi Y, Yamada KYH, Schork NJ, Hood WR, Hill GE, Miller RA, Seluanov A, Gorbunova V. Comparative transcriptomics reveals circadian and pluripotency networks as two pillars of longevity regulation. Cell Metab 2022; 34:836-856.e5. [PMID: 35580607 PMCID: PMC9364679 DOI: 10.1016/j.cmet.2022.04.011] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 02/24/2022] [Accepted: 04/22/2022] [Indexed: 01/24/2023]
Abstract
Mammals differ more than 100-fold in maximum lifespan. Here, we conducted comparative transcriptomics on 26 species with diverse lifespans. We identified thousands of genes with expression levels negatively or positively correlated with a species' maximum lifespan (Neg- or Pos-MLS genes). Neg-MLS genes are primarily involved in energy metabolism and inflammation. Pos-MLS genes show enrichment in DNA repair, microtubule organization, and RNA transport. Expression of Neg- and Pos-MLS genes is modulated by interventions, including mTOR and PI3K inhibition. Regulatory networks analysis showed that Neg-MLS genes are under circadian regulation possibly to avoid persistent high expression, whereas Pos-MLS genes are targets of master pluripotency regulators OCT4 and NANOG and are upregulated during somatic cell reprogramming. Pos-MLS genes are highly expressed during embryogenesis but significantly downregulated after birth. This work provides targets for anti-aging interventions by defining pathways correlating with longevity across mammals and uncovering circadian and pluripotency networks as central regulators of longevity.
Collapse
Affiliation(s)
- J Yuyang Lu
- Department of Biology, University of Rochester, Rochester, NY 14627, USA
| | - Matthew Simon
- Department of Biology, University of Rochester, Rochester, NY 14627, USA
| | - Yang Zhao
- Department of Biology, University of Rochester, Rochester, NY 14627, USA
| | - Julia Ablaeva
- Department of Biology, University of Rochester, Rochester, NY 14627, USA
| | - Nancy Corson
- Department of Biology, University of Rochester, Rochester, NY 14627, USA
| | - Yongwook Choi
- Quantitative Medicine and Systems Biology Division, Translational Genomics Research Institute, Phoenix, AZ 85004, USA
| | - KayLene Y H Yamada
- Department of Biological Sciences, Auburn University, Auburn, AL 36849, USA
| | - Nicholas J Schork
- Quantitative Medicine and Systems Biology Division, Translational Genomics Research Institute, Phoenix, AZ 85004, USA
| | - Wendy R Hood
- Department of Biological Sciences, Auburn University, Auburn, AL 36849, USA
| | - Geoffrey E Hill
- Department of Biological Sciences, Auburn University, Auburn, AL 36849, USA
| | - Richard A Miller
- Department of Pathology and Geriatrics Center, University of Michigan, Ann Arbor, MI 48109, USA
| | - Andrei Seluanov
- Department of Biology, University of Rochester, Rochester, NY 14627, USA.
| | - Vera Gorbunova
- Department of Biology, University of Rochester, Rochester, NY 14627, USA.
| |
Collapse
|
13
|
Wang X, Xu C, Sun H. DNA Damage Repair-Related Genes Signature for Immune Infiltration and Outcome in Cervical Cancer. Front Genet 2022; 13:733164. [PMID: 35309134 PMCID: PMC8927729 DOI: 10.3389/fgene.2022.733164] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 02/08/2022] [Indexed: 12/11/2022] Open
Abstract
Background: The mechanism of DNA damage repair plays an important role in many solid tumors represented by cervical cancer. Purpose: The purpose of this study was to explore the effect of DNA damage repair-related genes on immune function of patients with cervical cancer, and to establish and evaluate a prognosis model based on DNA damage repair-related genes. Methods: In the study, we analyzed the genes related to DNA damage and repair, and obtained two subtypes (F1 and F2). We selected two groups of samples for different selection, and studied which pathways were enriched expression. For different subtypes, the immune score was explored to explain immune infiltration. We got the key genes through screening, and established the prognosis model through the key genes. These 11 key genes were correlated with the expression of common Clusters of Differentiation (CD) genes in order to explore the effects of these genes on immunity. Results: Through the Least absolute shrinkage and selection operator (LASSO) method, we screened 11 genes from 232 candidate genes as the key genes for the prognosis score. Through the Kaplan-Meier method, four genes (HAP1, MCM5, RNASEH2A, CETN2) with significant prognostic significance were screened into the final model, forming a Nomogram with C-index of 0.716 (0.649–1.0). Conclusion: In cervical cancer, DNA damage repair related genes and immune cell infection characteristics have certain association, and DNA damage repair related genes and immune cell infection characteristics can effectively predict the prognosis.
Collapse
Affiliation(s)
- Xinghao Wang
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Chen Xu
- Department of Surgical Oncology, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Hongzan Sun
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, China
- *Correspondence: Hongzan Sun,
| |
Collapse
|
14
|
Floriot S, Bellutti L, Castille J, Moison P, Messiaen S, Passet B, Boulanger L, Boukadiri A, Tourpin S, Beauvallet C, Vilotte M, Riviere J, Péchoux C, Bertaud M, Vilotte JL, Livera G. CEP250 is Required for Maintaining Centrosome Cohesion in the Germline and Fertility in Male Mice. Front Cell Dev Biol 2022; 9:754054. [PMID: 35127699 PMCID: PMC8809461 DOI: 10.3389/fcell.2021.754054] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 12/24/2021] [Indexed: 12/02/2022] Open
Abstract
Male gametogenesis involves both mitotic divisions to amplify germ cell progenitors that gradually differentiate and meiotic divisions. Centrosomal regulation is essential for both types of divisions, with centrioles remaining tightly paired during the interphase. Here, we generated and characterized the phenotype of mutant mice devoid of Cep250/C-Nap1, a gene encoding for a docking protein for fibers linking centrioles, and characterized their phenotype. The Cep250-/- mice presented with no major defects, apart from male infertility due to a reduction in the spermatogonial pool and the meiotic blockade. Spermatogonial stem cells expressing Zbtb16 were not affected, whereas the differentiating spermatogonia were vastly lost. These cells displayed abnormal γH2AX-staining, accompanied by an increase in the apoptotic rate. The few germ cells that survived at this stage, entered the meiotic prophase I and were arrested at a pachytene-like stage, likely due to synapsis defects and the unrepaired DNA double-strand breaks. In these cells, centrosomes split up precociously, with γ-tubulin foci being separated whereas these were closely associated in wild-type cells. Interestingly, this lack of cohesion was also observed in wild-type female meiocytes, likely explaining the normal fertility of Cep250-/- female mice. Taken together, this study proposes a specific requirement of centrosome cohesion in the male germline, with a crucial role of CEP250 in both differentiating spermatogonia and meiotic spermatocytes.
Collapse
Affiliation(s)
- Sandrine Floriot
- INRAe, AgroParisTech, Université Paris-Saclay, GABI, Jouy-en-Josas, France
| | - Laura Bellutti
- Laboratory of Development of the Gonads, UMRE008 Genetic Stability Stem Cells and Radiations, IRCM/IBFJ CEA, Université de Paris, Université Paris-Saclay, Paris, France
- *Correspondence: Laura Bellutti, ; Gabriel Livera,
| | - Johan Castille
- INRAe, AgroParisTech, Université Paris-Saclay, GABI, Jouy-en-Josas, France
| | - Pauline Moison
- Laboratory of Development of the Gonads, UMRE008 Genetic Stability Stem Cells and Radiations, IRCM/IBFJ CEA, Université de Paris, Université Paris-Saclay, Paris, France
| | - Sébastien Messiaen
- Laboratory of Development of the Gonads, UMRE008 Genetic Stability Stem Cells and Radiations, IRCM/IBFJ CEA, Université de Paris, Université Paris-Saclay, Paris, France
| | - Bruno Passet
- INRAe, AgroParisTech, Université Paris-Saclay, GABI, Jouy-en-Josas, France
| | - Laurent Boulanger
- INRAe, AgroParisTech, Université Paris-Saclay, GABI, Jouy-en-Josas, France
| | - Abdelhak Boukadiri
- INRAe, AgroParisTech, Université Paris-Saclay, GABI, Jouy-en-Josas, France
| | - Sophie Tourpin
- Laboratory of Development of the Gonads, UMRE008 Genetic Stability Stem Cells and Radiations, IRCM/IBFJ CEA, Université de Paris, Université Paris-Saclay, Paris, France
| | | | - Marthe Vilotte
- INRAe, AgroParisTech, Université Paris-Saclay, GABI, Jouy-en-Josas, France
| | - Julie Riviere
- INRAe, AgroParisTech, Université Paris-Saclay, GABI, Jouy-en-Josas, France
| | - Christine Péchoux
- INRAe, AgroParisTech, Université Paris-Saclay, GABI, Jouy-en-Josas, France
| | - Maud Bertaud
- INRAe, AgroParisTech, Université Paris-Saclay, GABI, Jouy-en-Josas, France
| | - Jean-Luc Vilotte
- INRAe, AgroParisTech, Université Paris-Saclay, GABI, Jouy-en-Josas, France
| | - Gabriel Livera
- Laboratory of Development of the Gonads, UMRE008 Genetic Stability Stem Cells and Radiations, IRCM/IBFJ CEA, Université de Paris, Université Paris-Saclay, Paris, France
- *Correspondence: Laura Bellutti, ; Gabriel Livera,
| |
Collapse
|
15
|
Gupta S, Ozimek-Kulik JE, Phillips JK. Nephronophthisis-Pathobiology and Molecular Pathogenesis of a Rare Kidney Genetic Disease. Genes (Basel) 2021; 12:genes12111762. [PMID: 34828368 PMCID: PMC8623546 DOI: 10.3390/genes12111762] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 10/26/2021] [Accepted: 10/27/2021] [Indexed: 12/17/2022] Open
Abstract
The exponential rise in our understanding of the aetiology and pathophysiology of genetic cystic kidney diseases can be attributed to the identification of cystogenic genes over the last three decades. The foundation of this was laid by positional cloning strategies which gradually shifted towards next-generation sequencing (NGS) based screenings. This shift has enabled the discovery of novel cystogenic genes at an accelerated pace unlike ever before and, most notably, the past decade has seen the largest increase in identification of the genes which cause nephronophthisis (NPHP). NPHP is a monogenic autosomal recessive cystic kidney disease caused by mutations in a diverse clade of over 26 identified genes and is the most common genetic cause of renal failure in children. NPHP gene types present with some common pathophysiological features alongside a diverse range of extra-renal phenotypes associated with specific syndromic presentations. This review provides a timely update on our knowledge of this disease, including epidemiology, pathophysiology, anatomical and molecular features. We delve into the diversity of the NPHP causing genes and discuss known molecular mechanisms and biochemical pathways that may have possible points of intersection with polycystic kidney disease (the most studied renal cystic pathology). We delineate the pathologies arising from extra-renal complications and co-morbidities and their impact on quality of life. Finally, we discuss the current diagnostic and therapeutic modalities available for disease management, outlining possible avenues of research to improve the prognosis for NPHP patients.
Collapse
Affiliation(s)
- Shabarni Gupta
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW 2109, Australia; (J.E.O.-K.); (J.K.P.)
- Correspondence:
| | - Justyna E. Ozimek-Kulik
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW 2109, Australia; (J.E.O.-K.); (J.K.P.)
- School of Women’s and Children’s Health, University of New South Wales, Sydney, NSW 2031, Australia
- Department of Paediatric Nephrology, Sydney Children’s Hospital Network, Children’s Hospital at Westmead, Sydney, NSW 2145, Australia
| | - Jacqueline Kathleen Phillips
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW 2109, Australia; (J.E.O.-K.); (J.K.P.)
| |
Collapse
|
16
|
Akagawa R, Nabeshima YI, Kawauchi T. Alternative Functions of Cell Cycle-Related and DNA Repair Proteins in Post-mitotic Neurons. Front Cell Dev Biol 2021; 9:753175. [PMID: 34746147 PMCID: PMC8564117 DOI: 10.3389/fcell.2021.753175] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 09/28/2021] [Indexed: 11/13/2022] Open
Abstract
Proper regulation of neuronal morphological changes is essential for neuronal migration, maturation, synapse formation, and high-order function. Many cytoplasmic proteins involved in the regulation of neuronal microtubules and the actin cytoskeleton have been identified. In addition, some nuclear proteins have alternative functions in neurons. While cell cycle-related proteins basically control the progression of the cell cycle in the nucleus, some of them have an extra-cell cycle-regulatory function (EXCERF), such as regulating cytoskeletal organization, after exit from the cell cycle. Our expression analyses showed that not only cell cycle regulators, including cyclin A1, cyclin D2, Cdk4/6, p21cip1, p27kip1, Ink4 family, and RAD21, but also DNA repair proteins, including BRCA2, p53, ATM, ATR, RAD17, MRE11, RAD9, and Hus1, were expressed after neurogenesis, suggesting that these proteins have alternative functions in post-mitotic neurons. In this perspective paper, we discuss the alternative functions of the nuclear proteins in neuronal development, focusing on possible cytoplasmic roles.
Collapse
Affiliation(s)
- Remi Akagawa
- Laboratory of Molecular Life Science, Institute of Biomedical Research and Innovation, Foundation for Biomedical Research and Innovation at Kobe (FBRI), Kobe, Japan
| | - Yo-ichi Nabeshima
- Laboratory of Molecular Life Science, Institute of Biomedical Research and Innovation, Foundation for Biomedical Research and Innovation at Kobe (FBRI), Kobe, Japan
| | - Takeshi Kawauchi
- Laboratory of Molecular Life Science, Institute of Biomedical Research and Innovation, Foundation for Biomedical Research and Innovation at Kobe (FBRI), Kobe, Japan
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
17
|
De novo Assembly and Analysis of Tissue-Specific Transcriptomes of the Edible Red Sea Urchin Loxechinus albus Using RNA-Seq. BIOLOGY 2021; 10:biology10100995. [PMID: 34681094 PMCID: PMC8533317 DOI: 10.3390/biology10100995] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 09/27/2021] [Accepted: 09/29/2021] [Indexed: 02/07/2023]
Abstract
Simple Summary Edible red sea urchin (Loxechinus albus) is an endemic species of echinoderm distributed along the Chilean coasts. This resource has been overexploited in recent years, depleting their natural populations. At present, there are few reported gene sequences available in public databases, restricting the molecular studies associated with aquaculture for this species. The aim of this study was to present the first annotated reference transcriptome of L. albus using NGS technologies and the differential expression transcripts analysis of the evaluated tissues. The transcriptome data obtained in this study will serve as a reference for future molecular research in the edible red sea urchin and other sea urchin species. Abstract Edible red sea urchin (Loxechinus albus) is an endemic echinoderm species of the Chilean coasts. The worldwide demand for high-quality gonads of this species has addressed the depletion of its natural populations. Studies on this sea urchin are limited, and genomic information is almost nonexistent. Hence, generate a transcriptome is crucial information that will considerably enrich molecular data and promote future findings for the L. albus aquaculture. Here, we obtained transcriptomic data of the edible red sea urchin by Illumina platform. Total RNA was extracted from gonads, intestines, and coelomocytes of juvenile urchins, and samples were sequenced using MiSeq Illumina technology. A total of 91,119,300 paired-end reads were de novo assembled, 185,239 transcripts produced, and a reference transcriptome created with 38.8% GC content and an N50 of 1769 bp. Gene ontology analysis revealed notable differences in the expression profiles between gonads, intestines, and coelomocytes, allowing the detection of transcripts associated with specific biological processes and KEGG pathways. These data were validated using 12 candidate transcripts by real-time qPCR. This dataset will provide a valuable molecular resource for L. albus and other species of sea urchins.
Collapse
|
18
|
Lovera M, Lüders J. The ciliary impact of nonciliary gene mutations. Trends Cell Biol 2021; 31:876-887. [PMID: 34183231 DOI: 10.1016/j.tcb.2021.06.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 05/31/2021] [Accepted: 06/03/2021] [Indexed: 01/15/2023]
Abstract
Mutations in genes encoding centriolar or ciliary proteins cause diseases collectively known as 'ciliopathies'. Interestingly, the Human Phenotype Ontology database lists numerous disorders that display clinical features reminiscent of ciliopathies but do not involve defects in the centriole-cilium proteome. Instead, defects in different cellular compartments may impair cilia indirectly and cause additional, nonciliopathy phenotypes. This phenotypic heterogeneity, perhaps combined with the field's centriole-cilium-centric view, may have hindered the recognition of ciliary contributions. Identifying these diseases and dissecting how the underlying gene mutations impair cilia not only will add to our understanding of cilium assembly and function but also may open up new therapeutic avenues.
Collapse
Affiliation(s)
- Marta Lovera
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Baldiri Reixac 10, 08028 Barcelona, Spain
| | - Jens Lüders
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Baldiri Reixac 10, 08028 Barcelona, Spain.
| |
Collapse
|
19
|
Etoposide Triggers Cellular Senescence by Inducing Multiple Centrosomes and Primary Cilia in Adrenocortical Tumor Cells. Cells 2021; 10:cells10061466. [PMID: 34208028 PMCID: PMC8230646 DOI: 10.3390/cells10061466] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Revised: 06/05/2021] [Accepted: 06/09/2021] [Indexed: 11/17/2022] Open
Abstract
Etoposide (ETO) has been used in treating adrenocortical tumor (ACT) cells. Our previous study showed that ETO inhibits ACT cell growth. In the present study, we show that ETO treatment at IC50 (10 μM) inhibited ACT cell growth by inducing cellular senescence rather than apoptosis. Several markers of cellular senescence, including enlarged nuclei, activated senescence-associated β-galactosidase activity, elevated levels of p53 and p21, and down-regulation of Lamin B1, were observed. We further found that ETO induced multiple centrosomes. The inhibition of multiple centrosomes accomplished by treating cells with either roscovitine or centrinone or through the overexpression of NR5A1/SF-1 alleviated ETO-induced senescence, suggesting that ETO triggered senescence via multiple centrosomes. Primary cilia also played a role in ETO-induced senescence. In the mechanism, DNA-PK-Chk2 signaling was activated by ETO treatment; inhibition of this signaling cascade alleviated multiple ETO-induced centrosomes and primary cilia followed by reducing cellular senescence. In addition to DNA damage signaling, autophagy was also triggered by ETO treatment for centrosomal events and senescence. Importantly, the inactivation of DNA-PK-Chk2 signaling reduced ETO-triggered autophagy; however, the inhibition of autophagy did not affect DNA-PK-Chk2 activation. Thus, ETO activated the DNA-PK-Chk2 cascade to facilitate autophagy. The activated autophagy further induced multiple centrosomes and primary cilia followed by triggering senescence.
Collapse
|
20
|
Flynn M, Whitton L, Donohoe G, Morrison CG, Morris DW. Altered gene regulation as a candidate mechanism by which ciliopathy gene SDCCAG8 contributes to schizophrenia and cognitive function. Hum Mol Genet 2021; 29:407-417. [PMID: 31868218 DOI: 10.1093/hmg/ddz292] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 11/29/2019] [Accepted: 12/02/2019] [Indexed: 12/14/2022] Open
Abstract
Mutations in genes that encode centrosomal/ciliary proteins cause severe cognitive deficits, while common single-nucleotide polymorphisms in these genes are associated with schizophrenia (SZ) and cognition in genome-wide association studies. The role of these genes in neuropsychiatric disorders is unknown. The ciliopathy gene SDCCAG8 is associated with SZ and educational attainment (EA). Genome editing of SDCCAG8 caused defects in primary ciliogenesis and cilium-dependent cell signalling. Transcriptomic analysis of SDCCAG8-deficient cells identified differentially expressed genes that are enriched in neurodevelopmental processes such as generation of neurons and synapse organization. These processes are enriched for genes associated with SZ, human intelligence (IQ) and EA. Phenotypic analysis of SDCCAG8-deficent neuronal cells revealed impaired migration and neuronal differentiation. These data implicate ciliary signalling in the aetiology of SZ and cognitive dysfunction. We found that centrosomal/ciliary genes are enriched for association with IQ, suggesting altered gene regulation as a general model for neurodevelopmental impacts of centrosomal/ciliary genes.
Collapse
Affiliation(s)
- Mairéad Flynn
- Cognitive Genetics and Cognitive Therapy Group, Neuroimaging and Cognitive Genomics (NICOG) Centre, School of Psychology and Discipline of Biochemistry, National University of Ireland Galway, Ireland.,Centre for Chromosome Biology, School of Natural Sciences, National University of Ireland Galway, Ireland
| | - Laura Whitton
- Cognitive Genetics and Cognitive Therapy Group, Neuroimaging and Cognitive Genomics (NICOG) Centre, School of Psychology and Discipline of Biochemistry, National University of Ireland Galway, Ireland
| | - Gary Donohoe
- Cognitive Genetics and Cognitive Therapy Group, Neuroimaging and Cognitive Genomics (NICOG) Centre, School of Psychology and Discipline of Biochemistry, National University of Ireland Galway, Ireland
| | - Ciaran G Morrison
- Centre for Chromosome Biology, School of Natural Sciences, National University of Ireland Galway, Ireland
| | - Derek W Morris
- Cognitive Genetics and Cognitive Therapy Group, Neuroimaging and Cognitive Genomics (NICOG) Centre, School of Psychology and Discipline of Biochemistry, National University of Ireland Galway, Ireland
| |
Collapse
|
21
|
Yoshino Y, Fang Z, Qi H, Kobayashi A, Chiba N. Dysregulation of the centrosome induced by BRCA1 deficiency contributes to tissue-specific carcinogenesis. Cancer Sci 2021; 112:1679-1687. [PMID: 33606355 PMCID: PMC8088922 DOI: 10.1111/cas.14859] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 02/11/2021] [Accepted: 02/16/2021] [Indexed: 12/12/2022] Open
Abstract
Alterations in breast cancer gene 1 (BRCA1), a tumor suppressor gene, increase the risk of breast and ovarian cancers. BRCA1 forms a heterodimer with BRCA1-associated RING domain protein 1 (BARD1) and functions in multiple cellular processes, including DNA repair and centrosome regulation. BRCA1 acts as a tumor suppressor by promoting homologous recombination (HR) repair, and alterations in BRCA1 cause HR deficiency, not only in breast and ovarian tissues but also in other tissues. The molecular mechanisms underlying BRCA1 alteration-induced carcinogenesis remain unclear. Centrosomes are the major microtubule-organizing centers and function in bipolar spindle formation. The regulation of centrosome number is critical for chromosome segregation in mitosis, which maintains genomic stability. BRCA1/BARD1 function in centrosome regulation together with Obg-like ATPase (OLA1) and receptor for activating protein C kinase 1 (RACK1). Cancer-derived variants of BRCA1, BARD1, OLA1, and RACK1 do not interact, and aberrant expression of these proteins results in abnormal centrosome duplication in mammary-derived cells, and rarely in other cell types. RACK1 is involved in centriole duplication in the S phase by promoting polo-like kinase 1 activation by Aurora A, which is critical for centrosome duplication. Centriole number is higher in cells derived from mammary tissues compared with in those derived from other tissues, suggesting that tissue-specific centrosome characterization may shed light on the tissue specificity of BRCA1-associated carcinogenesis. Here, we explored the role of the BRCA1-containing complex in centrosome regulation and the effect of its deficiency on tissue-specific carcinogenesis.
Collapse
Affiliation(s)
- Yuki Yoshino
- Department of Cancer BiologyInstitute of Aging, Development, and CancerTohoku UniversitySendaiJapan
- Laboratory of Cancer BiologyGraduate School of Life SciencesTohoku UniversitySendaiJapan
- Department of Cancer BiologyTohoku University Graduate School of MedicineSendaiJapan
| | - Zhenzhou Fang
- Department of Cancer BiologyInstitute of Aging, Development, and CancerTohoku UniversitySendaiJapan
- Department of Cancer BiologyTohoku University Graduate School of MedicineSendaiJapan
| | - Huicheng Qi
- Department of Cancer BiologyInstitute of Aging, Development, and CancerTohoku UniversitySendaiJapan
- Department of Cancer BiologyTohoku University Graduate School of MedicineSendaiJapan
| | - Akihiro Kobayashi
- Department of Cancer BiologyInstitute of Aging, Development, and CancerTohoku UniversitySendaiJapan
- Department of Cancer BiologyTohoku University Graduate School of MedicineSendaiJapan
| | - Natsuko Chiba
- Department of Cancer BiologyInstitute of Aging, Development, and CancerTohoku UniversitySendaiJapan
- Laboratory of Cancer BiologyGraduate School of Life SciencesTohoku UniversitySendaiJapan
- Department of Cancer BiologyTohoku University Graduate School of MedicineSendaiJapan
| |
Collapse
|
22
|
Primary cilia and the DNA damage response: linking a cellular antenna and nuclear signals. Biochem Soc Trans 2021; 49:829-841. [PMID: 33843966 DOI: 10.1042/bst20200751] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 03/09/2021] [Accepted: 03/10/2021] [Indexed: 11/17/2022]
Abstract
The maintenance of genome stability involves integrated biochemical activities that detect DNA damage or incomplete replication, delay the cell cycle, and direct DNA repair activities on the affected chromatin. These processes, collectively termed the DNA damage response (DDR), are crucial for cell survival and to avoid disease, particularly cancer. Recent work has highlighted links between the DDR and the primary cilium, an antenna-like, microtubule-based signalling structure that extends from a centriole docked at the cell surface. Ciliary dysfunction gives rise to a range of complex human developmental disorders termed the ciliopathies. Mutations in ciliopathy genes have been shown to impact on several functions that relate to centrosome integrity, DNA damage signalling, responses to problems in DNA replication and the control of gene expression. This review covers recent findings that link cilia and the DDR and explores the various roles played by key genes in these two contexts. It outlines how proteins encoded by ciliary genes impact checkpoint signalling, DNA replication and repair, gene expression and chromatin remodelling. It discusses how these diverse activities may integrate nuclear responses with those that affect a structure of the cell periphery. Additional directions for exploration of the interplay between these pathways are highlighted, with a focus on new ciliary gene candidates that alter genome stability.
Collapse
|
23
|
Chen YC, Hsieh HH, Chang HC, Wang HC, Lin WJ, Lin JJ. CDC25B induces cellular senescence and correlates with tumor suppression in a p53-dependent manner. J Biol Chem 2021; 296:100564. [PMID: 33745968 PMCID: PMC8054198 DOI: 10.1016/j.jbc.2021.100564] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 03/10/2021] [Accepted: 03/17/2021] [Indexed: 01/13/2023] Open
Abstract
The phosphatase cell division cycle 25B (Cdc25B) regulates cell cycle progression. Increased Cdc25B levels are often detected in cancer cell lines and human cancers and have been implicated in contributing to tumor growth, potentially by providing cancer cells with the ability to bypass checkpoint controls. However, the specific mechanism by which increased Cdc25B impacts tumor progression is not clear. Here we analyzed The Cancer Genome Atlas (TCGA) database and found that patients with high CDC25B expression had the expected poor survival. However, we also found that high CDC25B expression had a p53-dependent tumor suppressive effect in lung cancer and possibly several other cancer types. Looking in more detail at the tumor suppressive function of Cdc25B, we found that increased Cdc25B expression caused inhibition of cell growth in human normal fibroblasts. This effect was not due to alteration of specific cell cycle stage or inhibition of apoptosis, nor by induction of the DNA damage response. Instead, increased CDC25B expression led cells into senescence. We also found that p53 was required to induce senescence, which might explain the p53-dependent tumor suppressive function of Cdc25B. Mechanistically, we found that the Cdc25B phosphatase activity was required to induce senescence. Further analysis also found that Cdc25B stabilized p53 through binding and dephosphorylating p53. Together, this study identified a tumor-suppressive function of Cdc25B that is mediated through a p53-dependent senescence pathway.
Collapse
Affiliation(s)
- Ying-Chieh Chen
- Institute of Biopharmaceutical Sciences, National Yang-Ming University, Taipei, Taiwan
| | - Hsi-Hsien Hsieh
- Institute of Biopharmaceutical Sciences, National Yang-Ming University, Taipei, Taiwan
| | - Hsi-Chi Chang
- Institute of Biopharmaceutical Sciences, National Yang-Ming University, Taipei, Taiwan
| | - Hsin-Chiao Wang
- Institute of Biochemistry and Molecular Biology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Wey-Jinq Lin
- Institute of Biopharmaceutical Sciences, National Yang-Ming University, Taipei, Taiwan.
| | - Jing-Jer Lin
- Institute of Biopharmaceutical Sciences, National Yang-Ming University, Taipei, Taiwan; Institute of Biochemistry and Molecular Biology, National Taiwan University College of Medicine, Taipei, Taiwan.
| |
Collapse
|
24
|
Burigotto M, Mattivi A, Migliorati D, Magnani G, Valentini C, Roccuzzo M, Offterdinger M, Pizzato M, Schmidt A, Villunger A, Maffini S, Fava LL. Centriolar distal appendages activate the centrosome-PIDDosome-p53 signalling axis via ANKRD26. EMBO J 2021; 40:e104844. [PMID: 33350486 PMCID: PMC7883297 DOI: 10.15252/embj.2020104844] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 10/22/2020] [Accepted: 11/04/2020] [Indexed: 01/08/2023] Open
Abstract
Centrosome amplification results into genetic instability and predisposes cells to neoplastic transformation. Supernumerary centrosomes trigger p53 stabilization dependent on the PIDDosome (a multiprotein complex composed by PIDD1, RAIDD and Caspase-2), whose activation results in cleavage of p53's key inhibitor, MDM2. Here, we demonstrate that PIDD1 is recruited to mature centrosomes by the centriolar distal appendage protein ANKRD26. PIDDosome-dependent Caspase-2 activation requires not only PIDD1 centrosomal localization, but also its autoproteolysis. Following cytokinesis failure, supernumerary centrosomes form clusters, which appear to be necessary for PIDDosome activation. In addition, in the context of DNA damage, activation of the complex results from a p53-dependent elevation of PIDD1 levels independently of centrosome amplification. We propose that PIDDosome activation can in both cases be promoted by an ANKRD26-dependent local increase in PIDD1 concentration close to the centrosome. Collectively, these findings provide a paradigm for how centrosomes can contribute to cell fate determination by igniting a signalling cascade.
Collapse
Affiliation(s)
- Matteo Burigotto
- Armenise‐Harvard Laboratory of Cell DivisionDepartment of Cellular, Computational and Integrative Biology—CIBIOUniversity of TrentoTrentoItaly
| | - Alessia Mattivi
- Armenise‐Harvard Laboratory of Cell DivisionDepartment of Cellular, Computational and Integrative Biology—CIBIOUniversity of TrentoTrentoItaly
| | - Daniele Migliorati
- Armenise‐Harvard Laboratory of Cell DivisionDepartment of Cellular, Computational and Integrative Biology—CIBIOUniversity of TrentoTrentoItaly
| | - Giovanni Magnani
- Armenise‐Harvard Laboratory of Cell DivisionDepartment of Cellular, Computational and Integrative Biology—CIBIOUniversity of TrentoTrentoItaly
| | - Chiara Valentini
- Armenise‐Harvard Laboratory of Cell DivisionDepartment of Cellular, Computational and Integrative Biology—CIBIOUniversity of TrentoTrentoItaly
| | - Michela Roccuzzo
- Advanced Imaging Core FacilityDepartment of Cellular, Computational and Integrative Biology—CIBIOUniversity of TrentoTrentoItaly
| | - Martin Offterdinger
- Division of NeurobiochemistryBioopticsBiocenterMedical University of InnsbruckInnsbruckAustria
| | - Massimo Pizzato
- Laboratory of Virus‐Cell InteractionDepartment of Cellular, Computational and Integrative Biology—CIBIOUniversity of TrentoTrentoItaly
| | - Alexander Schmidt
- Proteomics Core FacilityBiozentrumUniversity of BaselBaselSwitzerland
| | - Andreas Villunger
- Division of Developmental ImmunologyBiocenterMedical University of InnsbruckInnsbruckAustria
- CeMM Research Center for Molecular Medicine of the Austrian Academy of SciencesViennaAustria
| | - Stefano Maffini
- Department of Mechanistic Cell BiologyMax Planck Institute of Molecular PhysiologyDortmundGermany
| | - Luca L Fava
- Armenise‐Harvard Laboratory of Cell DivisionDepartment of Cellular, Computational and Integrative Biology—CIBIOUniversity of TrentoTrentoItaly
| |
Collapse
|
25
|
Drosophila female germline stem cells undergo mitosis without nuclear breakdown. Curr Biol 2021; 31:1450-1462.e3. [PMID: 33548191 DOI: 10.1016/j.cub.2021.01.033] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 11/18/2020] [Accepted: 01/11/2021] [Indexed: 02/02/2023]
Abstract
Stem cell homeostasis requires nuclear lamina (NL) integrity. In Drosophila germ cells, compromised NL integrity activates the ataxia telangiectasia and Rad3-related (ATR) and checkpoint kinase 2 (Chk2) checkpoint kinases, blocking germ cell differentiation and causing germline stem cell (GSC) loss. Checkpoint activation occurs upon loss of either the NL protein emerin or its partner barrier-to-autointegration factor, two proteins required for nuclear reassembly at the end of mitosis. Here, we examined how mitosis contributes to NL structural defects linked to checkpoint activation. These analyses led to the unexpected discovery that wild-type female GSCs utilize a non-canonical mode of mitosis, one that retains a permeable but intact nuclear envelope and NL. We show that the interphase NL is remodeled during mitosis for insertion of centrosomes that nucleate the mitotic spindle within the confines of the nucleus. We show that depletion or loss of NL components causes mitotic defects, including compromised chromosome segregation associated with altered centrosome positioning and structure. Further, in emerin mutant GSCs, centrosomes remain embedded in the interphase NL. Notably, these embedded centrosomes carry large amounts of pericentriolar material and nucleate astral microtubules, revealing a role for emerin in the regulation of centrosome structure. Epistasis studies demonstrate that defects in centrosome structure are upstream of checkpoint activation, suggesting that these centrosome defects might trigger checkpoint activation and GSC loss. Connections between NL proteins and centrosome function have implications for mechanisms associated with NL dysfunction in other stem cell populations, including NL-associated diseases, such as laminopathies.
Collapse
|
26
|
Genotoxic stress-activated DNA-PK-p53 cascade and autophagy cooperatively induce ciliogenesis to maintain the DNA damage response. Cell Death Differ 2021; 28:1865-1879. [PMID: 33462409 PMCID: PMC8184926 DOI: 10.1038/s41418-020-00713-8] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 12/03/2020] [Accepted: 12/16/2020] [Indexed: 11/16/2022] Open
Abstract
The DNA-PK maintains cell survival when DNA damage occurs. In addition, aberrant activation of the DNA-PK induces centrosome amplification, suggesting additional roles for this kinase. Here, we showed that the DNA-PK-p53 cascade induced primary cilia formation (ciliogenesis), thus maintaining the DNA damage response under genotoxic stress. Treatment with genotoxic drugs (etoposide, neocarzinostatin, hydroxyurea, or cisplatin) led to ciliogenesis in human retina (RPE1), trophoblast (HTR8), lung (A459), and mouse Leydig progenitor (TM3) cell lines. Upon genotoxic stress, several DNA damage signaling were activated, but only the DNA-PK-p53 cascade contributed to ciliogenesis, as pharmacological inhibition or genetic depletion of this pathway decreased genotoxic stress-induced ciliogenesis. Interestingly, in addition to localizing to the nucleus, activated DNA-PK localized to the base of the primary cilium (mother centriole) and daughter centriole. Genotoxic stress also induced autophagy. Inhibition of autophagy initiation or lysosomal degradation or depletion of ATG7 decreased genotoxic stress-induced ciliogenesis. Besides, inhibition of ciliogenesis by depletion of IFT88 or CEP164 attenuated the genotoxic stress-induced DNA damage response. Thus, our study uncovered the interplay among genotoxic stress, the primary cilium, and the DNA damage response.
Collapse
|
27
|
Ma S, Rong Z, Liu C, Qin X, Zhang X, Chen Q. DNA damage promotes microtubule dynamics through a DNA-PK-AKT axis for enhanced repair. J Cell Biol 2021; 220:211656. [PMID: 33404607 PMCID: PMC7791344 DOI: 10.1083/jcb.201911025] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 11/01/2020] [Accepted: 12/02/2020] [Indexed: 01/09/2023] Open
Abstract
DNA double-strand breaks (DSBs) are mainly repaired by c-NHEJ and HR pathways. The enhanced DSB mobility after DNA damage is critical for efficient DSB repair. Although microtubule dynamics have been shown to regulate DSB mobility, the reverse effect of DSBs to microtubule dynamics remains elusive. Here, we uncovered a novel DSB-induced microtubule dynamics stress response (DMSR), which promotes DSB mobility and facilitates c-NHEJ repair. DMSR is accompanied by interphase centrosome maturation, which occurs in a DNA-PK-AKT-dependent manner. Depletion of PCM proteins attenuates DMSR and the mobility of DSBs, resulting in delayed c-NHEJ. Remarkably, DMSR occurs only in G1 or G0 cells and lasts around 6 h. Both inhibition of DNA-PK and depletion of 53BP1 abolish DMSR. Taken together, our study reveals a positive DNA repair mechanism in G1 or G0 cells in which DSBs actively promote microtubule dynamics and facilitate the c-NHEJ process.
Collapse
Affiliation(s)
- Shuyun Ma
- Department of Radiation and Medical Oncology, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China,Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, China
| | - Zeming Rong
- Department of Radiation and Medical Oncology, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China,Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, China
| | - Chen Liu
- Department of Radiation and Medical Oncology, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China,Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, China
| | - Xiaobing Qin
- Department of Radiation and Medical Oncology, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China,Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, China
| | - Xiaoyan Zhang
- College of Biomedicine and Health, Huazhong Agricultural University, Wuhan, China,College of Life Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Qiang Chen
- Department of Radiation and Medical Oncology, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China,Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, China,Correspondence to Qiang Chen:
| |
Collapse
|
28
|
Otsuka K, Yoshino Y, Qi H, Chiba N. The Function of BARD1 in Centrosome Regulation in Cooperation with BRCA1/OLA1/RACK1. Genes (Basel) 2020; 11:genes11080842. [PMID: 32722046 PMCID: PMC7464954 DOI: 10.3390/genes11080842] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 07/21/2020] [Accepted: 07/22/2020] [Indexed: 12/17/2022] Open
Abstract
Breast cancer gene 1 (BRCA1)-associated RING domain protein 1 (BARD1) forms a heterodimer with BRCA1, a tumor suppressor associated with hereditary breast and ovarian cancer. BRCA1/BARD1 functions in multiple cellular processes including DNA repair and centrosome regulation. Centrosomes are the major microtubule-organizing centers in animal cells and are critical for the formation of a bipolar mitotic spindle. BRCA1 and BARD1 localize to the centrosome during the cell cycle, and the BRCA1/BARD1 dimer ubiquitinates centrosomal proteins to regulate centrosome function. We identified Obg-like ATPase 1 (OLA1) and receptor for activated C kinase (RACK1) as BRCA1/BARD1-interating proteins that bind to BARD1 and BRCA1 and localize the centrosomes during the cell cycle. Cancer-derived variants of BRCA1, BARD1, OLA1, and RACK1 failed to interact, and aberrant expression of these proteins caused centrosome amplification due to centriole overduplication only in mammary tissue-derived cells. In S-G2 phase, the number of centrioles was higher in mammary tissue-derived cells than in cells from other tissues, suggesting their involvement in tissue-specific carcinogenesis by BRCA1 and BARD1 germline mutations. We described the function of BARD1 in centrosome regulation in cooperation with BRCA1/OLA1/RACK1, as well as the effect of their dysfunction on carcinogenesis.
Collapse
Affiliation(s)
- Kei Otsuka
- Department of Cancer Biology, Institute of Development, Aging and Cancer (IDAC), Tohoku University, 4-1 Seiryomachi Aoba-ku, Sendai 980-8575, Japan; (K.O.); (Y.Y.); (H.Q.)
- Laboratory of Cancer Biology, Graduate School of Life Sciences, Tohoku University, 4-1 Seiryomachi Aoba-ku, Sendai 980-8575, Japan
| | - Yuki Yoshino
- Department of Cancer Biology, Institute of Development, Aging and Cancer (IDAC), Tohoku University, 4-1 Seiryomachi Aoba-ku, Sendai 980-8575, Japan; (K.O.); (Y.Y.); (H.Q.)
- Laboratory of Cancer Biology, Graduate School of Life Sciences, Tohoku University, 4-1 Seiryomachi Aoba-ku, Sendai 980-8575, Japan
- Department of Cancer Biology, Tohoku University Graduate School of Medicine, 4-1 Seiryomachi Aoba-ku, Sendai 980-8575, Japan
| | - Huicheng Qi
- Department of Cancer Biology, Institute of Development, Aging and Cancer (IDAC), Tohoku University, 4-1 Seiryomachi Aoba-ku, Sendai 980-8575, Japan; (K.O.); (Y.Y.); (H.Q.)
- Department of Cancer Biology, Tohoku University Graduate School of Medicine, 4-1 Seiryomachi Aoba-ku, Sendai 980-8575, Japan
| | - Natsuko Chiba
- Department of Cancer Biology, Institute of Development, Aging and Cancer (IDAC), Tohoku University, 4-1 Seiryomachi Aoba-ku, Sendai 980-8575, Japan; (K.O.); (Y.Y.); (H.Q.)
- Laboratory of Cancer Biology, Graduate School of Life Sciences, Tohoku University, 4-1 Seiryomachi Aoba-ku, Sendai 980-8575, Japan
- Department of Cancer Biology, Tohoku University Graduate School of Medicine, 4-1 Seiryomachi Aoba-ku, Sendai 980-8575, Japan
- Correspondence:
| |
Collapse
|
29
|
Moonlighting in Mitosis: Analysis of the Mitotic Functions of Transcription and Splicing Factors. Cells 2020; 9:cells9061554. [PMID: 32604778 PMCID: PMC7348712 DOI: 10.3390/cells9061554] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Revised: 06/22/2020] [Accepted: 06/23/2020] [Indexed: 12/14/2022] Open
Abstract
Moonlighting proteins can perform one or more additional functions besides their primary role. It has been posited that a protein can acquire a moonlighting function through a gradual evolutionary process, which is favored when the primary and secondary functions are exerted in different cellular compartments. Transcription factors (TFs) and splicing factors (SFs) control processes that occur in interphase nuclei and are strongly reduced during cell division, and are therefore in a favorable situation to evolve moonlighting mitotic functions. However, recently published moonlighting protein databases, which comprise almost 400 proteins, do not include TFs and SFs with secondary mitotic functions. We searched the literature and found several TFs and SFs with bona fide moonlighting mitotic functions, namely they localize to specific mitotic structure(s), interact with proteins enriched in the same structure(s), and are required for proper morphology and functioning of the structure(s). In addition, we describe TFs and SFs that localize to mitotic structures but cannot be classified as moonlighting proteins due to insufficient data on their biochemical interactions and mitotic roles. Nevertheless, we hypothesize that most TFs and SFs with specific mitotic localizations have either minor or redundant moonlighting functions, or are evolving towards the acquisition of these functions.
Collapse
|
30
|
Sterpka A, Yang J, Strobel M, Zhou Y, Pauplis C, Chen X. Diverged morphology changes of astrocytic and neuronal primary cilia under reactive insults. Mol Brain 2020; 13:28. [PMID: 32122360 PMCID: PMC7053156 DOI: 10.1186/s13041-020-00571-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Accepted: 02/24/2020] [Indexed: 12/13/2022] Open
Abstract
Primary cilia are centriole-derived sensory organelles that are present in most mammalian cells, including astrocytes and neurons. Evidence is emerging that astrocyte and neuronal primary cilia demonstrate a dichotomy in the mature mouse brain. However, it is unknown how astrocytic and neuronal primary cilia change their morphology and ciliary proteins when exposed to reactive insults including epilepsy and traumatic brain injury. We used a double transgenic mouse strain (Arl13b-mCherry; Centrin2-GFP), in which we found spontaneous seizures, and a cortical injury model to examine the morphological changes of astrocytic and neuronal primary cilia under reactive conditions. Transgenic overexpression of Arl13b drastically increases the length of astrocytic and neuronal primary cilia in the hippocampus, as well as the cilia lengths of cultured astrocytes and neurons. Spontaneous seizures shorten Arl13b-positive astrocytic cilia and AC3-positive neuronal cilia in the hippocampus. In a cortical injury model, Arl13b is not detectable in primary cilia, but Arl13b protein relocates to the cell body and has robust expression in the proximity of injured tissues. In contrast, the number of AC3-positive cilia near injured tissues remains unchanged, but their lengths become shorter. These results on astrocytic cilia implicate Arl13b in regulating astrocyte proliferation and tissue regeneration, while the shortening of AC3-positive cilia suggests adaptive changes of neuronal primary cilia under excitotoxicity.
Collapse
Affiliation(s)
- Ashley Sterpka
- Department of Molecular, Cellular and Biomedical Sciences, College of Life Sciences and Agriculture, University of New Hampshire, 389 Rudman Hall, 46 College Road, Durham, NH, 03824, USA
| | - Juan Yang
- Department of Molecular, Cellular and Biomedical Sciences, College of Life Sciences and Agriculture, University of New Hampshire, 389 Rudman Hall, 46 College Road, Durham, NH, 03824, USA
| | - Matthew Strobel
- Department of Molecular, Cellular and Biomedical Sciences, College of Life Sciences and Agriculture, University of New Hampshire, 389 Rudman Hall, 46 College Road, Durham, NH, 03824, USA
| | - Yuxin Zhou
- Department of Molecular, Cellular and Biomedical Sciences, College of Life Sciences and Agriculture, University of New Hampshire, 389 Rudman Hall, 46 College Road, Durham, NH, 03824, USA
| | - Connor Pauplis
- Department of Molecular, Cellular and Biomedical Sciences, College of Life Sciences and Agriculture, University of New Hampshire, 389 Rudman Hall, 46 College Road, Durham, NH, 03824, USA
| | - Xuanmao Chen
- Department of Molecular, Cellular and Biomedical Sciences, College of Life Sciences and Agriculture, University of New Hampshire, 389 Rudman Hall, 46 College Road, Durham, NH, 03824, USA.
| |
Collapse
|
31
|
Remo A, Li X, Schiebel E, Pancione M. The Centrosome Linker and Its Role in Cancer and Genetic Disorders. Trends Mol Med 2020; 26:380-393. [PMID: 32277932 DOI: 10.1016/j.molmed.2020.01.011] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 11/26/2019] [Accepted: 01/21/2020] [Indexed: 02/07/2023]
Abstract
Centrosome cohesion, the joining of the two centrosomes of a cell, is increasingly appreciated as a major regulator of cell functions such as Golgi organization and cilia positioning. One major element of centrosome cohesion is the centrosome linker that consists of a growing number of proteins. The timely disassembly of the centrosome linker enables centrosomes to separate and assemble a functional bipolar mitotic spindle that is crucial for maintaining genomic integrity. Exciting new findings link centrosome linker defects to cell transformation and genetic disorders. We review recent data on the molecular mechanisms of the assembly and disassembly of the centrosome linker, and discuss how defects in the proper execution of these processes cause DNA damage and genomic instability leading to disease.
Collapse
Affiliation(s)
- Andrea Remo
- Pathology Unit, Mater Salutis Hospital, Azienda Unità Locale Socio Sanitaria (AULSS) 9 'Scaligera', Verona, Italy
| | - Xue Li
- Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH), Deutsches Krebsforschungszentrum (DKFZ)-ZMBH Allianz, Heidelberg, Germany; Heidelberg Biosciences International Graduate School (HBIGS), Universität Heidelberg, Heidelberg, Germany
| | - Elmar Schiebel
- Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH), Deutsches Krebsforschungszentrum (DKFZ)-ZMBH Allianz, Heidelberg, Germany.
| | - Massimo Pancione
- Department of Sciences and Technologies, University of Sannio, Benevento, Italy; Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Complutense University of Madrid, Madrid, Spain.
| |
Collapse
|
32
|
Ali H, Braga L, Giacca M. Cardiac regeneration and remodelling of the cardiomyocyte cytoarchitecture. FEBS J 2020; 287:417-438. [PMID: 31743572 DOI: 10.1111/febs.15146] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 09/27/2019] [Accepted: 11/18/2019] [Indexed: 12/13/2022]
Abstract
Adult mammals are unable to regenerate their hearts after cardiac injury, largely due to the incapacity of cardiomyocytes (CMs) to undergo cell division. However, mammalian embryonic and fetal CMs, similar to CMs from fish and amphibians during their entire life, exhibit robust replicative activity, which stops abruptly after birth and never significantly resumes. Converging evidence indicates that formation of the highly ordered and stable cytoarchitecture of mammalian mature CMs is coupled with loss of their proliferative potential. Here, we review the available information on the role of the cardiac cytoskeleton and sarcomere in the regulation of CM proliferation. The actin cytoskeleton, the intercalated disc, the microtubular network and the dystrophin-glycoprotein complex each sense mechanical cues from the surrounding environment. Furthermore, they participate in the regulation of CM proliferation by impinging on the yes-associated protein/transcriptional co-activator with PDZ-binding motif, β-catenin and myocardin-related transcription factor transcriptional co-activators. Mastering the molecular mechanisms regulating CM proliferation would permit the development of innovative strategies to stimulate cardiac regeneration in adult individuals, a hitherto unachieved yet fundamental therapeutic goal.
Collapse
Affiliation(s)
- Hashim Ali
- British Heart Foundation Centre of Research Excellence, School of Cardiovascular Medicine & Sciences, King's College London, UK.,Molecular Medicine Laboratory, International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy
| | - Luca Braga
- British Heart Foundation Centre of Research Excellence, School of Cardiovascular Medicine & Sciences, King's College London, UK.,Molecular Medicine Laboratory, International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy
| | - Mauro Giacca
- British Heart Foundation Centre of Research Excellence, School of Cardiovascular Medicine & Sciences, King's College London, UK.,Molecular Medicine Laboratory, International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy.,Department of Medical, Surgical and Health Sciences, University of Trieste, Italy
| |
Collapse
|
33
|
Casar Tena T, Maerz LD, Szafranski K, Groth M, Blätte TJ, Donow C, Matysik S, Walther P, Jeggo PA, Burkhalter MD, Philipp M. Resting cells rely on the DNA helicase component MCM2 to build cilia. Nucleic Acids Res 2019; 47:134-151. [PMID: 30329080 PMCID: PMC6326816 DOI: 10.1093/nar/gky945] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2018] [Accepted: 10/04/2018] [Indexed: 12/24/2022] Open
Abstract
Minichromosome maintenance (MCM) proteins facilitate replication by licensing origins and unwinding the DNA double strand. Interestingly, the number of MCM hexamers greatly exceeds the number of firing origins suggesting additional roles of MCMs. Here we show a hitherto unanticipated function of MCM2 in cilia formation in human cells and zebrafish that is uncoupled from replication. Zebrafish depleted of MCM2 develop ciliopathy-phenotypes including microcephaly and aberrant heart looping due to malformed cilia. In non-cycling human fibroblasts, loss of MCM2 promotes transcription of a subset of genes, which cause cilia shortening and centriole overduplication. Chromatin immunoprecipitation experiments show that MCM2 binds to transcription start sites of cilia inhibiting genes. We propose that such binding may block RNA polymerase II-mediated transcription. Depletion of a second MCM (MCM7), which functions in complex with MCM2 during its canonical functions, reveals an overlapping cilia-deficiency phenotype likely unconnected to replication, although MCM7 appears to regulate a distinct subset of genes and pathways. Our data suggests that MCM2 and 7 exert a role in ciliogenesis in post-mitotic tissues.
Collapse
Affiliation(s)
- Teresa Casar Tena
- Institute of Biochemistry and Molecular Biology, Ulm University, 89081 Ulm, Germany
| | - Lars D Maerz
- Institute of Biochemistry and Molecular Biology, Ulm University, 89081 Ulm, Germany
| | - Karol Szafranski
- Leibniz Institute on Aging, Fritz Lipmann Institute, 07745 Jena, Germany
| | - Marco Groth
- Leibniz Institute on Aging, Fritz Lipmann Institute, 07745 Jena, Germany
| | - Tamara J Blätte
- Institute of Biochemistry and Molecular Biology, Ulm University, 89081 Ulm, Germany
| | - Cornelia Donow
- Institute of Biochemistry and Molecular Biology, Ulm University, 89081 Ulm, Germany
| | - Sabrina Matysik
- Institute of Biochemistry and Molecular Biology, Ulm University, 89081 Ulm, Germany
| | - Paul Walther
- Central Facility for Electron Microscopy, Ulm University, 89081 Ulm, Germany
| | - Penelope A Jeggo
- Genome Damage and Stability Centre, University of Sussex, Brighton BN1 9RQ, UK
| | - Martin D Burkhalter
- Institute of Biochemistry and Molecular Biology, Ulm University, 89081 Ulm, Germany
| | - Melanie Philipp
- Institute of Biochemistry and Molecular Biology, Ulm University, 89081 Ulm, Germany
| |
Collapse
|
34
|
The Nuclear Arsenal of Cilia. Dev Cell 2019; 49:161-170. [DOI: 10.1016/j.devcel.2019.03.009] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 12/07/2018] [Accepted: 03/08/2019] [Indexed: 12/31/2022]
|
35
|
Chumová J, Kourová H, Trögelová L, Halada P, Binarová P. Microtubular and Nuclear Functions of γ-Tubulin: Are They LINCed? Cells 2019; 8:cells8030259. [PMID: 30893853 PMCID: PMC6468392 DOI: 10.3390/cells8030259] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Revised: 03/07/2019] [Accepted: 03/14/2019] [Indexed: 01/02/2023] Open
Abstract
γ-Tubulin is a conserved member of the tubulin superfamily with a function in microtubule nucleation. Proteins of γ-tubulin complexes serve as nucleation templates as well as a majority of other proteins contributing to centrosomal and non-centrosomal nucleation, conserved across eukaryotes. There is a growing amount of evidence of γ-tubulin functions besides microtubule nucleation in transcription, DNA damage response, chromatin remodeling, and on its interactions with tumor suppressors. However, the molecular mechanisms are not well understood. Furthermore, interactions with lamin and SUN proteins of the LINC complex suggest the role of γ-tubulin in the coupling of nuclear organization with cytoskeletons. γ-Tubulin that belongs to the clade of eukaryotic tubulins shows characteristics of both prokaryotic and eukaryotic tubulins. Both human and plant γ-tubulins preserve the ability of prokaryotic tubulins to assemble filaments and higher-order fibrillar networks. γ-Tubulin filaments, with bundling and aggregating capacity, are suggested to perform complex scaffolding and sequestration functions. In this review, we discuss a plethora of γ-tubulin molecular interactions and cellular functions, as well as recent advances in understanding the molecular mechanisms behind them.
Collapse
Affiliation(s)
- Jana Chumová
- Institute of Microbiology of the Czech Academy of Sciences, Vídeňská 1083, 142 20 Prague, Czech Republic.
| | - Hana Kourová
- Institute of Microbiology of the Czech Academy of Sciences, Vídeňská 1083, 142 20 Prague, Czech Republic.
| | - Lucie Trögelová
- Institute of Microbiology of the Czech Academy of Sciences, Vídeňská 1083, 142 20 Prague, Czech Republic.
| | - Petr Halada
- Institute of Microbiology of the Czech Academy of Sciences, Vídeňská 1083, 142 20 Prague, Czech Republic.
| | - Pavla Binarová
- Institute of Microbiology of the Czech Academy of Sciences, Vídeňská 1083, 142 20 Prague, Czech Republic.
| |
Collapse
|
36
|
Wang H, Huang Y, Shi J, Zhi Y, Yuan F, Yu J, Chen Z, Yang J. XPC deficiency leads to centrosome amplification by inhibiting BRCA1 expression upon cisplatin-mediated DNA damage in human bladder cancer. Cancer Lett 2018; 444:136-146. [PMID: 30579971 DOI: 10.1016/j.canlet.2018.12.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 12/03/2018] [Accepted: 12/11/2018] [Indexed: 12/15/2022]
Abstract
Xeroderma pigmentosum group C (XPC) is a well-known DNA damage recognition protein. Defects in XPC lead to carcinogenesis and progression of many human cancers. In the current study, we defined a novel, important role of XPC in preventing centrosome amplification during cisplatin-mediated DNA damage response. From experiments with human bladder cancer tissue, urothelial tissue from Xpc knockout mice and XPC-silenced cell lines, we found that attenuated XPC expression was associated with increased centrosome amplification in human bladder cancer. A significant increase in centrosome amplification was observed in XPC-silenced cells upon cisplatin treatment. XPC deficiency leads to reduced BRCA1 expression via upregulating its transcriptional repressor, Pit-1. The BRCA1 downregulation results in more DNA double strand breaks accumulation and persistent activation of the ATM-Chk1/Chk2 signaling, resulting in a prolonged G2/M arrest during which centrosome can over-duplicate and lead to centrosome amplification. XPC complementation in silenced cells could reduce Pit-1 expression, increase BRCA1 expression and recover the status of centrosome amplification. Our study reveals a new function for XPC in preventing chromosomal instability, providing new information on cancer chemotherapy and potential clinical significance for cancer management.
Collapse
Affiliation(s)
- Huanhuan Wang
- Department of Cell Biology, The Third Military Medical University, Chongqing, PR China
| | - Yaqin Huang
- Department of Cell Biology, The Third Military Medical University, Chongqing, PR China
| | - Jiazhong Shi
- Department of Cell Biology, The Third Military Medical University, Chongqing, PR China
| | - Yi Zhi
- Department of Urology, Third Affiliated Hospital of Chongqing Medical University, Chongqing, PR China
| | - Fang Yuan
- Chongqing University Cancer Hospital, Chongqing, PR China
| | - Jin Yu
- Department of Cell Biology, The Third Military Medical University, Chongqing, PR China
| | - Zhiwen Chen
- Urology Institute of People's Liberation Army, Southwest Hospital, The Third Military Medical University, Chongqing, PR China; Southwest Cancer Center, Southwest Hospital, The Third Military Medical University, Chongqing, China.
| | - Jin Yang
- Department of Cell Biology, The Third Military Medical University, Chongqing, PR China.
| |
Collapse
|
37
|
Hearn T. ALMS1 and Alström syndrome: a recessive form of metabolic, neurosensory and cardiac deficits. J Mol Med (Berl) 2018; 97:1-17. [PMID: 30421101 PMCID: PMC6327082 DOI: 10.1007/s00109-018-1714-x] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Revised: 10/25/2018] [Accepted: 10/30/2018] [Indexed: 12/12/2022]
Abstract
Alström syndrome (AS) is characterised by metabolic deficits, retinal dystrophy, sensorineural hearing loss, dilated cardiomyopathy and multi-organ fibrosis. Elucidating the function of the mutated gene, ALMS1, is critical for the development of specific treatments and may uncover pathways relevant to a range of other disorders including common forms of obesity and type 2 diabetes. Interest in ALMS1 is heightened by the recent discovery of its involvement in neonatal cardiomyocyte cell cycle arrest, a process with potential relevance to regenerative medicine. ALMS1 encodes a ~ 0.5 megadalton protein that localises to the base of centrioles. Some studies have suggested a role for this protein in maintaining centriole-nucleated sensory organelles termed primary cilia, and AS is now considered to belong to the growing class of human genetic disorders linked to ciliary dysfunction (ciliopathies). However, mechanistic details are lacking, and recent studies have implicated ALMS1 in several processes including endosomal trafficking, actin organisation, maintenance of centrosome cohesion and transcription. In line with a more complex picture, multiple isoforms of the protein likely exist and non-centrosomal sites of localisation have been reported. This review outlines the evidence for both ciliary and extra-ciliary functions of ALMS1.
Collapse
Affiliation(s)
- Tom Hearn
- Institute of Life Science, Swansea University Medical School, Singleton Park, Swansea, SA2 8PP, UK.
| |
Collapse
|
38
|
Abstract
Mitosis is controlled by reversible protein phosphorylation involving specific kinases and phosphatases. A handful of major mitotic protein kinases, such as the cyclin B-CDK1 complex, the Aurora kinases, and Polo-like kinase 1 (PLK1), cooperatively regulate distinct mitotic processes. Research has identified proteins and mechanisms that integrate these kinases into signaling cascades that guide essential mitotic events. These findings have important implications for our understanding of the mechanisms of mitotic regulation and may advance the development of novel antimitotic drugs. We review collected evidence that in vertebrates, the Aurora kinases serve as catalytic subunits of distinct complexes formed with the four scaffold proteins Bora, CEP192, INCENP, and TPX2, which we deem "core" Aurora cofactors. These complexes and the Aurora-PLK1 cascades organized by Bora, CEP192, and INCENP control crucial aspects of mitosis and all pathways of spindle assembly. We compare the mechanisms of Aurora activation in relation to the different spindle assembly pathways and draw a functional analogy between the CEP192 complex and the chromosomal passenger complex that may reflect the coevolution of centrosomes, kinetochores, and the actomyosin cleavage apparatus. We also analyze the roles and mechanisms of Aurora-PLK1 signaling in the cell and centrosome cycles and in the DNA damage response.
Collapse
Affiliation(s)
- Vladimir Joukov
- N.N. Petrov National Medical Research Center of Oncology, Saint-Petersburg 197758, Russian Federation.
| | | |
Collapse
|
39
|
Hossain D, Ferreira Barbosa JA, Cohen ÉA, Tsang WY. HIV-1 Vpr hijacks EDD-DYRK2-DDB1 DCAF1 to disrupt centrosome homeostasis. J Biol Chem 2018; 293:9448-9460. [PMID: 29724823 PMCID: PMC6005440 DOI: 10.1074/jbc.ra117.001444] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Revised: 04/17/2018] [Indexed: 11/06/2022] Open
Abstract
Viruses exploit the host cell machinery for their own profit. To evade innate immune sensing and promote viral replication, HIV type 1 (HIV-1) subverts DNA repair regulatory proteins and induces G2/M arrest. The preintegration complex of HIV-1 is known to traffic along microtubules and accumulate near the microtubule-organizing center. The centrosome is the major microtubule-organizing center in most eukaryotic cells, but precisely how HIV-1 impinges on centrosome biology remains poorly understood. We report here that the HIV-1 accessory protein viral protein R (Vpr) localized to the centrosome through binding to DCAF1, forming a complex with the ubiquitin ligase EDD-DYRK2-DDB1DCAF1 and Cep78, a resident centrosomal protein previously shown to inhibit EDD-DYRK2-DDB1DCAF1 Vpr did not affect ubiquitination of Cep78. Rather, it enhanced ubiquitination of an EDD-DYRK2-DDB1DCAF1 substrate, CP110, leading to its degradation, an effect that could be overcome by Cep78 expression. The down-regulation of CP110 and elongation of centrioles provoked by Vpr were independent of G2/M arrest. Infection of T lymphocytes with HIV-1, but not with HIV-1 lacking Vpr, promoted CP110 degradation and centriole elongation. Elongated centrioles recruited more γ-tubulin to the centrosome, resulting in increased microtubule nucleation. Our results suggest that Vpr is targeted to the centrosome where it hijacks a ubiquitin ligase, disrupting organelle homeostasis, which may contribute to HIV-1 pathogenesis.
Collapse
Affiliation(s)
- Delowar Hossain
- From the Institut de recherches cliniques de Montréal, Montreal, Quebec H2W 1R7, Canada
- the Division of Experimental Medicine, McGill University, Montreal, Quebec H4A 3J1, Canada
| | | | - Éric A Cohen
- From the Institut de recherches cliniques de Montréal, Montreal, Quebec H2W 1R7, Canada
- the Division of Experimental Medicine, McGill University, Montreal, Quebec H4A 3J1, Canada
- the Department of Microbiology, Infectiology, and Immunology, Université de Montréal, Montreal, Quebec H3C 3J7, Canada, and
| | - William Y Tsang
- From the Institut de recherches cliniques de Montréal, Montreal, Quebec H2W 1R7, Canada,
- the Division of Experimental Medicine, McGill University, Montreal, Quebec H4A 3J1, Canada
- the Department of Pathology and Cell Biology, Université de Montréal, Montreal, Quebec H3C 3J7, Canada
| |
Collapse
|
40
|
Etoposide-induced DNA damage affects multiple cellular pathways in addition to DNA damage response. Oncotarget 2018; 9:24122-24139. [PMID: 29844877 PMCID: PMC5963631 DOI: 10.18632/oncotarget.24517] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2017] [Accepted: 02/10/2018] [Indexed: 02/06/2023] Open
Abstract
DNA damage response (DDR) coordinates lesion repair and checkpoint activation. DDR is intimately connected with transcription. However, the relationship between DDR and transcription has not been clearly established. We report here RNA-sequencing analyses of MCF7 cells containing double-strand breaks induced by etoposide. While etoposide does not apparently cause global changes in mRNA abundance, it altered some gene expression. At the setting of fold alteration ≥ 2 and false discovery rate (FDR) ≤ 0.001, FDR < 0.05, or p < 0.05, etoposide upregulated 96, 268, or 860 genes and downregulated 41, 133, or 503 genes in MCF7 cells. Among these differentially expressed genes (DEGs), the processes of biogenesis, metabolism, cell motility, signal transduction, and others were affected; the pathways of Ras GTPase activity, RNA binding, cytokine-mediated signaling, kinase regulatory activity, protein binding, and translation were upregulated, and those pathways related to coated vesicle, calmodulin binding, and microtubule-based movement were downregulated. We further identified RABL6, RFTN2, FAS-AS1, and TCEB3CL as new DDR-affected genes in MCF7 and T47D cells. By metabolic labelling using 4-thiouridine, we observed dynamic alterations in the transcription of these genes in etoposide-treated MCF7 and T47D cells. During 0-2 hour etoposide treatment, RABL6 transcription was robustly increased at 0.5 and 1 hour in MCF7 cells and at 2 hours in T47D cells, while FAS-AS1 transcription was dramatically and steadily elevated in both cell lines. Taken together, we demonstrate dynamic alterations in transcription and that these changes affect multiple cellular processes in etoposide-induced DDR.
Collapse
|
41
|
Patwardhan D, Mani S, Passemard S, Gressens P, El Ghouzzi V. STIL balancing primary microcephaly and cancer. Cell Death Dis 2018; 9:65. [PMID: 29352115 PMCID: PMC5833631 DOI: 10.1038/s41419-017-0101-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Revised: 10/04/2017] [Accepted: 10/23/2017] [Indexed: 11/25/2022]
Abstract
Cell division and differentiation are two fundamental physiological processes that need to be tightly balanced to achieve harmonious development of an organ or a tissue without jeopardizing its homeostasis. The role played by the centriolar protein STIL is highly illustrative of this balance at different stages of life as deregulation of the human STIL gene expression has been associated with either insufficient brain development (primary microcephaly) or cancer, two conditions resulting from perturbations in cell cycle and chromosomal segregation. This review describes the recent advances on STIL functions in the control of centriole duplication and mitotic spindle integrity, and discusses how pathological perturbations of its finely tuned expression result in chromosomal instability in both embryonic and postnatal situations, highlighting the concept that common key factors are involved in developmental steps and tissue homeostasis.
Collapse
Affiliation(s)
- Dhruti Patwardhan
- PROTECT, INSERM, Université Paris Diderot, Sorbonne Paris Cité, Paris, France
- Centre for Neuroscience, IISC Bangalore, India
| | - Shyamala Mani
- PROTECT, INSERM, Université Paris Diderot, Sorbonne Paris Cité, Paris, France
- Curadev Pharma, B 87, Sector 83, Noida, UP, 201305,, India
| | - Sandrine Passemard
- PROTECT, INSERM, Université Paris Diderot, Sorbonne Paris Cité, Paris, France
- AP HP, Hôpital Robert Debré, Service de Génétique Clinique, Paris, France
| | - Pierre Gressens
- PROTECT, INSERM, Université Paris Diderot, Sorbonne Paris Cité, Paris, France
- Centre for the Developing Brain, Division of Imaging Sciences and Biomedical Engineering, King's College London, King's Health Partners, St. Thomas' Hospital, London, UK
| | - Vincent El Ghouzzi
- PROTECT, INSERM, Université Paris Diderot, Sorbonne Paris Cité, Paris, France.
| |
Collapse
|
42
|
DNA Damage as a Driver for Growth Delay: Chromosome Instability Syndromes with Intrauterine Growth Retardation. BIOMED RESEARCH INTERNATIONAL 2017; 2017:8193892. [PMID: 29238724 PMCID: PMC5702399 DOI: 10.1155/2017/8193892] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/04/2017] [Revised: 06/16/2017] [Accepted: 07/17/2017] [Indexed: 12/20/2022]
Abstract
DNA is constantly exposed to endogenous and exogenous mutagenic stimuli that are capable of producing diverse lesions. In order to protect the integrity of the genetic material, a wide array of DNA repair systems that can target each specific lesion has evolved. Despite the availability of several repair pathways, a common general program known as the DNA damage response (DDR) is stimulated to promote lesion detection, signaling, and repair in order to maintain genetic integrity. The genes that participate in these pathways are subject to mutation; a loss in their function would result in impaired DNA repair and genomic instability. When the DDR is constitutionally altered, every cell of the organism, starting from development, will show DNA damage and subsequent genomic instability. The cellular response to this is either uncontrolled proliferation and cell cycle deregulation that ensues overgrowth, or apoptosis and senescence that result in tissue hypoplasia. These diverging growth abnormalities can clinically translate as cancer or growth retardation; both features can be found in chromosome instability syndromes (CIS). The analysis of the clinical, cellular, and molecular phenotypes of CIS with intrauterine growth retardation allows inferring that replication alteration is their unifying feature.
Collapse
|
43
|
Ogden A, Rida PCG, Aneja R. Centrosome amplification: a suspect in breast cancer and racial disparities. Endocr Relat Cancer 2017; 24:T47-T64. [PMID: 28515047 PMCID: PMC5837860 DOI: 10.1530/erc-17-0072] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Accepted: 05/17/2017] [Indexed: 12/31/2022]
Abstract
The multifaceted involvement of centrosome amplification (CA) in tumorigenesis is coming into focus following years of meticulous experimentation, which have elucidated the powerful abilities of CA to promote cellular invasion, disrupt stem cell division, drive chromosomal instability (CIN) and perturb tissue architecture, activities that can accelerate tumor progression. Integration of the extant in vitro, in vivo and clinical data suggests that in some tissues CA may be a tumor-initiating event, in others a consequential 'hit' in multistep tumorigenesis, and in some others, non-tumorigenic. However, in vivo data are limited and primarily focus on PLK4 (which has CA-independent mechanisms by which it promotes aggressive cellular phenotypes). In vitro breast cancer models suggest that CA can promote tumorigenesis in breast cancer cells in the setting of p53 loss or mutation, which can both trigger CA and promote cellular tolerance to its tendency to slow proliferation and induce aneuploidy. It is thus our perspective that CA is likely an early hit in multistep breast tumorigenesis that may sometimes be lost to preserve aggressive karyotypes acquired through centrosome clustering-mediated CIN, both numerical and structural. We also envision that the robust link between p53 and CA may underlie, to a considerable degree, racial health disparity in breast cancer outcomes. This question is clinically significant because, if it is true, then analysis of centrosomal profiles and administration of centrosome declustering drugs could prove highly efficacious in risk stratifying breast cancers and treating African American (AA) women with breast cancer.
Collapse
Affiliation(s)
- Angela Ogden
- Department of BiologyGeorgia State University, Atlanta, Georgia, USA
| | | | - Ritu Aneja
- Department of BiologyGeorgia State University, Atlanta, Georgia, USA
| |
Collapse
|
44
|
Walz G. Role of primary cilia in non-dividing and post-mitotic cells. Cell Tissue Res 2017; 369:11-25. [PMID: 28361305 PMCID: PMC5487853 DOI: 10.1007/s00441-017-2599-7] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Revised: 02/20/2017] [Accepted: 02/27/2017] [Indexed: 12/12/2022]
Abstract
The essential role of primary (non-motile) cilia during the development of multi-cellular tissues and organs is well established and is underlined by severe disease manifestations caused by mutations in cilia-associated molecules that are collectively termed ciliopathies. However, the role of primary cilia in non-dividing and terminally differentiated, post-mitotic cells is less well understood. Although the prevention of cells from re-entering the cell cycle may represent a major chore, primary cilia have recently been linked to DNA damage responses, autophagy and mitochondria. Given this connectivity, primary cilia in non-dividing cells are well positioned to form a signaling hub outside of the nucleus. Such a center could integrate information to initiate responses and to maintain cellular homeostasis if cell survival is jeopardized. These more discrete functions may remain undetected until differentiated cells are confronted with emergencies.
Collapse
Affiliation(s)
- Gerd Walz
- Renal Division, Department of Medicine, University Freiburg Medical Center, Hugstetter Strasse 55, 79106, Freiburg, Germany.
| |
Collapse
|
45
|
Abstract
The organization of microtubule networks is crucial for controlling chromosome segregation during cell division, for positioning and transport of different organelles, and for cell polarity and morphogenesis. The geometry of microtubule arrays strongly depends on the localization and activity of the sites where microtubules are nucleated and where their minus ends are anchored. Such sites are often clustered into structures known as microtubule-organizing centers, which include the centrosomes in animals and spindle pole bodies in fungi. In addition, other microtubules, as well as membrane compartments such as the cell nucleus, the Golgi apparatus, and the cell cortex, can nucleate, stabilize, and tether microtubule minus ends. These activities depend on microtubule-nucleating factors, such as γ-tubulin-containing complexes and their activators and receptors, and microtubule minus end-stabilizing proteins with their binding partners. Here, we provide an overview of the current knowledge on how such factors work together to control microtubule organization in different systems.
Collapse
Affiliation(s)
- Jingchao Wu
- Cell Biology, Department of Biology, Faculty of Science, Utrecht University, 3584 Utrecht, The Netherlands; ,
| | - Anna Akhmanova
- Cell Biology, Department of Biology, Faculty of Science, Utrecht University, 3584 Utrecht, The Netherlands; ,
| |
Collapse
|
46
|
Rabinowicz N, Mangala LS, Brown KR, Checa-Rodriguez C, Castiel A, Moskovich O, Zarfati G, Trakhtenbrot L, Levy-Barda A, Jiang D, Rodriguez-Aguayo C, Pradeep S, van Praag Y, Lopez-Berestein G, David A, Novikov I, Huertas P, Rottapel R, Sood AK, Izraeli S. Targeting the centriolar replication factor STIL synergizes with DNA damaging agents for treatment of ovarian cancer. Oncotarget 2017; 8:27380-27392. [PMID: 28423708 PMCID: PMC5432342 DOI: 10.18632/oncotarget.16068] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Accepted: 02/20/2017] [Indexed: 01/19/2023] Open
Abstract
Advanced ovarian cancer is an incurable disease. Thus, novel therapies are required. We wished to identify new therapeutic targets for ovarian cancer. ShRNA screen performed in 42 ovarian cancer cell lines identified the centriolar replication factor STIL as an essential gene for ovarian cancer cells. This was verified in-vivo in orthotopic human ovarian cancer mouse models. STIL depletion by administration of siRNA in neutral liposomes resulted in robust anti-tumor effect that was further enhanced in combination with cisplatin. Consistent with this finding, STIL depletion enhanced the extent of DNA double strand breaks caused by DNA damaging agents. This was associated with centrosomal depletion, ongoing genomic instability and enhanced formation of micronuclei. Interestingly, the ongoing DNA damage was not associated with reduced DNA repair. Indeed, we observed that depletion of STIL enhanced canonical homologous recombination repair and increased BRCA1 and RAD51 foci in response to DNA double strand breaks. Thus, inhibition of STIL significantly enhances the efficacy of DNA damaging chemotherapeutic drugs in treatment of ovarian cancer.
Collapse
Affiliation(s)
- Noa Rabinowicz
- Cancer Research Center, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel
- Department of Human Molecular Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Lingegowda S. Mangala
- Department of Gynecologic Oncology, MD Anderson Cancer Center, Houston, Texas, USA
- Center for RNA Interference and Non-Coding RNA, MD Anderson Cancer Center, Houston, Texas, USA
| | - Kevin R. Brown
- Donnelly Centre and The Banting and Best Department of Medical Research, University of Toronto, Toronto, Ontario, Canada
| | - Cintia Checa-Rodriguez
- Department of Genetics, University of Sevilla and Centro Andaluz de Biología Molecular y Medicina Regenerativa (CABIMER), Sevilla, Spain
| | - Asher Castiel
- Cancer Research Center, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel
- Department of Human Molecular Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Oren Moskovich
- Cancer Research Center, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel
- Department of Human Molecular Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Giulia Zarfati
- Cancer Research Center, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel
| | - Luba Trakhtenbrot
- Cancer Research Center, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel
| | - Adva Levy-Barda
- Cancer Research Center, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel
| | - Dahai Jiang
- Department of Gynecologic Oncology, MD Anderson Cancer Center, Houston, Texas, USA
- Center for RNA Interference and Non-Coding RNA, MD Anderson Cancer Center, Houston, Texas, USA
| | - Cristian Rodriguez-Aguayo
- Center for RNA Interference and Non-Coding RNA, MD Anderson Cancer Center, Houston, Texas, USA
- Department of Experimental Therapeutics, MD Anderson Cancer Center, Houston, Texas, USA
| | - Sunila Pradeep
- Department of Gynecologic Oncology, MD Anderson Cancer Center, Houston, Texas, USA
| | - Yael van Praag
- Cancer Research Center, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel
| | - Gabriel Lopez-Berestein
- Center for RNA Interference and Non-Coding RNA, MD Anderson Cancer Center, Houston, Texas, USA
- Department of Experimental Therapeutics, MD Anderson Cancer Center, Houston, Texas, USA
| | - Ahuvit David
- Cancer Research Center, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel
- Department of Human Molecular Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Ilya Novikov
- Biostatistical Unit, Gertner Institute for Epidemiology and Health Policy Research, Ramat Gan, Israel
| | - Pablo Huertas
- Department of Genetics, University of Sevilla and Centro Andaluz de Biología Molecular y Medicina Regenerativa (CABIMER), Sevilla, Spain
| | - Robert Rottapel
- Princess Margaret Cancer Center, University Health Network, Toronto, Ontario, Canada
| | - Anil K. Sood
- Department of Gynecologic Oncology, MD Anderson Cancer Center, Houston, Texas, USA
- Center for RNA Interference and Non-Coding RNA, MD Anderson Cancer Center, Houston, Texas, USA
- Department of Cancer Biology, MD Anderson Cancer Center, Houston, Texas, USA
| | - Shai Izraeli
- Cancer Research Center, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel
- Department of Human Molecular Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- The Gene Development and Environment Pediatric Research Institute, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel
| |
Collapse
|
47
|
Abstract
The maintenance of genome stability is critical for proper cell function, and loss of this stability contributes to many human diseases and developmental disorders. Therefore, cells have evolved partially redundant mechanisms to monitor and protect the genome. One subcellular organelle implicated in the maintenance of genome stability is the centrosome, best known as the primary microtubule organizing center of most animal cells. Centrosomes serve many different roles throughout the cell cycle, and many of those roles, including mitotic spindle assembly, nucleation of the interphase microtubule array, DNA damage response, and efficient cell cycle progression, have been proposed to help maintain genome stability. As a result, the centrosome is itself a highly regulated entity. Here, we review evidence concerning the significance of the centrosome in promoting genome integrity. Recent advances permitting acute and persistent centrosome removal suggest we still have much to learn regarding the specific function and actual importance of centrosomes in different contexts, as well as how cells may compensate for centrosome dysfunction to maintain the integrity of the genome. Although many animal cells survive and proliferate in the absence of centrosomes, they do so aberrantly. Based on these and other studies, we conclude that centrosomes serve as critical, multifunctional organelles that promote genome stability.
Collapse
Affiliation(s)
- Dorothy A Lerit
- Cell Biology and Physiology Center, National Heart, Lung, and Blood Institute; National Institutes of Health, Bethesda, MD, 20892, USA.
- National Institutes of Health, 50 South Drive, Building 50, Room 2122, Bethesda, MD, 20892, USA.
| | - John S Poulton
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, 27599, USA.
- University of North Carolina, Fordham 519, CB#3280, Chapel Hill, NC, 27599, USA.
| |
Collapse
|
48
|
Cosenza MR, Krämer A. Centrosome amplification, chromosomal instability and cancer: mechanistic, clinical and therapeutic issues. Chromosome Res 2016; 24:105-26. [PMID: 26645976 DOI: 10.1007/s10577-015-9505-5] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Centrosomes, the main microtubule-organizing centers in most animal cells, are of crucial importance for the assembly of a bipolar mitotic spindle and subsequent faithful segregation of chromosomes into two daughter cells. Centrosome abnormalities can be found in virtually all cancer types and have been linked to chromosomal instability (CIN) and tumorigenesis. Although our knowledge on centrosome structure, replication, and amplification has greatly increased within recent years, still only very little is known on nature, causes, and consequences of centrosome aberrations in primary tumor tissues. In this review, we summarize our current insights into the mechanistic link between centrosome aberrations, aneuploidy, CIN and tumorigenesis. Mechanisms of induction and cellular consequences of aneuploidy, tetraploidization and CIN, as well as origin and effects of supernumerary centrosomes will be discussed. In addition, animal models for both CIN and centrosome amplification will be outlined. Finally, we describe approaches to exploit centrosome amplification, aneuploidy and CIN for novel and specific anticancer treatment strategies based on the modulation of chromosome missegregation rates.
Collapse
Affiliation(s)
- Marco Raffaele Cosenza
- Clinical Cooperation Unit Molecular Hematology/Oncology, German Cancer Research Center (DKFZ) and Department of Internal Medicine V, University of Heidelberg, Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
| | - Alwin Krämer
- Clinical Cooperation Unit Molecular Hematology/Oncology, German Cancer Research Center (DKFZ) and Department of Internal Medicine V, University of Heidelberg, Im Neuenheimer Feld 280, 69120, Heidelberg, Germany.
| |
Collapse
|
49
|
Johnson CA, Collis SJ. Ciliogenesis and the DNA damage response: a stressful relationship. Cilia 2016; 5:19. [PMID: 27335639 PMCID: PMC4916530 DOI: 10.1186/s13630-016-0040-6] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Accepted: 03/22/2016] [Indexed: 01/27/2023] Open
Abstract
Both inherited and sporadic mutations can give rise to a plethora of human diseases. Through myriad diverse cellular processes, sporadic mutations can arise through a failure to accurately replicate the genetic code or by inaccurate separation of duplicated chromosomes into daughter cells. The human genome has therefore evolved to encode a large number of proteins that work together with regulators of the cell cycle to ensure that it remains error-free. This is collectively known as the DNA damage response (DDR), and genome stability mechanisms involve a complex network of signalling and processing factors that ensure redundancy and adaptability of these systems. The importance of genome stability mechanisms is best illustrated by the dramatic increased risk of cancer in individuals with underlying disruption to genome maintenance mechanisms. Cilia are microtubule-based sensory organelles present on most vertebrate cells, where they facilitate transduction of external signals into the cell. When not embedded within the specialised ciliary membrane, components of the primary cilium's basal body help form the microtubule organising centre that controls cellular trafficking and the mitotic segregation of chromosomes. Ciliopathies are a collection of diseases associated with functional disruption to cilia function through a variety of different mechanisms. Ciliopathy phenotypes can vary widely, and although some cellular overgrowth phenotypes are prevalent in a subset of ciliopathies, an increased risk of cancer is not noted as a clinical feature. However, recent studies have identified surprising genetic and functional links between cilia-associated proteins and genome maintenance factors. The purpose of this mini-review is to therefore highlight some of these discoveries and discuss their implications with regards to functional crosstalk between the DDR and ciliogenesis pathways, and how this may impact on the development of human disease.
Collapse
Affiliation(s)
- Colin A. Johnson
- />Section of Ophthalmology and Neurosciences, Wellcome Trust Brenner Building, Leeds Institute of Molecular Medicine, St. James’s University Hospital, Leeds, LS9 7TF UK
| | - Spencer J. Collis
- />Genome Stability Group, Department of Oncology and Metabolism, Academic Unit of Molecular Oncology, Medical School, University of Sheffield, Beech Hill Road, Sheffield, S10 2RX UK
| |
Collapse
|