1
|
Martino J, Siri SO, Calzetta NL, Paviolo NS, Garro C, Pansa MF, Carbajosa S, Brown AC, Bocco JL, Gloger I, Drewes G, Madauss KP, Soria G, Gottifredi V. Inhibitors of Rho kinases (ROCK) induce multiple mitotic defects and synthetic lethality in BRCA2-deficient cells. eLife 2023; 12:e80254. [PMID: 37073955 PMCID: PMC10185344 DOI: 10.7554/elife.80254] [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: 05/13/2022] [Accepted: 04/18/2023] [Indexed: 04/20/2023] Open
Abstract
The trapping of Poly-ADP-ribose polymerase (PARP) on DNA caused by PARP inhibitors (PARPi) triggers acute DNA replication stress and synthetic lethality (SL) in BRCA2-deficient cells. Hence, DNA damage is accepted as a prerequisite for SL in BRCA2-deficient cells. In contrast, here we show that inhibiting ROCK in BRCA2-deficient cells triggers SL independently from acute replication stress. Such SL is preceded by polyploidy and binucleation resulting from cytokinesis failure. Such initial mitosis abnormalities are followed by other M phase defects, including anaphase bridges and abnormal mitotic figures associated with multipolar spindles, supernumerary centrosomes and multinucleation. SL was also triggered by inhibiting Citron Rho-interacting kinase, another enzyme that, similarly to ROCK, regulates cytokinesis. Together, these observations demonstrate that cytokinesis failure triggers mitotic abnormalities and SL in BRCA2-deficient cells. Furthermore, the prevention of mitotic entry by depletion of Early mitotic inhibitor 1 (EMI1) augmented the survival of BRCA2-deficient cells treated with ROCK inhibitors, thus reinforcing the association between M phase and cell death in BRCA2-deficient cells. This novel SL differs from the one triggered by PARPi and uncovers mitosis as an Achilles heel of BRCA2-deficient cells.
Collapse
Affiliation(s)
| | | | | | | | - Cintia Garro
- Centro de Investigaciones en Bioquímica Clínica e Inmunología, CIBICI-CONICET, Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de CórdobaCórdobaArgentina
- OncoPrecisionCórdobaArgentina
| | - Maria F Pansa
- Centro de Investigaciones en Bioquímica Clínica e Inmunología, CIBICI-CONICET, Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de CórdobaCórdobaArgentina
| | - Sofía Carbajosa
- Centro de Investigaciones en Bioquímica Clínica e Inmunología, CIBICI-CONICET, Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de CórdobaCórdobaArgentina
- OncoPrecisionCórdobaArgentina
| | - Aaron C Brown
- Center for Molecular Medicine, Maine Medical Center Research InstituteScarboroughUnited States
| | - José Luis Bocco
- Centro de Investigaciones en Bioquímica Clínica e Inmunología, CIBICI-CONICET, Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de CórdobaCórdobaArgentina
| | - Israel Gloger
- GlaxoSmithKline-Trust in Science, Global Health R&DStevenageUnited Kingdom
| | - Gerard Drewes
- GlaxoSmithKline-Trust in Science, Global Health R&DStevenageUnited Kingdom
| | - Kevin P Madauss
- GlaxoSmithKline-Trust in Science, Global Health R&DUpper ProvidenceUnited States
| | - Gastón Soria
- Centro de Investigaciones en Bioquímica Clínica e Inmunología, CIBICI-CONICET, Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de CórdobaCórdobaArgentina
- OncoPrecisionCórdobaArgentina
| | | |
Collapse
|
2
|
Gorry R, Brennan K, Lavin PTM, Sheridan R, Mc Gee MM. Phosphorylation of the prolyl isomerase Cyclophilin A regulates its localisation and release from the centrosome during mitosis. Cell Cycle 2023; 22:951-966. [PMID: 36691345 PMCID: PMC10054169 DOI: 10.1080/15384101.2023.2167430] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Accepted: 11/30/2022] [Indexed: 01/25/2023] Open
Abstract
The centrosome acts as a protein platform from which proteins are deployed to function throughout the cell cycle. Previously, we have shown that the prolyl isomerase Cyclophilin A (CypA) localizes to the centrosome in interphase and re-localizes to the midbody during mitosis where it functions in cytokinesis. In this study, investigation of CypA by SDS-PAGE during the cell cycle reveals that it undergoes a mobility shift during mitosis, indicative of a post-translational modification, which may correlate with its subcellular re-localization. Due to the lack of a phospho-specific antibody, we used site-directed mutagenesis to demonstrate that the previously identified serine 77 phosphorylation site within CypA is important for control of CypA centrosome localization. Furthermore, CypA is shown to interact with the mitotic NIMA-related kinase 2 (Nek2) during interphase and mitosis, while also interacting with the Nek2-antagonist PP1 during interphase but not during mitosis, suggesting a potential role for the Nek2-PP1 complex in CypA phospho-regulation. In support of this, Nek2 is capable of phosphorylating CypA in vitro. Overall, this work reveals that phosphorylation of CypA at serine 77 is important for its release from the centrosome during mitosis and may be regulated by the activity of Nek2 and PP1 during the cell cycle.
Collapse
Affiliation(s)
- Rebecca Gorry
- School of Biomolecular and Biomedical Science, University College Dublin, Belfield, Ireland
- Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Ireland
| | - Kieran Brennan
- School of Biomolecular and Biomedical Science, University College Dublin, Belfield, Ireland
- Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Ireland
| | - Paul TM Lavin
- School of Biomolecular and Biomedical Science, University College Dublin, Belfield, Ireland
- Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Ireland
| | - Rebecca Sheridan
- School of Biomolecular and Biomedical Science, University College Dublin, Belfield, Ireland
- Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Ireland
| | - Margaret M Mc Gee
- School of Biomolecular and Biomedical Science, University College Dublin, Belfield, Ireland
- Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Ireland
| |
Collapse
|
3
|
Marocco L, Umrath F, Sachsenmaier S, Rabiner R, Wülker N, Danalache M. 5-Aminolevulinic Acid-Mediated Photodynamic Therapy Potentiates the Effectiveness of Doxorubicin in Ewing Sarcomas. Biomedicines 2022; 10:biomedicines10112900. [PMID: 36428464 PMCID: PMC9687703 DOI: 10.3390/biomedicines10112900] [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: 10/19/2022] [Revised: 11/03/2022] [Accepted: 11/09/2022] [Indexed: 11/16/2022] Open
Abstract
Ewing sarcomas (ES) are aggressive primary bone tumors that require radical therapy. Promising low toxicity, 5-aminolevulinic acid (5-ALA)-mediated photodynamic therapy (PDT) could enhance the effectiveness of conventional treatment modalities (e.g., doxorubicin (DOX)), improving, thus, the anti-tumorigenic effects. In this study, we investigated the effects of DOX and 5-ALA PDT alone or in combination on three different human ES cell lines. Cell viability, reactive oxygen species (ROS) production, and cellular stiffness were measured 24 h after PDT (blue light-wavelength 436 nm with 5-ALA) with or without DOX. ES cell lines have a different sensitivity to the same doses and exposure of 5-ALA PDT. DOX in combination with 5-ALA PDT was found to be effective in impairing the viability of all ES cells while also increasing cytotoxic activity by high ROS production. The stiffness of the ES cells increased significantly (p < 0.05) post treatment. Overall, our results showed that across multiple ES cell lines, 5-ALA PDT can successfully and safely be combined with DOX to potentiate the therapeutic effect. The 5-ALA PDT has the potential to be a highly effective treatment when used alone or in conjunction with other treatments. More research is needed to assess the effectiveness of 5-ALA PDT in in vivo settings.
Collapse
Affiliation(s)
- Lea Marocco
- Laboratory of Cell Biology, Department of Orthopaedic Surgery, University Hospital of Tübingen, 72072 Tübingen, Germany
- Correspondence:
| | - Felix Umrath
- Laboratory of Cell Biology, Department of Orthopaedic Surgery, University Hospital of Tübingen, 72072 Tübingen, Germany
- Department of Oral and Maxillofacial Surgery, University Hospital of Tübingen, 72076 Tübingen, Germany
| | - Saskia Sachsenmaier
- Department of Orthopaedic Surgery, University Hospital of Tübingen, 72076 Tübingen, Germany
| | | | - Nikolaus Wülker
- Department of Orthopaedic Surgery, University Hospital of Tübingen, 72076 Tübingen, Germany
| | - Marina Danalache
- Laboratory of Cell Biology, Department of Orthopaedic Surgery, University Hospital of Tübingen, 72072 Tübingen, Germany
| |
Collapse
|
4
|
Fukuda M, Tojo Y, Sato A, Saito H, Nakanishi A, Miki Y. BRCA2 represses the transcriptional activity of pS2 by E2-ERα. Biochem Biophys Res Commun 2022; 588:75-82. [PMID: 34952473 DOI: 10.1016/j.bbrc.2021.12.054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 12/15/2021] [Indexed: 02/07/2023]
Abstract
Germline mutations to the breast cancer 2 (BRCA2) gene have been associated with hereditary breast cancer. In addition to estrogen uptake, BRCA2 expression increases in the S phase of the cell cycle and largely contributes to DNA damage repair associated with DNA replication. However, the role of BRCA2 in estrogen induction remains unclear. An expression plasmid was created to induce BRCA2 activation upon the addition of estradiol by introducing mutations to the binding sequences for the transcription factors USF1, E2F1, and NF-κB within the promoter region of BRCA2. Then, the estrogen receptor (ER) sites of the proteins that interact with BRCA2 upon the addition of estradiol were identified. Both proteins were bound by the helical domain of BRCA2 and activation function-2 of the ER, suggesting that this binding may regulate the transcriptional activity of pS2, a target gene of the estradiol-ER, by suppressing the binding of SRC-1, a coactivator required for activation of the transcription factor.
Collapse
Affiliation(s)
- Mio Fukuda
- Department of Specialized Surgeries, Tokyo Medical and Dental University (TMDU), 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan
| | - Yo Tojo
- Department of Molecular Genetics, Medical Research Institute, Tokyo Medical and Dental University (TMDU), 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan
| | - Ami Sato
- Department of Molecular Genetics, Medical Research Institute, Tokyo Medical and Dental University (TMDU), 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan
| | - Hiroko Saito
- Department of Genetic Diagnosis, The Cancer Institute, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo, 135-8550, Japan
| | - Akira Nakanishi
- Department of Molecular Genetics, Medical Research Institute, Tokyo Medical and Dental University (TMDU), 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan.
| | - Yoshio Miki
- Department of Molecular Genetics, Medical Research Institute, Tokyo Medical and Dental University (TMDU), 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan; Department of Genetic Diagnosis, The Cancer Institute, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo, 135-8550, Japan.
| |
Collapse
|
5
|
Le HP, Heyer WD, Liu J. Guardians of the Genome: BRCA2 and Its Partners. Genes (Basel) 2021; 12:genes12081229. [PMID: 34440403 PMCID: PMC8394001 DOI: 10.3390/genes12081229] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 08/04/2021] [Accepted: 08/06/2021] [Indexed: 12/28/2022] Open
Abstract
The tumor suppressor BRCA2 functions as a central caretaker of genome stability, and individuals who carry BRCA2 mutations are predisposed to breast, ovarian, and other cancers. Recent research advanced our mechanistic understanding of BRCA2 and its various interaction partners in DNA repair, DNA replication support, and DNA double-strand break repair pathway choice. In this review, we discuss the biochemical and structural properties of BRCA2 and examine how these fundamental properties contribute to DNA repair and replication fork stabilization in living cells. We highlight selected BRCA2 binding partners and discuss their role in BRCA2-mediated homologous recombination and fork protection. Improved mechanistic understanding of how BRCA2 functions in genome stability maintenance can enable experimental evidence-based evaluation of pathogenic BRCA2 mutations and BRCA2 pseudo-revertants to support targeted therapy.
Collapse
Affiliation(s)
- Hang Phuong Le
- Department of Microbiology and Molecular Genetics, University of California, Davis, CA 95616, USA; (H.P.L.); (W.-D.H.)
| | - Wolf-Dietrich Heyer
- Department of Microbiology and Molecular Genetics, University of California, Davis, CA 95616, USA; (H.P.L.); (W.-D.H.)
- Department of Molecular and Cellular Biology, University of California, Davis, CA 95616, USA
| | - Jie Liu
- Department of Microbiology and Molecular Genetics, University of California, Davis, CA 95616, USA; (H.P.L.); (W.-D.H.)
- Correspondence: ; Tel.: +1-530-752-3016
| |
Collapse
|
6
|
Missense Variants of Uncertain Significance: A Powerful Genetic Tool for Function Discovery with Clinical Implications. Cancers (Basel) 2021; 13:cancers13153719. [PMID: 34359619 PMCID: PMC8345083 DOI: 10.3390/cancers13153719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 07/19/2021] [Accepted: 07/21/2021] [Indexed: 11/16/2022] Open
Abstract
Simple Summary Variants of uncertain significance in the breast cancer susceptibility gene BRCA2 represent 50–80% of the results from genetic testing. These mutations may lead to the dysfunction of the gene, thus conferring breast cancer predisposition; however, because they are rare and their impact on the function is not easy to predict, their classification into benign or pathogenic variants remains a challenge. By focusing on three specific rare missense variants identified in breast cancer patients, in this review, we discuss how the functional evaluation of this type of variants can be used to reveal novel activities of BRCA2. Based on these findings, we suggest additional functional tests that might be required for accurate variant classification and how their characterization may be leveraged to find novel clinical strategies for patients bearing these mutations. Abstract The breast cancer susceptibility gene BRCA2 encodes a multifunctional protein required for the accurate repair of DNA double-strand breaks and replicative DNA lesions. In addition, BRCA2 exhibits emerging important roles in mitosis. As a result, mutations in BRCA2 may affect chromosomal integrity in multiple ways. However, many of the BRCA2 mutations found in breast cancer patients and their families are single amino acid substitutions, sometimes unique, and their relevance in cancer risk remains difficult to assess. In this review, we focus on three recent reports that investigated variants of uncertain significance (VUS) located in the N-terminal region of BRCA2. In this framework, we make the case for how the functional evaluation of VUS can be a powerful genetic tool not only for revealing novel aspects of BRCA2 function but also for re-evaluating cancer risk. We argue that other functions beyond homologous recombination deficiency or “BRCAness” may influence cancer risk. We hope our discussion will help the reader appreciate the potential of these functional studies in the prevention and diagnostics of inherited breast and ovarian cancer. Moreover, these novel aspects in BRCA2 function might help find new therapeutic strategies.
Collapse
|
7
|
Julien M, Ghouil R, Petitalot A, Caputo SM, Carreira A, Zinn-Justin S. Intrinsic Disorder and Phosphorylation in BRCA2 Facilitate Tight Regulation of Multiple Conserved Binding Events. Biomolecules 2021; 11:1060. [PMID: 34356684 PMCID: PMC8301801 DOI: 10.3390/biom11071060] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 07/09/2021] [Accepted: 07/13/2021] [Indexed: 12/26/2022] Open
Abstract
The maintenance of genome integrity in the cell is an essential process for the accurate transmission of the genetic material. BRCA2 participates in this process at several levels, including DNA repair by homologous recombination, protection of stalled replication forks, and cell division. These activities are regulated and coordinated via cell-cycle dependent modifications. Pathogenic variants in BRCA2 cause genome instability and are associated with breast and/or ovarian cancers. BRCA2 is a very large protein of 3418 amino acids. Most well-characterized variants causing a strong predisposition to cancer are mutated in the C-terminal 700 residues DNA binding domain of BRCA2. The rest of the BRCA2 protein is predicted to be disordered. Interactions involving intrinsically disordered regions (IDRs) remain difficult to identify both using bioinformatics tools and performing experimental assays. However, the lack of well-structured binding sites provides unique functional opportunities for BRCA2 to bind to a large set of partners in a tightly regulated manner. We here summarize the predictive and experimental arguments that support the presence of disorder in BRCA2. We describe how BRCA2 IDRs mediate self-assembly and binding to partners during DNA double-strand break repair, mitosis, and meiosis. We highlight how phosphorylation by DNA repair and cell-cycle kinases regulate these interactions. We finally discuss the impact of cancer-associated variants on the function of BRCA2 IDRs and more generally on genome stability and cancer risk.
Collapse
Affiliation(s)
- Manon Julien
- Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Université Paris-Sud, 91190 Gif-sur-Yvette, France; (M.J.); (R.G.)
- L’Institut de Biologie Intégrative de la Cellule (I2BC), UMR 9198, Paris-Saclay University, 91190 Gif-sur-Yvette, France;
| | - Rania Ghouil
- Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Université Paris-Sud, 91190 Gif-sur-Yvette, France; (M.J.); (R.G.)
- L’Institut de Biologie Intégrative de la Cellule (I2BC), UMR 9198, Paris-Saclay University, 91190 Gif-sur-Yvette, France;
| | - Ambre Petitalot
- Service de Génétique, Unité de Génétique Constitutionnelle, Institut Curie, 75005 Paris, France; (A.P.); (S.M.C.)
- Institut Curie, Paris Sciences Lettres Research University, 75005 Paris, France
| | - Sandrine M. Caputo
- Service de Génétique, Unité de Génétique Constitutionnelle, Institut Curie, 75005 Paris, France; (A.P.); (S.M.C.)
- Institut Curie, Paris Sciences Lettres Research University, 75005 Paris, France
| | - Aura Carreira
- L’Institut de Biologie Intégrative de la Cellule (I2BC), UMR 9198, Paris-Saclay University, 91190 Gif-sur-Yvette, France;
- Institut Curie, Paris Sciences Lettres Research University, 75005 Paris, France
- Unité Intégrité du Génome, ARN et Cancer, Institut Curie, CNRS UMR3348, 91405 Orsay, France
| | - Sophie Zinn-Justin
- Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Université Paris-Sud, 91190 Gif-sur-Yvette, France; (M.J.); (R.G.)
- L’Institut de Biologie Intégrative de la Cellule (I2BC), UMR 9198, Paris-Saclay University, 91190 Gif-sur-Yvette, France;
| |
Collapse
|
8
|
Ehlén Å, Sessa G, Zinn-Justin S, Carreira A. The phospho-dependent role of BRCA2 on the maintenance of chromosome integrity. Cell Cycle 2021; 20:731-741. [PMID: 33691600 PMCID: PMC8098065 DOI: 10.1080/15384101.2021.1892994] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 01/21/2021] [Accepted: 02/16/2021] [Indexed: 12/18/2022] Open
Abstract
Chromosomal instability is a hallmark of cancer. The tumor suppressor protein BRCA2 performs an important role in the maintenance of genome integrity particularly in interphase; as a mediator of homologous recombination DNA repair pathway, it participates in the repair of DNA double-strand breaks, inter-strand crosslinks and replicative DNA lesions. BRCA2 also protects stalled replication forks from aberrant degradation. Defects in these functions lead to structural chromosomal aberrations. BRCA2 is a large protein containing highly disordered regions that are heavily phosphorylated particularly in mitosis. The functions of these modifications are getting elucidated and reveal emerging activities in chromosome alignment, chromosome segregation and abscission during cell division. Defects in these activities result in numerical chromosomal aberrations. In addition to BRCA2, other factors of the DNA damage response (DDR) participate in mitosis in close association with cell cycle kinases and phosphatases suggesting that the maintenance of genome integrity functions of these factors extends beyond DNA repair. Here we will discuss the regulation of BRCA2 functions through phosphorylation by cell cycle kinases particularly in mitosis, and illustrate with some examples how BRCA2 and other DDR proteins partially rewire their interactions, essentially via phosphorylation, to fulfill mitotic specific functions that ensure chromosome stability.
Collapse
Affiliation(s)
- Åsa Ehlén
- Institut Curie, PSL University, CNRS, UMR3348, Orsay, France
- Paris-Saclay University CNRS, UMR3348, Orsay, France
| | - Gaetana Sessa
- Institut Curie, PSL University, CNRS, UMR3348, Orsay, France
- Paris-Saclay University CNRS, UMR3348, Orsay, France
| | - Sophie Zinn-Justin
- Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Univ Paris-Sud, Université Paris-Saclay, Gif-sur-Yvette Cedex, France
| | - Aura Carreira
- Institut Curie, PSL University, CNRS, UMR3348, Orsay, France
- Paris-Saclay University CNRS, UMR3348, Orsay, France
| |
Collapse
|
9
|
Abstract
Myosins constitute a superfamily of actin-based molecular motor proteins that mediates a variety of cellular activities including muscle contraction, cell migration, intracellular transport, the formation of membrane projections, cell adhesion, and cell signaling. The 12 myosin classes that are expressed in humans share sequence similarities especially in the N-terminal motor domain; however, their enzymatic activities, regulation, ability to dimerize, binding partners, and cellular functions differ. It is becoming increasingly apparent that defects in myosins are associated with diseases including cardiomyopathies, colitis, glomerulosclerosis, neurological defects, cancer, blindness, and deafness. Here, we review the current state of knowledge regarding myosins and disease.
Collapse
|
10
|
Alik A, Bouguechtouli C, Julien M, Bermel W, Ghouil R, Zinn‐Justin S, Theillet F. Sensitivity‐Enhanced
13
C‐NMR Spectroscopy for Monitoring Multisite Phosphorylation at Physiological Temperature and pH. Angew Chem Int Ed Engl 2020; 59:10411-10415. [DOI: 10.1002/anie.202002288] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 03/12/2020] [Indexed: 11/08/2022]
Affiliation(s)
- Ania Alik
- Université Paris-Saclay CEA CNRS Institute for Integrative Biology of the Cell (I2BC) 91198 Gif-sur-Yvette France
| | - Chafiaa Bouguechtouli
- Université Paris-Saclay CEA CNRS Institute for Integrative Biology of the Cell (I2BC) 91198 Gif-sur-Yvette France
| | - Manon Julien
- Université Paris-Saclay CEA CNRS Institute for Integrative Biology of the Cell (I2BC) 91198 Gif-sur-Yvette France
| | - Wolfgang Bermel
- Bruker BioSpin GmbH Silberstreifen 76287 Rheinstetten Germany
| | - Rania Ghouil
- Université Paris-Saclay CEA CNRS Institute for Integrative Biology of the Cell (I2BC) 91198 Gif-sur-Yvette France
| | - Sophie Zinn‐Justin
- Université Paris-Saclay CEA CNRS Institute for Integrative Biology of the Cell (I2BC) 91198 Gif-sur-Yvette France
| | - Francois‐Xavier Theillet
- Université Paris-Saclay CEA CNRS Institute for Integrative Biology of the Cell (I2BC) 91198 Gif-sur-Yvette France
| |
Collapse
|
11
|
Alik A, Bouguechtouli C, Julien M, Bermel W, Ghouil R, Zinn‐Justin S, Theillet F. Sensitivity‐Enhanced
13
C‐NMR Spectroscopy for Monitoring Multisite Phosphorylation at Physiological Temperature and pH. Angew Chem Int Ed Engl 2020. [DOI: 10.1002/ange.202002288] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Affiliation(s)
- Ania Alik
- Université Paris-Saclay CEA CNRS Institute for Integrative Biology of the Cell (I2BC) 91198 Gif-sur-Yvette France
| | - Chafiaa Bouguechtouli
- Université Paris-Saclay CEA CNRS Institute for Integrative Biology of the Cell (I2BC) 91198 Gif-sur-Yvette France
| | - Manon Julien
- Université Paris-Saclay CEA CNRS Institute for Integrative Biology of the Cell (I2BC) 91198 Gif-sur-Yvette France
| | - Wolfgang Bermel
- Bruker BioSpin GmbH Silberstreifen 76287 Rheinstetten Germany
| | - Rania Ghouil
- Université Paris-Saclay CEA CNRS Institute for Integrative Biology of the Cell (I2BC) 91198 Gif-sur-Yvette France
| | - Sophie Zinn‐Justin
- Université Paris-Saclay CEA CNRS Institute for Integrative Biology of the Cell (I2BC) 91198 Gif-sur-Yvette France
| | - Francois‐Xavier Theillet
- Université Paris-Saclay CEA CNRS Institute for Integrative Biology of the Cell (I2BC) 91198 Gif-sur-Yvette France
| |
Collapse
|
12
|
Ehlén Å, Martin C, Miron S, Julien M, Theillet FX, Ropars V, Sessa G, Beaurepere R, Boucherit V, Duchambon P, El Marjou A, Zinn-Justin S, Carreira A. Proper chromosome alignment depends on BRCA2 phosphorylation by PLK1. Nat Commun 2020; 11:1819. [PMID: 32286328 PMCID: PMC7156385 DOI: 10.1038/s41467-020-15689-9] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Accepted: 03/20/2020] [Indexed: 12/18/2022] Open
Abstract
The BRCA2 tumor suppressor protein is involved in the maintenance of genome integrity through its role in homologous recombination. In mitosis, BRCA2 is phosphorylated by Polo-like kinase 1 (PLK1). Here we describe how this phosphorylation contributes to the control of mitosis. We identify a conserved phosphorylation site at T207 of BRCA2 that constitutes a bona fide docking site for PLK1 and is phosphorylated in mitotic cells. We show that BRCA2 bound to PLK1 forms a complex with the phosphatase PP2A and phosphorylated-BUBR1. Reducing BRCA2 binding to PLK1, as observed in BRCA2 breast cancer variants S206C and T207A, alters the tetrameric complex resulting in unstable kinetochore-microtubule interactions, misaligned chromosomes, faulty chromosome segregation and aneuploidy. We thus reveal a role of BRCA2 in the alignment of chromosomes, distinct from its DNA repair function, with important consequences on chromosome stability. These findings may explain in part the aneuploidy observed in BRCA2-mutated tumors.
Collapse
Affiliation(s)
- Åsa Ehlén
- Institut Curie, PSL Research University, CNRS, UMR3348, F-91405, Orsay, France
- Paris Sud University, Paris-Saclay University CNRS, UMR3348, F-91405, Orsay, France
| | - Charlotte Martin
- Institut Curie, PSL Research University, CNRS, UMR3348, F-91405, Orsay, France
- Paris Sud University, Paris-Saclay University CNRS, UMR3348, F-91405, Orsay, France
| | - Simona Miron
- Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Univ Paris-Sud, Université Paris-Saclay, Gif-sur-Yvette, Cedex, France
| | - Manon Julien
- Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Univ Paris-Sud, Université Paris-Saclay, Gif-sur-Yvette, Cedex, France
- Department of Biology, École Normale Supérieure, 94230, Cachan, France
| | - François-Xavier Theillet
- Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Univ Paris-Sud, Université Paris-Saclay, Gif-sur-Yvette, Cedex, France
| | - Virginie Ropars
- Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Univ Paris-Sud, Université Paris-Saclay, Gif-sur-Yvette, Cedex, France
| | - Gaetana Sessa
- Institut Curie, PSL Research University, CNRS, UMR3348, F-91405, Orsay, France
- Paris Sud University, Paris-Saclay University CNRS, UMR3348, F-91405, Orsay, France
| | - Romane Beaurepere
- Institut Curie, PSL Research University, CNRS, UMR3348, F-91405, Orsay, France
- Paris Sud University, Paris-Saclay University CNRS, UMR3348, F-91405, Orsay, France
| | - Virginie Boucherit
- Institut Curie, PSL Research University, CNRS, UMR3348, F-91405, Orsay, France
- Paris Sud University, Paris-Saclay University CNRS, UMR3348, F-91405, Orsay, France
| | - Patricia Duchambon
- Protein Expression and Purification Core Facility, Institut Curie, 26 rue d'Ulm, 75248, Paris, Cedex 05, France
- INSERM U1196, 91405, Orsay, Cedex, France
| | - Ahmed El Marjou
- Protein Expression and Purification Core Facility, Institut Curie, 26 rue d'Ulm, 75248, Paris, Cedex 05, France
- CNRS UMR144, 12 rue Lhomond, 75005, Paris, France
| | - Sophie Zinn-Justin
- Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Univ Paris-Sud, Université Paris-Saclay, Gif-sur-Yvette, Cedex, France.
| | - Aura Carreira
- Institut Curie, PSL Research University, CNRS, UMR3348, F-91405, Orsay, France.
- Paris Sud University, Paris-Saclay University CNRS, UMR3348, F-91405, Orsay, France.
| |
Collapse
|
13
|
Julien M, Miron S, Carreira A, Theillet FX, Zinn-Justin S. 1H, 13C and 15N backbone resonance assignment of the human BRCA2 N-terminal region. BIOMOLECULAR NMR ASSIGNMENTS 2020; 14:79-85. [PMID: 31900740 DOI: 10.1007/s12104-019-09924-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Accepted: 12/20/2019] [Indexed: 06/10/2023]
Abstract
The Breast Cancer susceptibility protein 2 (BRCA2) is involved in mechanisms that maintain genome stability, including DNA repair, replication and cell division. These functions are ensured by the folded C-terminal DNA binding domain of BRCA2 but also by its large regions predicted to be disordered. Several studies have shown that disordered regions of BRCA2 are subjected to phosphorylation, thus regulating BRCA2 interactions through the cell cycle. The N-terminal region of BRCA2 contains two highly conserved clusters of phosphorylation sites between amino acids 75 and 210. Upon phosphorylation by CDK, the cluster 1 is known to become a docking site for the kinase PLK1. The cluster 2 is phosphorylated by PLK1 at least at two positions. Both of these phosphorylation clusters are important for mitosis progression, in particular for chromosome segregation and cytokinesis. In order to identify the phosphorylated residues and to characterize the phosphorylation sites preferences and their functional consequences within BRCA2 N-terminus, we have produced and analyzed the BRCA2 fragment from amino acid 48 to amino acid 284 (BRCA248-284). Here, we report the assignment of 1H, 15N, 13CO, 13Cα and 13Cβ NMR chemical shifts of this region. Analysis of these chemical shifts confirmed that BRCA248-284 shows no stable fold: it is intrinsically disordered, with only short, transient α-helices.
Collapse
Affiliation(s)
- Manon Julien
- Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Univ Paris-Sud, Université Paris-Saclay, Gif-sur-Yvette Cedex, France
- Paris Sud University, Paris-Saclay University CNRS, UMR3348, 91405, Orsay, France
| | - Simona Miron
- Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Univ Paris-Sud, Université Paris-Saclay, Gif-sur-Yvette Cedex, France
| | - Aura Carreira
- Paris Sud University, Paris-Saclay University CNRS, UMR3348, 91405, Orsay, France
- Institut Curie, PSL Research University, UMR3348, 91405, Orsay, France
- CNRS, UMR3348, 91405, Orsay, France
| | - François-Xavier Theillet
- Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Univ Paris-Sud, Université Paris-Saclay, Gif-sur-Yvette Cedex, France
| | - Sophie Zinn-Justin
- Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Univ Paris-Sud, Université Paris-Saclay, Gif-sur-Yvette Cedex, France.
| |
Collapse
|
14
|
Trivedi DV, Nag S, Spudich A, Ruppel KM, Spudich JA. The Myosin Family of Mechanoenzymes: From Mechanisms to Therapeutic Approaches. Annu Rev Biochem 2020; 89:667-693. [PMID: 32169021 DOI: 10.1146/annurev-biochem-011520-105234] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Myosins are among the most fascinating enzymes in biology. As extremely allosteric chemomechanical molecular machines, myosins are involved in myriad pivotal cellular functions and are frequently sites of mutations leading to disease phenotypes. Human β-cardiac myosin has proved to be an excellent target for small-molecule therapeutics for heart muscle diseases, and, as we describe here, other myosin family members are likely to be potentially unique targets for treating other diseases as well. The first part of this review focuses on how myosins convert the chemical energy of ATP hydrolysis into mechanical movement, followed by a description of existing therapeutic approaches to target human β-cardiac myosin. The next section focuses on the possibility of targeting nonmuscle members of the human myosin family for several diseases. We end the review by describing the roles of myosin in parasites and the therapeutic potential of targeting them to block parasitic invasion of their hosts.
Collapse
Affiliation(s)
- Darshan V Trivedi
- Department of Biochemistry, Stanford University School of Medicine, Stanford, California 94305, USA; , , .,Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Suman Nag
- MyoKardia Inc., Brisbane, California 94005, USA;
| | - Annamma Spudich
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bangalore 560-097, India;
| | - Kathleen M Ruppel
- Department of Biochemistry, Stanford University School of Medicine, Stanford, California 94305, USA; , , .,Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, California 94305, USA.,Division of Pediatric Cardiology, Department of Pediatrics, Stanford University School of Medicine, Stanford, California 94305, USA
| | - James A Spudich
- Department of Biochemistry, Stanford University School of Medicine, Stanford, California 94305, USA; , , .,Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, California 94305, USA
| |
Collapse
|
15
|
The C. elegans BRCA2-ALP/Enigma Complex Regulates Axon Regeneration via a Rho GTPase-ROCK-MLC Phosphorylation Pathway. Cell Rep 2019; 24:1880-1889. [PMID: 30110643 DOI: 10.1016/j.celrep.2018.07.049] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Revised: 05/28/2018] [Accepted: 07/16/2018] [Indexed: 12/11/2022] Open
Abstract
The ability of specific neurons to regenerate their axons after injury is governed by cell-intrinsic regeneration pathways. However, the mechanisms regulating axon regeneration are not well understood. Here, we identify the brc-2 gene encoding a homolog of the mammalian BRCA2 tumor suppressor as a regulator of axon regeneration in Caenorhabditis elegans motor neurons. We show that the RHO-1/Rho GTPase-LET-502/ROCK (Rho-associated coiled-coil kinase)-regulatory non-muscle myosin light-chain (MLC-4/MLC) phosphorylation signaling pathway regulates axon regeneration. BRC-2 functions between RHO-1 and LET-502, suggesting that BRC-2 is required for the activation of LET-502 by RHO-1-GTP. We also find that one component that interacts with BRC-2, the ALP (α-actinin-associated LIM protein)/Enigma protein ALP-1, is required for regeneration and acts between LET-502 and MLC-4 phosphorylation. Furthermore, we demonstrate that ALP-1 associates with LET-502 and MLC-4. Thus, ALP-1 serves as a platform to activate MLC-4 phosphorylation mediated by the RHO-1-LET-502 signaling pathway.
Collapse
|
16
|
Halder D, Saha S, Singh RK, Ghosh I, Mallick D, Dey SK, Ghosh A, Das BB, Ghosh S, Jana SS. Nonmuscle myosin IIA and IIB differentially modulate migration and alter gene expression in primary mouse tumorigenic cells. Mol Biol Cell 2019; 30:1463-1476. [PMID: 30995168 PMCID: PMC6724700 DOI: 10.1091/mbc.e18-12-0790] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Revised: 04/04/2019] [Accepted: 04/10/2019] [Indexed: 12/22/2022] Open
Abstract
Though many cancers are known to show up-regulation of nonmuscle myosin (NM) IIA and IIB, the mechanism by which NMIIs aid in cancer development remains unexplored. Here we demonstrate that tumor-generating, fibroblast-like cells isolated from 3-methylcholanthrene (3MC)-induced murine tumor exhibit distinct phospho-dependent localization of NMIIA and NMIIB at the perinuclear area and tip of the filopodia and affect cell migration differentially. While NMIIA-KD affects protrusion dynamics and increases cell directionality, NMIIB-KD lowers migration speed and increases filopodial branching. Strategically located NMIIs at the perinuclear area colocalize with the linker of nucleoskeleton and cytoskeleton (LINC) protein Nesprin2 and maintain the integrity of the nuclear-actin cap. Interestingly, knockdown of NMIIs results in altered expression of genes involved in epithelial-to-mesenchymal transition, angiogenesis, and cellular senescence. NMIIB-KD cells display down-regulation of Gsc and Serpinb2, which is strikingly similar to Nesprin2-KD cells as assessed by quantitative PCR analysis. Further gene network analysis predicts that NMIIA and NMIIB may act on similar pathways but through different regulators. Concomitantly, knockdown of NMIIA or NMIIB lowers the growth rate and tumor volume of 3MC-induced tumor in vivo. Altogether, these results open a new window to further investigate the effect of LINC-associated perinuclear actomyosin complex on mechanoresponsive gene expression in the growing tumor.
Collapse
Affiliation(s)
- Debdatta Halder
- School of Biological Sciences, Indian Association for the Cultivation of Science, Jadavpur, Kolkata 700032, India
| | - Shekhar Saha
- School of Biological Sciences, Indian Association for the Cultivation of Science, Jadavpur, Kolkata 700032, India
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, VA 22908
| | - Raman K. Singh
- School of Biological Sciences, Indian Association for the Cultivation of Science, Jadavpur, Kolkata 700032, India
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 7610010, Israel
| | - Indranil Ghosh
- School of Biological Sciences, Indian Association for the Cultivation of Science, Jadavpur, Kolkata 700032, India
| | - Ditipriya Mallick
- School of Biological Sciences, Indian Association for the Cultivation of Science, Jadavpur, Kolkata 700032, India
| | - Sumit K. Dey
- School of Biological Sciences, Indian Association for the Cultivation of Science, Jadavpur, Kolkata 700032, India
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY 14853
| | - Arijit Ghosh
- School of Biological Sciences, Indian Association for the Cultivation of Science, Jadavpur, Kolkata 700032, India
| | - Benu Brata Das
- School of Biological Sciences, Indian Association for the Cultivation of Science, Jadavpur, Kolkata 700032, India
| | | | - Siddhartha S. Jana
- School of Biological Sciences, Indian Association for the Cultivation of Science, Jadavpur, Kolkata 700032, India
| |
Collapse
|
17
|
Dias KB, Flores APC, Hildebrand LC, de Oliveira MG, Lamers ML, Rados PV, Magnusson AS, Filho MS. Non-muscle myosin II as a predictive factor in head and neck squamous cell carcinoma. Med Oral Patol Oral Cir Bucal 2019; 24:e346-e353. [PMID: 31011146 PMCID: PMC6530949 DOI: 10.4317/medoral.22898] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Accepted: 02/27/2019] [Indexed: 12/11/2022] Open
Abstract
Background The present study attempted to provide information regarding non-muscle myosin II (MII) isoforms immunoreactivity in patients with head and neck squamous cell carcinoma (HNSCC) and analysis of the patients’ clinical status after 5 years of monitoring. Material and Methods A semiquantitative analysis of the immunoreactivity of the MII isoforms was performed in 54 surgical specimens and its correlation with clinical and pathological variables and prognosis was verified. Data were analyzed using chi-square, Mann-Whitney and Kruskal-Wallis tests. To evaluate the survival over the total monitoring time and any connection with the proteins studied, the Kaplan-Meier analysis was used. P values ≤0.05 were considered statistically significant. Results In the advanced stages of pathological tumor-node-metastasis, the expression of MIIB in adjacent non-neoplastic epithelial tissues tended to increase (p = 0.057). In tumoral zones there was an association of high expression among the three isoforms (MIIA/MIIB p=0,001, MIIB/MIIC p=0,006 and MIIA/MIIC p=0,012). Negative clinical evolution in patients was directly correlated to increased MIIC expression in the tumoral zone of invasion in HNSCC (p = 0.017). Based on clinical evolution after the monitoring period, patients with tumors expressing MIIC had poorer prognoses (p = 0.048). Conclusions The present study suggests that MIIB expression in non-neoplastic adjacent epithelial tissues may indicate a potential for regional metastasis and that MIIC expression in the tumoral zone of invasion is predictive of negative evolution of the disease. Key words:Head and neck squamous cell carcinoma, oral cancer, myosin type II, non-muscle myosin, immunohistochemistry.
Collapse
Affiliation(s)
- K-B Dias
- Departmentof Oral Pathology, School of Dentistry, Universidade Federal do Rio Grande do Sul, Av Ramiro Barcelos 2492/503, 90035-003 - Porto Alegre, Rio Grande do Sul, Brazil,
| | | | | | | | | | | | | | | |
Collapse
|
18
|
Abstract
Fanconi anaemia (FA) is a genetic disorder that is characterized by bone marrow failure (BMF), developmental abnormalities and predisposition to cancer. Together with other proteins involved in DNA repair processes and cell division, the FA proteins maintain genome homeostasis, and germline mutation of any one of the genes that encode FA proteins causes FA. Monoallelic inactivation of some FA genes, such as FA complementation group D1 (FANCD1; also known as the breast and ovarian cancer susceptibility gene BRCA2), leads to adult-onset cancer predisposition but does not cause FA, and somatic mutations in FA genes occur in cancers in the general population. Carcinogenesis resulting from a dysregulated FA pathway is multifaceted, as FA proteins monitor multiple complementary genome-surveillance checkpoints throughout interphase, where monoubiquitylation of the FANCD2-FANCI heterodimer by the FA core complex promotes recruitment of DNA repair effectors to chromatin lesions to resolve DNA damage and mitosis. In this Review, we discuss how the FA pathway safeguards genome integrity throughout the cell cycle and show how studies of FA have revealed opportunities to develop rational therapeutics for this genetic disease and for malignancies that acquire somatic mutations within the FA pathway.
Collapse
Affiliation(s)
- Grzegorz Nalepa
- Department of Pediatrics, Section of Pediatric Hematology-Oncology, Wells Center for Pediatric Research, Indiana University School of Medicine, 1044 W Walnut Street, R4-421, Indianapolis, Indiana 46202, USA
- Riley Hospital for Children at Indiana University Health, 705 Riley Hospital Drive, Room 5900, Indianapolis, Indiana 46202, USA
- Department of Biochemistry, Indiana University School of Medicine
- Department of Medical and Molecular Genetics, Indiana University School of Medicine
| | - D Wade Clapp
- Riley Hospital for Children at Indiana University Health, 705 Riley Hospital Drive, Room 5900, Indianapolis, Indiana 46202, USA
- Department of Biochemistry, Indiana University School of Medicine
- Department of Microbiology and Immunology, Indiana University School of Medicine
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana, 46202, USA
| |
Collapse
|
19
|
Liao Y, Lin D, Cui P, Abbasi B, Chen C, Zhang Z, Zhang Y, Dong Y, Rui R, Ju S. Polo-like kinase 1 inhibition results in misaligned chromosomes and aberrant spindles in porcine oocytes during the first meiotic division. Reprod Domest Anim 2018; 53:256-265. [PMID: 29143380 DOI: 10.1111/rda.13102] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2017] [Accepted: 09/26/2017] [Indexed: 01/15/2023]
Abstract
Polo-like kinase 1 (Plk1), a type of serine/threonine protein kinase, has been implicated in various functions in the regulation of mitotic processes. However, these kinase's roles in meiotic division are not fully understood, particularly in the meiotic maturation of porcine oocytes. In this study, the expression and spatiotemporal localization of Plk1 were initially assessed in the meiotic process of pig oocytes by utilizing Western blotting with immunofluorescent staining combined with confocal microscopy imaging technique. The results showed that Plk1 was expressed and exhibited a dynamic subcellular localization throughout the meiotic process. After germinal vesicle breakdown (GVBD), Plk1 was detected prominently around the condensed chromosomes and subsequently exhibited a similar subcellular localization to α-tubulin throughout subsequent meiotic phases, with particular enrichment being observed near spindle poles at MI and MII. Inhibition of Plk1 via a highly selective inhibitor, GSK461364, led to the failure of first polar body extrusion in porcine oocytes, with the majority of the treated oocytes being arrested in GVBD. Further subcellular structure examination results indicated that Plk1 inhibition caused the great majority of oocytes with spindle abnormalities and chromosome misalignment during the first meiotic division. The results of this study illustrate that Plk1 is critical for the first meiotic division in porcine oocytes through its influence on spindle organization and chromosome alignment, which further affects the ensuing meiotic cell cycle progression.
Collapse
Affiliation(s)
- Y Liao
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - D Lin
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - P Cui
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - B Abbasi
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - C Chen
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Z Zhang
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Y Zhang
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Y Dong
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - R Rui
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - S Ju
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| |
Collapse
|
20
|
Chinthalapudi K, Heissler SM, Preller M, Sellers JR, Manstein DJ. Mechanistic insights into the active site and allosteric communication pathways in human nonmuscle myosin-2C. eLife 2017; 6:32742. [PMID: 29256864 PMCID: PMC5749951 DOI: 10.7554/elife.32742] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Accepted: 12/18/2017] [Indexed: 01/10/2023] Open
Abstract
Despite a generic, highly conserved motor domain, ATP turnover kinetics and their activation by F-actin vary greatly between myosin-2 isoforms. Here, we present a 2.25 Å pre-powerstroke state (ADP⋅VO4) crystal structure of the human nonmuscle myosin-2C motor domain, one of the slowest myosins characterized. In combination with integrated mutagenesis, ensemble-solution kinetics, and molecular dynamics simulation approaches, the structure reveals an allosteric communication pathway that connects the distal end of the motor domain with the active site. Disruption of this pathway by mutation of hub residue R788, which forms the center of a cluster of interactions connecting the converter, the SH1-SH2 helix, the relay helix, and the lever, abolishes nonmuscle myosin-2 specific kinetic signatures. Our results provide insights into structural changes in the myosin motor domain that are triggered upon F-actin binding and contribute critically to the mechanochemical behavior of stress fibers, actin arcs, and cortical actin-based structures.
Collapse
Affiliation(s)
- Krishna Chinthalapudi
- Institute for Biophysical Chemistry, OE4350, Hannover Medical School, Hannover, Germany.,Division for Structural Biochemistry, OE8830, Hannover Medical School, Hannover, Germany.,Cell Adhesion Laboratory, Department of Integrative Structural and Computational Biology, The Scripps Research Institute, Jupiter, United States
| | - Sarah M Heissler
- Institute for Biophysical Chemistry, OE4350, Hannover Medical School, Hannover, Germany.,Laboratory of Molecular Physiology, NHLBI, National Institutes of Health, Bethesda, United States
| | - Matthias Preller
- Institute for Biophysical Chemistry, OE4350, Hannover Medical School, Hannover, Germany.,Centre for Structural Systems Biology (CSSB), German Electron Synchrotron (DESY), Hamburg, Germany
| | - James R Sellers
- Laboratory of Molecular Physiology, NHLBI, National Institutes of Health, Bethesda, United States
| | - Dietmar J Manstein
- Institute for Biophysical Chemistry, OE4350, Hannover Medical School, Hannover, Germany.,Division for Structural Biochemistry, OE8830, Hannover Medical School, Hannover, Germany
| |
Collapse
|
21
|
Zhang Z, Chen C, Ma L, Yu Q, Li S, Abbasi B, Yang J, Rui R, Ju S. Plk1 is essential for proper chromosome segregation during meiosis I/meiosis II transition in pig oocytes. Reprod Biol Endocrinol 2017; 15:69. [PMID: 28851440 PMCID: PMC5575893 DOI: 10.1186/s12958-017-0289-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Accepted: 08/13/2017] [Indexed: 02/23/2023] Open
Abstract
BACKGROUND Polo-like kinase 1 (Plk1), as a characteristic regulator in meiosis, organizes multiple biological events of cell division. Although Plk1 has been implicated in various functions in somatic cell mitotic processes, considerably less is known regarding its function during the transition from metaphase I (MI) to metaphase II (MII) stage in oocyte meiotic progression. METHODS In this study, the possible role of Plk1 during the MI-to-MII stage transition in pig oocytes was addressed. Initially, the spatiotemporal expression and subcellular localization pattern of Plk1 were revealed in pig oocytes from MI to MII stage using indirect immunofluorescence and confocal microscopy imaging techniques combined with western blot analyses. Moreover, a highly selective Plk1 inhibitor, GSK461364, was used to determine the potential role of Plk1 during this MI-to-MII transition progression. RESULTS Upon expression, Plk1 exhibited a specific dynamic intracellular localization, and co-localization of Plk1 with α-tubulin was revealed in the meiotic spindle of pig oocyte during the transition from MI to MII stage. GSK461364 treatment significantly blocked the first polar body (pbI) emission in a dose-dependent manner and resulted in a failure of meiotic maturation, with a larger percentage of the GSK461364-treated oocytes arresting in the anaphase-telophase I (ATI) stage. Further subcellular structure examination results showed that inhibition of Plk1 with GSK461364 had no visible effect on spindle assembly but caused a significantly higher proportion of the treated oocytes to have obvious defects in homologous chromosome segregation at ATI stage. CONCLUSIONS Thus, these results indicate that Plk1 plays an essential role during the meiosis I/meiosis II transition in porcine oocytes, and the regulation is associated with Plk1's effects on homologous chromosome segregation in the ATI stage.
Collapse
Affiliation(s)
- Zixiao Zhang
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095 China
| | - Changchao Chen
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095 China
| | - Liying Ma
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095 China
| | - Qiuchen Yu
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095 China
| | - Shuai Li
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095 China
| | - Benazir Abbasi
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095 China
| | - Jiayi Yang
- Nanjing Foreign Languages School, Nanjing, 210008 China
| | - Rong Rui
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095 China
| | - Shiqiang Ju
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095 China
| |
Collapse
|
22
|
Meyer S, Stevens A, Paredes R, Schneider M, Walker MJ, Williamson AJK, Gonzalez-Sanchez MB, Smetsers S, Dalal V, Teng HY, White DJ, Taylor S, Muter J, Pierce A, de Leonibus C, Rockx DAP, Rooimans MA, Spooncer E, Stauffer S, Biswas K, Godthelp B, Dorsman J, Clayton PE, Sharan SK, Whetton AD. Acquired cross-linker resistance associated with a novel spliced BRCA2 protein variant for molecular phenotyping of BRCA2 disruption. Cell Death Dis 2017; 8:e2875. [PMID: 28617445 PMCID: PMC5520920 DOI: 10.1038/cddis.2017.264] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Revised: 03/29/2017] [Accepted: 05/05/2017] [Indexed: 12/25/2022]
Abstract
BRCA2 encodes a protein with a fundamental role in homologous recombination that is essential for normal development. Carrier status of mutations in BRCA2 is associated with familial breast and ovarian cancer, while bi-allelic BRCA2 mutations can cause Fanconi anemia (FA), a cancer predisposition syndrome with cellular cross-linker hypersensitivity. Cancers associated with BRCA2 mutations can acquire chemo-resistance on relapse. We modeled acquired cross-linker resistance with an FA-derived BRCA2-mutated acute myeloid leukemia (AML) platform. Associated with acquired cross-linker resistance was the expression of a functional BRCA2 protein variant lacking exon 5 and exon 7 (BRCA2ΔE5+7), implying a role for BRCA2 splicing for acquired chemo-resistance. Integrated network analysis of transcriptomic and proteomic differences for phenotyping of BRCA2 disruption infers impact on transcription and chromatin remodeling in addition to the DNA damage response. The striking overlap with transcriptional profiles of FA patient hematopoiesis and BRCA mutation associated ovarian cancer helps define and explicate the ‘BRCAness’ profile.
Collapse
Affiliation(s)
- Stefan Meyer
- Stem Cell &Leukaemia Proteomics Laboratory, Manchester Cancer Research Centre, Division of Molecular and Clinical Cancer Sciences, Faculty of Biology, Medicine &Health, University of Manchester, Manchester, UK.,Manchester Academic Health Science Centre, Manchester, UK.,Department of Paediatric and Adolescent Oncology, Royal Manchester Children's Hospital, Manchester, UK.,Young Oncology Unit, Christie Hospital, Manchester, UK
| | - Adam Stevens
- Manchester Academic Health Science Centre, Manchester, UK.,Department of Paediatric Endocrinology, Faculty of Biology, Medicine &Health, University of Manchester, Manchester, UK
| | - Roberto Paredes
- Stem Cell &Leukaemia Proteomics Laboratory, Manchester Cancer Research Centre, Division of Molecular and Clinical Cancer Sciences, Faculty of Biology, Medicine &Health, University of Manchester, Manchester, UK.,Manchester Academic Health Science Centre, Manchester, UK
| | - Marion Schneider
- Stem Cell &Leukaemia Proteomics Laboratory, Manchester Cancer Research Centre, Division of Molecular and Clinical Cancer Sciences, Faculty of Biology, Medicine &Health, University of Manchester, Manchester, UK.,Manchester Academic Health Science Centre, Manchester, UK
| | - Michael J Walker
- Stem Cell &Leukaemia Proteomics Laboratory, Manchester Cancer Research Centre, Division of Molecular and Clinical Cancer Sciences, Faculty of Biology, Medicine &Health, University of Manchester, Manchester, UK.,Manchester Academic Health Science Centre, Manchester, UK
| | - Andrew J K Williamson
- Stem Cell &Leukaemia Proteomics Laboratory, Manchester Cancer Research Centre, Division of Molecular and Clinical Cancer Sciences, Faculty of Biology, Medicine &Health, University of Manchester, Manchester, UK.,Manchester Academic Health Science Centre, Manchester, UK
| | - Maria-Belen Gonzalez-Sanchez
- Stem Cell &Leukaemia Proteomics Laboratory, Manchester Cancer Research Centre, Division of Molecular and Clinical Cancer Sciences, Faculty of Biology, Medicine &Health, University of Manchester, Manchester, UK.,Manchester Academic Health Science Centre, Manchester, UK
| | - Stephanie Smetsers
- Department of Clinical Genetics, Section Oncogenetics, VU University Medical Center, Amsterdam, The Netherlands
| | - Vineet Dalal
- Stem Cell &Leukaemia Proteomics Laboratory, Manchester Cancer Research Centre, Division of Molecular and Clinical Cancer Sciences, Faculty of Biology, Medicine &Health, University of Manchester, Manchester, UK.,Manchester Academic Health Science Centre, Manchester, UK
| | - Hsiang Ying Teng
- Stem Cell &Leukaemia Proteomics Laboratory, Manchester Cancer Research Centre, Division of Molecular and Clinical Cancer Sciences, Faculty of Biology, Medicine &Health, University of Manchester, Manchester, UK.,Manchester Academic Health Science Centre, Manchester, UK
| | - Daniel J White
- Stem Cell &Leukaemia Proteomics Laboratory, Manchester Cancer Research Centre, Division of Molecular and Clinical Cancer Sciences, Faculty of Biology, Medicine &Health, University of Manchester, Manchester, UK.,Manchester Academic Health Science Centre, Manchester, UK
| | - Sam Taylor
- Stem Cell &Leukaemia Proteomics Laboratory, Manchester Cancer Research Centre, Division of Molecular and Clinical Cancer Sciences, Faculty of Biology, Medicine &Health, University of Manchester, Manchester, UK.,Manchester Academic Health Science Centre, Manchester, UK
| | - Joanne Muter
- Stem Cell &Leukaemia Proteomics Laboratory, Manchester Cancer Research Centre, Division of Molecular and Clinical Cancer Sciences, Faculty of Biology, Medicine &Health, University of Manchester, Manchester, UK.,Manchester Academic Health Science Centre, Manchester, UK
| | - Andrew Pierce
- Stem Cell &Leukaemia Proteomics Laboratory, Manchester Cancer Research Centre, Division of Molecular and Clinical Cancer Sciences, Faculty of Biology, Medicine &Health, University of Manchester, Manchester, UK.,Manchester Academic Health Science Centre, Manchester, UK
| | - Chiara de Leonibus
- Manchester Academic Health Science Centre, Manchester, UK.,Department of Paediatric Endocrinology, Faculty of Biology, Medicine &Health, University of Manchester, Manchester, UK
| | - Davy A P Rockx
- Department of Clinical Genetics, Section Oncogenetics, VU University Medical Center, Amsterdam, The Netherlands
| | - Martin A Rooimans
- Department of Clinical Genetics, Section Oncogenetics, VU University Medical Center, Amsterdam, The Netherlands
| | - Elaine Spooncer
- Stem Cell &Leukaemia Proteomics Laboratory, Manchester Cancer Research Centre, Division of Molecular and Clinical Cancer Sciences, Faculty of Biology, Medicine &Health, University of Manchester, Manchester, UK.,Manchester Academic Health Science Centre, Manchester, UK
| | - Stacey Stauffer
- Mouse Cancer Genetics Program; Center for Cancer Research; Frederick National Laboratory for Cancer Research; National Cancer Institute, Frederick, MD, USA
| | - Kajal Biswas
- Mouse Cancer Genetics Program; Center for Cancer Research; Frederick National Laboratory for Cancer Research; National Cancer Institute, Frederick, MD, USA
| | - Barbara Godthelp
- Department of Toxicogenetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Josephine Dorsman
- Department of Clinical Genetics, Section Oncogenetics, VU University Medical Center, Amsterdam, The Netherlands
| | - Peter E Clayton
- Manchester Academic Health Science Centre, Manchester, UK.,Department of Paediatric Endocrinology, Faculty of Biology, Medicine &Health, University of Manchester, Manchester, UK
| | - Shyam K Sharan
- Mouse Cancer Genetics Program; Center for Cancer Research; Frederick National Laboratory for Cancer Research; National Cancer Institute, Frederick, MD, USA
| | - Anthony D Whetton
- Stem Cell &Leukaemia Proteomics Laboratory, Manchester Cancer Research Centre, Division of Molecular and Clinical Cancer Sciences, Faculty of Biology, Medicine &Health, University of Manchester, Manchester, UK.,Manchester Academic Health Science Centre, Manchester, UK.,Stoller Biomarker Discovery Centre, University of Manchester, Manchester, UK
| |
Collapse
|
23
|
Zhang Z, Chen C, Cui P, Liao Y, Yao L, Zhang Y, Rui R, Ju S. Plk1 inhibition leads to a failure of mitotic division during the first mitotic division in pig embryos. J Assist Reprod Genet 2017; 34:399-407. [PMID: 28074435 PMCID: PMC5360688 DOI: 10.1007/s10815-016-0864-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Accepted: 12/19/2016] [Indexed: 12/01/2022] Open
Abstract
PURPOSE This study was conducted to examine the dynamic distribution of polo-like 1 kinase (Plk1) and the possible role it plays in first mitotic division during early porcine embryo development. METHODS Indirect immunofluorescence and confocal microscopy imaging techniques combined with western blot analyses were used to study the dynamic expression and subcellular localization of Plk1 protein in pig parthenogenetic embryos. Finally, a selective Plk1 inhibitor, GSK461364, was used to evaluate the potential role of Plk1 during this special stage. RESULTS The results showed that Plk1 upon expression exhibited specific dynamic intracellular localization, which closely correlated with the α-tubulin distribution during the first mitotic division. GSK461364 treatment resulted in cleavage failure, with majority of the GSK461364-treated embryos being arrested in prometaphase. Further results of the subcellular structure examination showed that GSK461364 treatment led to a significantly higher proportion of the treated embryos having abnormal spindles and misarranged chromosomes at the prometaphase stage. CONCLUSIONS Thus, these results indicated that Plk1 is essential for porcine embryos to complete the first mitotic division. Furthermore, Plk1 regulation was associated with effects on spindle assembly and chromosome arrangement.
Collapse
Affiliation(s)
- Zixiao Zhang
- College of Veterinary Medicine, Nanjing Agricultural University, Jiangsu, 210095, China
| | - Changchao Chen
- College of Veterinary Medicine, Nanjing Agricultural University, Jiangsu, 210095, China
| | - Panpan Cui
- College of Veterinary Medicine, Nanjing Agricultural University, Jiangsu, 210095, China
| | - Yaya Liao
- College of Veterinary Medicine, Nanjing Agricultural University, Jiangsu, 210095, China
| | - Lingyun Yao
- College of Veterinary Medicine, Nanjing Agricultural University, Jiangsu, 210095, China
| | - Yue Zhang
- College of Veterinary Medicine, Nanjing Agricultural University, Jiangsu, 210095, China
| | - Rong Rui
- College of Veterinary Medicine, Nanjing Agricultural University, Jiangsu, 210095, China
| | - Shiqiang Ju
- College of Veterinary Medicine, Nanjing Agricultural University, Jiangsu, 210095, China.
| |
Collapse
|
24
|
Saha S, Halder D, Goswami S, Jana SS. N-terminal polar amino acids of the C2 insert of nonmuscle myosin II-C2 regulate its functional properties. FEBS Lett 2016; 590:4223-4232. [PMID: 27714782 DOI: 10.1002/1873-3468.12446] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Revised: 09/10/2016] [Accepted: 09/13/2016] [Indexed: 11/10/2022]
Abstract
In this study, we investigated the regions in the alternatively spliced C2 insert of nonmuscle myosin (NM) II-C conferring unique functional properties to the protein. We used constructs carrying deletions within different regions of C2 in neuronal cells; namely, the polar N terminus, the proline/serine-rich middle, and the nonpolar C terminus. We compared the wild-type NM II-C2 and deletion mutants with respect to ATPase activity, coassembly with NM II-B, regulation by myosin light-chain kinase (MLCK), and solubility, to determine the C2 region(s) involved in these processes. In addition, we examined the ability of the mutants to rescue the neurite-shortening phenotype upon NM II-C2 knockdown in Neuro-2a cells. Our data highlight the importance of the polar N terminus in NM II-C2 function.
Collapse
Affiliation(s)
- Shekhar Saha
- Department of Biological Chemistry, Indian Association for the Cultivation of Science, Kolkata, India
| | - Debdatta Halder
- Department of Biological Chemistry, Indian Association for the Cultivation of Science, Kolkata, India
| | - Swagata Goswami
- Department of Biological Chemistry, Indian Association for the Cultivation of Science, Kolkata, India
| | - Siddhartha S Jana
- Department of Biological Chemistry, Indian Association for the Cultivation of Science, Kolkata, India
| |
Collapse
|
25
|
Abstract
Mechanotransduction is the process through which cells survey the mechanical properties of their environment, convert these mechanical inputs into biochemical signals, and modulate their phenotype in response. These mechanical inputs, which may be encoded in the form of extracellular matrix stiffness, dimensionality, and adhesion, all strongly influence cell morphology, migration, and fate decisions. One mechanism through which cells on planar or pseudo-planar matrices exert tensile forces and interrogate microenvironmental mechanics is through stress fibers, which are bundles composed of actin filaments and, in most cases, non-muscle myosin II filaments. Stress fibers form a continuous structural network that is mechanically coupled to the extracellular matrix through focal adhesions. Furthermore, myosin-driven contractility plays a central role in the ability of stress fibers to sense matrix mechanics and generate tension. Here, we review the distinct roles that non-muscle myosin II plays in driving mechanosensing and focus specifically on motility. In a closely related discussion, we also describe stress fiber classification schemes and the differing roles of various myosin isoforms in each category. Finally, we briefly highlight recent studies exploring mechanosensing in three-dimensional environments, in which matrix content, structure, and mechanics are often tightly interrelated. Stress fibers and the myosin motors therein represent an intriguing and functionally important biological system in which mechanics, biochemistry, and architecture all converge.
Collapse
Affiliation(s)
- Stacey Lee
- Department of Bioengineering, University of California, Berkeley, Berkeley, CA, 94720, USA; UC Berkeley-UCSF Graduate Program in Bioengineering, University of California, Berkeley, Berkeley, CA, 94720, USA
| | - Sanjay Kumar
- Department of Bioengineering, University of California, Berkeley, Berkeley, CA, 94720, USA; UC Berkeley-UCSF Graduate Program in Bioengineering, University of California, Berkeley, Berkeley, CA, 94720, USA; Department of Chemical and Biomolecular Engineering, University of California, Berkeley, Berkeley, CA, 94720, USA
| |
Collapse
|
26
|
Martinez JS, Baldeyron C, Carreira A. Molding BRCA2 function through its interacting partners. Cell Cycle 2016; 14:3389-95. [PMID: 26566862 DOI: 10.1080/15384101.2015.1093702] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The role of the tumor suppressor BRCA2 has been shaped over 2 decades thanks to the discovery of its protein and nucleic acid partners, biochemical and structural studies of the protein, and the functional evaluation of germline variants identified in breast cancer patients. Yet, the pathogenic and functional effect of many germline mutations in BRCA2 remains undetermined, and the heterogeneity of BRCA2-associated tumors challenges the identification of causative variants that drive tumorigenesis. In this review, we propose an overview of the established and emerging interacting partners and functional pathways attributed to BRCA2, and we speculate on how variants altering these functions may contribute to cancer susceptibility.
Collapse
Affiliation(s)
- Juan S Martinez
- a Institut Curie; Centre de Recherche ; Orsay , France.,b CNRS UMR3348; Genotoxic Stress and Cancer; Centre Universitaire ; Orsay , France
| | - Céline Baldeyron
- a Institut Curie; Centre de Recherche ; Orsay , France.,b CNRS UMR3348; Genotoxic Stress and Cancer; Centre Universitaire ; Orsay , France
| | - Aura Carreira
- a Institut Curie; Centre de Recherche ; Orsay , France.,b CNRS UMR3348; Genotoxic Stress and Cancer; Centre Universitaire ; Orsay , France
| |
Collapse
|
27
|
Thirthagiri E, Klarmann KD, Shukla AK, Southon E, Biswas K, Martin BK, North SL, Magidson V, Burkett S, Haines DC, Noer K, Matthai R, Tessarollo L, Loncarek J, Keller JR, Sharan SK. BRCA2 minor transcript lacking exons 4-7 supports viability in mice and may account for survival of humans with a pathogenic biallelic mutation. Hum Mol Genet 2016; 25:1934-1945. [PMID: 26920070 DOI: 10.1093/hmg/ddw066] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Accepted: 02/22/2016] [Indexed: 11/14/2022] Open
Abstract
The breast cancer gene, BRCA2, is essential for viability, yet patients with Fanconi anemia-D1 subtype are born alive with biallelic mutations in this gene. The hypomorphic nature of the mutations is believed to support viability, but this is not always apparent. One such mutation is IVS7+2T>G, which causes premature protein truncation due to skipping of exon 7. We previously identified a transcript lacking exons 4-7, which restores the open-reading frame, encodes a DNA repair proficient protein and is expressed in IVS7+2T>G carriers. However, because the exons 4-7 encoded region contains several residues required for normal cell-cycle regulation and cytokinesis, this transcript's ability to support viability can be argued. To address this, we generated a Brca2 knock-in mouse model lacking exons 4-7 and demonstrated that these exons are dispensable for viability as well as tumor-free survival. This study provides the first in vivo evidence of the functional significance of a minor transcript of BRCA2 that can play a major role in the survival of humans who are homozygous for a clearly pathogenic mutation. Our results highlight the importance of assessing protein function restoration by premature truncating codon bypass by alternative splicing when evaluating the functional significance of variants such as nonsense and frame-shift mutations that are assumed to be clearly pathogenic. Our findings will impact not only the assessment of variants that map to this region, but also influence counseling paradigms and treatment options for such mutation carriers.
Collapse
Affiliation(s)
| | - Kimberly D Klarmann
- Mouse Cancer Genetics Program, Center for Cancer Research, Basic Sciences Program
| | | | - Eileen Southon
- Mouse Cancer Genetics Program, Center for Cancer Research, Basic Sciences Program
| | - Kajal Biswas
- Mouse Cancer Genetics Program, Center for Cancer Research
| | - Betty K Martin
- Mouse Cancer Genetics Program, Center for Cancer Research, Basic Sciences Program
| | | | | | - Sandra Burkett
- Mouse Cancer Genetics Program, Center for Cancer Research
| | - Diana C Haines
- Pathology/Histotechnology Laboratory, Leidos Biomedical Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA
| | - Kathleen Noer
- Frederick CCR Flow Cytometry Core Cancer and Inflammation Program, National Cancer Institute, Frederick, MD 21702, USA
| | - Roberta Matthai
- Frederick CCR Flow Cytometry Core Cancer and Inflammation Program, National Cancer Institute, Frederick, MD 21702, USA
| | | | | | - Jonathan R Keller
- Mouse Cancer Genetics Program, Center for Cancer Research, Basic Sciences Program,
| | - Shyam K Sharan
- Mouse Cancer Genetics Program, Center for Cancer Research,
| |
Collapse
|
28
|
Newell-Litwa KA, Horwitz R, Lamers ML. Non-muscle myosin II in disease: mechanisms and therapeutic opportunities. Dis Model Mech 2015; 8:1495-515. [PMID: 26542704 PMCID: PMC4728321 DOI: 10.1242/dmm.022103] [Citation(s) in RCA: 94] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The actin motor protein non-muscle myosin II (NMII) acts as a master regulator of cell morphology, with a role in several essential cellular processes, including cell migration and post-synaptic dendritic spine plasticity in neurons. NMII also generates forces that alter biochemical signaling, by driving changes in interactions between actin-associated proteins that can ultimately regulate gene transcription. In addition to its roles in normal cellular physiology, NMII has recently emerged as a critical regulator of diverse, genetically complex diseases, including neuronal disorders, cancers and vascular disease. In the context of these disorders, NMII regulatory pathways can be directly mutated or indirectly altered by disease-causing mutations. NMII regulatory pathway genes are also increasingly found in disease-associated copy-number variants, particularly in neuronal disorders such as autism and schizophrenia. Furthermore, manipulation of NMII-mediated contractility regulates stem cell pluripotency and differentiation, thus highlighting the key role of NMII-based pharmaceuticals in the clinical success of stem cell therapies. In this Review, we discuss the emerging role of NMII activity and its regulation by kinases and microRNAs in the pathogenesis and prognosis of a diverse range of diseases, including neuronal disorders, cancer and vascular disease. We also address promising clinical applications and limitations of NMII-based inhibitors in the treatment of these diseases and the development of stem-cell-based therapies.
Collapse
Affiliation(s)
- Karen A Newell-Litwa
- Department of Cell Biology, University of Virginia, Charlottesville, VA 22908, USA
| | - Rick Horwitz
- Department of Cell Biology, University of Virginia, Charlottesville, VA 22908, USA
| | - Marcelo L Lamers
- Department of Morphological Sciences, Institute of Basic Health Science, Federal University of Rio Grande do Sul, Porto Alegre, Rio Grande do Sul 90610-010, Brazil
| |
Collapse
|
29
|
Ghosh S, Bhunia AK, Paun BC, Gilbert SF, Dhru U, Patel K, Kern SE. Genome annotation by shotgun inactivation of a native gene in hemizygous cells: application to BRCA2 with implication of hypomorphic variants. Hum Mutat 2015; 36:260-9. [PMID: 25451944 DOI: 10.1002/humu.22736] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2014] [Accepted: 11/19/2014] [Indexed: 12/30/2022]
Abstract
The greatest interpretive challenge of modern medicine may be to functionally annotate the vast variation of human genomes. Demonstrating a proposed approach, we created a library of BRCA2 exon 27 shotgun-mutant plasmids including solitary and multiplex mutations to generate human knockin clones using homologous recombination. This 55-mutation, 13-clone syngeneic variance library (SyVaL) comprised severely affected clones having early-stop nonsense mutations, functionally hypomorphic clones having multiple missense mutations emphasizing the potential to identify and assess hypomorphic mutations in novel proteomic and epidemiologic studies, and neutral clones having multiple missense mutations. Efficient coverage of nonessential amino acids was provided by mutation multiplexing. Severe mutations were distinguished from hypomorphic or neutral changes by chemosensitivity assays (hypersensitivity to mitomycin C and acetaldehyde), by analysis of RAD51 focus formation, and by mitotic multipolarity. A multiplex unbiased approach of generating all-human SyVaLs in medically important genes, with random mutations in native genes, would provide databases of variants that could be functionally annotated without concerns arising from exogenous cDNA constructs or interspecies interactions, as a basis for subsequent proteomic domain mapping or clinical calibration if desired. Such gene-irrelevant approaches could be scaled up for multiple genes of clinical interest, providing distributable cellular libraries linked to public-shared functional databases.
Collapse
Affiliation(s)
- Soma Ghosh
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland, 21287
| | | | | | | | | | | | | |
Collapse
|
30
|
Differential Contributions of Nonmuscle Myosin II Isoforms and Functional Domains to Stress Fiber Mechanics. Sci Rep 2015; 5:13736. [PMID: 26336830 PMCID: PMC4559901 DOI: 10.1038/srep13736] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Accepted: 08/04/2015] [Indexed: 01/14/2023] Open
Abstract
While is widely acknowledged that nonmuscle myosin II (NMMII) enables stress fibers (SFs) to generate traction forces against the extracellular matrix, little is known about how specific NMMII isoforms and functional domains contribute to SF mechanics. Here we combine biophotonic and genetic approaches to address these open questions. First, we suppress the NMMII isoforms MIIA and MIIB and apply femtosecond laser nanosurgery to ablate and investigate the viscoelastic retraction of individual SFs. SF retraction dynamics associated with MIIA and MIIB suppression qualitatively phenocopy our earlier measurements in the setting of Rho kinase (ROCK) and myosin light chain kinase (MLCK) inhibition, respectively. Furthermore, fluorescence imaging and photobleaching recovery reveal that MIIA and MIIB are enriched in and more stably localize to ROCK- and MLCK-controlled central and peripheral SFs, respectively. Additional domain-mapping studies surprisingly reveal that deletion of the head domain speeds SF retraction, which we ascribe to reduced drag from actomyosin crosslinking and frictional losses. We propose a model in which ROCK/MIIA and MLCK/MIIB functionally regulate common pools of SFs, with MIIA crosslinking and motor functions jointly contributing to SF retraction dynamics and cellular traction forces.
Collapse
|
31
|
Kassianidou E, Kumar S. A biomechanical perspective on stress fiber structure and function. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2015; 1853:3065-74. [PMID: 25896524 DOI: 10.1016/j.bbamcr.2015.04.006] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/06/2015] [Revised: 04/05/2015] [Accepted: 04/08/2015] [Indexed: 01/11/2023]
Abstract
Stress fibers are actomyosin-based bundles whose structural and contractile properties underlie numerous cellular processes including adhesion, motility and mechanosensing. Recent advances in high-resolution live-cell imaging and single-cell force measurement have dramatically sharpened our understanding of the assembly, connectivity, and evolution of various specialized stress fiber subpopulations. This in turn has motivated interest in understanding how individual stress fibers generate tension and support cellular structure and force generation. In this review, we discuss approaches for measuring the mechanical properties of single stress fibers. We begin by discussing studies conducted in cell-free settings, including strategies based on isolation of intact stress fibers and reconstitution of stress fiber-like structures from purified components. We then discuss measurements obtained in living cells based both on inference of stress fiber properties from whole-cell mechanical measurements (e.g., atomic force microscopy) and on direct interrogation of single stress fibers (e.g., subcellular laser nanosurgery). We conclude by reviewing various mathematical models of stress fiber function that have been developed based on these experimental measurements. An important future challenge in this area will be the integration of these sophisticated biophysical measurements with the field's increasingly detailed molecular understanding of stress fiber assembly, dynamics, and signal transduction. This article is part of a Special Issue entitled: Mechanobiology.
Collapse
Affiliation(s)
- Elena Kassianidou
- Department of Bioengineering, University of California, Berkeley, United States
| | - Sanjay Kumar
- Department of Bioengineering, University of California, Berkeley, United States.
| |
Collapse
|
32
|
Ma X, Adelstein RS. The role of vertebrate nonmuscle Myosin II in development and human disease. BIOARCHITECTURE 2014; 4:88-102. [PMID: 25098841 DOI: 10.4161/bioa.29766] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Three different genes each located on a different chromosome encode the heavy chains of nonmuscle myosin II in humans and mice. This review explores the functional consequences of the presence of three isoforms during embryonic development and beyond. The roles of the various isoforms in cell division, cell-cell adhesion, blood vessel formation and neuronal cell migration are addressed in animal models and at the cellular level. Particular emphasis is placed on the role of nonmuscle myosin II during cardiac and brain development, and during closure of the neural tube and body wall. Questions addressed include the consequences on organ development, of lowering or ablating a particular isoform as well as the effect of substituting one isoform for another, all in vivo. Finally the roles of the three isoforms in human diseases such as cancer as well as in syndromes affecting a variety of organs in humans are reviewed.
Collapse
Affiliation(s)
- Xuefei Ma
- Laboratory of Molecular Cardiology; National Heart, Lung, and Blood Institute; National Institutes of Health; Bethesda, MD USA
| | - Robert S Adelstein
- Laboratory of Molecular Cardiology; National Heart, Lung, and Blood Institute; National Institutes of Health; Bethesda, MD USA
| |
Collapse
|