1
|
Alves Domingos H, Green M, Ouzounidis VR, Finlayson C, Prevo B, Cheerambathur DK. The kinetochore protein KNL-1 regulates the actin cytoskeleton to control dendrite branching. J Cell Biol 2025; 224:e202311147. [PMID: 39625434 PMCID: PMC11613958 DOI: 10.1083/jcb.202311147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 07/23/2024] [Accepted: 11/14/2024] [Indexed: 12/11/2024] Open
Abstract
The function of the nervous system is intimately tied to its complex and highly interconnected architecture. Precise control of dendritic branching in individual neurons is central to building the complex structure of the nervous system. Here, we show that the kinetochore protein KNL-1 and its associated KMN (Knl1/Mis12/Ndc80 complex) network partners, typically known for their role in chromosome-microtubule coupling during mitosis, control dendrite branching in the Caenorhabditis elegans mechanosensory PVD neuron. KNL-1 restrains excess dendritic branching and promotes contact-dependent repulsion events, ensuring robust sensory behavior and preventing premature neurodegeneration. Unexpectedly, KNL-1 loss resulted in significant alterations of the actin cytoskeleton alongside changes in microtubule dynamics within dendrites. We show that KNL-1 modulates F-actin dynamics to generate proper dendrite architecture and that its N-terminus can initiate F-actin assembly. These findings reveal that the postmitotic neuronal KMN network acts to shape the developing nervous system by regulating the actin cytoskeleton and provide new insight into the mechanisms controlling dendrite architecture.
Collapse
Affiliation(s)
- Henrique Alves Domingos
- Institute of Cell Biology, School of Biological Sciences, The University of Edinburgh, Edinburgh, UK
| | - Mattie Green
- Institute of Cell Biology, School of Biological Sciences, The University of Edinburgh, Edinburgh, UK
| | - Vasileios R. Ouzounidis
- Institute of Cell Biology, School of Biological Sciences, The University of Edinburgh, Edinburgh, UK
| | - Cameron Finlayson
- Institute of Cell Biology, School of Biological Sciences, The University of Edinburgh, Edinburgh, UK
| | - Bram Prevo
- Institute of Cell Biology, School of Biological Sciences, The University of Edinburgh, Edinburgh, UK
| | - Dhanya K. Cheerambathur
- Institute of Cell Biology, School of Biological Sciences, The University of Edinburgh, Edinburgh, UK
| |
Collapse
|
2
|
Knight A, Sugin S, Jurisicova A. Searching for the 'X' factor: investigating the genetics of primary ovarian insufficiency. J Ovarian Res 2024; 17:238. [PMID: 39609914 PMCID: PMC11603650 DOI: 10.1186/s13048-024-01555-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 11/10/2024] [Indexed: 11/30/2024] Open
Abstract
Primary ovarian insufficiency (POI) is the cessation of ovarian function before the age of 40. The causes of POI are heterogeneous, but substantial evidence exists to support a genetic basis of POI, particularly in the critical involvement of genes on the X chromosome. Recent studies have revealed novel candidate genes through the identification of copy number variations associated with POI. This review summarizes the genes located on the X chromosome with variants shown to be associated with POI in humans and/or in mice. Additionally, we present evidence to support the potential involvement of these candidate genes in the etiology of POI. We conducted a literature search in PubMed to identify case studies and screenings for the genetic causes of POI. We then performed systematic searches for the proposed candidate genes to investigate their potential reproductive roles. Of the X-linked candidate genes investigated, 10 were found to have variants associated with cases of POI in humans. An additional 10 genes were found to play a supportive role in POI. Other genes were not implicated in any cases of POI but were associated with various roles in reproduction. In the majority of cases where variants were identified through whole-exome sequencing, rather than targeted screening of candidate genes, more than one genetic variant was identified. Overall, this review supports past findings that the X chromosome plays a critical role in ovarian function, as demonstrated by a link between POI and various disruptions to genes on the X chromosome. Current genetic screening for POI, which includes only FMR1, is inadequate to capture the majority of cases with a genetic origin. An expanded genetic testing may improve health outcomes for individuals with POI as it could lead to better early interventions and education about these health risks.
Collapse
Affiliation(s)
- Anya Knight
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, Canada
| | - Sara Sugin
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, Canada
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, 25 Orde Street, Room 6-1016-1, Toronto, ON, M5T 3H7, Canada
| | - Andrea Jurisicova
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, Canada.
- Department of Obstetrics and Gynecology, Temerty Faculty of Medicine, University of Toronto, Toronto, Canada.
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, 25 Orde Street, Room 6-1016-1, Toronto, ON, M5T 3H7, Canada.
| |
Collapse
|
3
|
Wei J, Wen Q, Zhan S, Cao J, Jiang Y, Lian J, Mai Y, Qiu M, Liu Y, Chen P, Lin Q, Wei X, Wei Y, Huang Q, Zhang R, He S, Yuan G, Wei Q, Zhou Z, Yu H. Functional genetic variants of the disulfidptosis-related INF2 gene predict survival of hepatitis B virus-related hepatocellular carcinoma. Carcinogenesis 2024; 45:199-209. [PMID: 38270181 DOI: 10.1093/carcin/bgae003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 12/18/2023] [Accepted: 01/23/2024] [Indexed: 01/26/2024] Open
Abstract
Disulfidptosis is a novel form of programmed cell death involved in migration and invasion of cancer cells, but few studies investigated the roles of genetic variants in disulfidptosis-related genes in survival of patients with hepatitis B virus (HBV)-related hepatocellular carcinoma (HCC). We used Cox proportional hazards regression analyses, Kaplan-Meier curves and receiver operating characteristic curves to assess effects of genetic variants in 14 disulfidptosis-related genes on overall survival of 866 HBV-HCC patients. The Bayesian false discovery probability was used for multiple testing corrections. We also investigated biological mechanisms of the significant variants through expression quantitative trait loci analyses using the data from publicly available databases, luciferase reporter assays and differential expression analyses. As a result, we identified two independently functional single nucleotide polymorphisms (SNPs) (INF2 rs4072285 G > A and INF2 rs4444271 A > T) that predicted overall survival of HBV-HCC patients, with adjusted hazard ratios of 1.60 (95% CI = 1.22-2.11, P = 0.001) and 1.50 (95% CI = 1.80-1.90, P < 0.001), respectively, after multiple testing correction. Luciferase reporter assays indicated that both INF2 rs4072285 A and INF2 rs4444271 T alleles increased INF2 mRNA expression levels (P < 0.001) that were also higher in HCC tumor tissues than in adjacent normal tissues (P < 0.001); such elevated INF2 expression levels were associated with a poorer survival of HBV-HCC patients (P < 0.001) in the TCGA database. In summary, this study supported that INF2 rs4072285 and INF2 rs4444271 may be novel biomarkers for survival of HBV-HCC patients.
Collapse
Affiliation(s)
- Junjie Wei
- Department of Experimental Research, Guangxi Medical University Cancer Hospital, Nanning 530021, China
- Department of Epidemiology and Health Statistics, School of Public Health, Guangxi Medical University, Nanning 530021, China
| | - Qiuping Wen
- Department of Experimental Research, Guangxi Medical University Cancer Hospital, Nanning 530021, China
- Key Cultivated Laboratory of Cancer Molecular Medicine of Guangxi Health Commission, Guangxi Medical University Cancer Hospital, Nanning 530021, China
| | - Shicheng Zhan
- Department of Epidemiology and Health Statistics, School of Public Health, Guangxi Medical University, Nanning 530021, China
| | - Ji Cao
- Department of Cancer Prevention and Control, Guangxi Medical University Cancer Hospital, Nanning 530021, China
| | - Yanji Jiang
- Department of Scientific Research, Guangxi Medical University Cancer Hospital, Nanning 530021, China
| | - Jiawei Lian
- Department of Epidemiology and Health Statistics, School of Public Health, Guangxi Medical University, Nanning 530021, China
| | - Yuejiao Mai
- Department of Epidemiology and Health Statistics, School of Public Health, Guangxi Medical University, Nanning 530021, China
| | - Moqin Qiu
- Department of Respiratory Oncology, Guangxi Medical University Cancer Hospital, Nanning 530021, China
| | - Yingchun Liu
- Department of Experimental Research, Guangxi Medical University Cancer Hospital, Nanning 530021, China
- Key Cultivated Laboratory of Cancer Molecular Medicine of Guangxi Health Commission, Guangxi Medical University Cancer Hospital, Nanning 530021, China
| | - Peiqin Chen
- Department of Experimental Research, Guangxi Medical University Cancer Hospital, Nanning 530021, China
| | - Qiuling Lin
- Department of Clinical Research, Guangxi Medical University Cancer Hospital, Nanning 530021, China
| | - Xiaoxia Wei
- Department of Clinical Research, Guangxi Medical University Cancer Hospital, Nanning 530021, China
| | - Yuying Wei
- Department of Experimental Research, Guangxi Medical University Cancer Hospital, Nanning 530021, China
| | - Qiongguang Huang
- Department of Epidemiology and Health Statistics, School of Public Health, Guangxi Medical University, Nanning 530021, China
| | - Ruoxin Zhang
- School of Public Health, Key Laboratory of Public Health Safety, Ministry of Education, Fudan University, Shanghai 200032, China
| | - Songqing He
- Division of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, China
- Guangxi Key Laboratory of Immunology and Metabolism for Liver Diseases, Nanning, Guangxi 530021, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Nanning 530021, China
| | - Guandou Yuan
- Division of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, China
- Guangxi Key Laboratory of Immunology and Metabolism for Liver Diseases, Nanning, Guangxi 530021, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Nanning 530021, China
| | - Qingyi Wei
- Duke Cancer Institute, Duke University Medical Center, 10 Bryn Searle Dr., Durham, NC, 27710, USA
- Department of Population Health Sciences, Duke University School of Medicine, Durham, NC 27710, USA
| | - Zihan Zhou
- Department of Cancer Prevention and Control, Guangxi Medical University Cancer Hospital, Nanning 530021, China
| | - Hongping Yu
- Department of Experimental Research, Guangxi Medical University Cancer Hospital, Nanning 530021, China
- Key Cultivated Laboratory of Cancer Molecular Medicine of Guangxi Health Commission, Guangxi Medical University Cancer Hospital, Nanning 530021, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Nanning 530021, China
| |
Collapse
|
4
|
Soh JEC, Shimizu A, Sato A, Ogita H. Novel cardiovascular protective effects of RhoA signaling and its therapeutic implications. Biochem Pharmacol 2023; 218:115899. [PMID: 37907138 DOI: 10.1016/j.bcp.2023.115899] [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: 09/19/2023] [Revised: 10/24/2023] [Accepted: 10/26/2023] [Indexed: 11/02/2023]
Abstract
Ras homolog gene family member A (RhoA) belongs to the Rho GTPase superfamily, which was first studied in cancers as one of the essential regulators controlling cellular function. RhoA has long attracted attention as a key molecule involved in cell signaling and gene transcription, through which it affects cellular processes. A series of studies have demonstrated that RhoA plays crucial roles under both physiological states and pathological conditions in cardiovascular diseases. RhoA has been identified as an important regulator in cardiac remodeling by regulating actin stress fiber dynamics and cytoskeleton formation. However, its underlying mechanisms remain poorly understood, preventing definitive conclusions being drawn about its protective role in the cardiovascular system. In this review, we outline the characteristics of RhoA and its related signaling molecules, and present an overview of RhoA classical function and the corresponding cellular responses of RhoA under physiological and pathological conditions. Overall, we provide an update on the novel signaling under RhoA in the cardiovascular system and its potential clinical and therapeutic targets in cardiovascular medicine.
Collapse
Affiliation(s)
- Joanne Ern Chi Soh
- Division of Molecular Medical Biochemistry, Department of Biochemistry and Molecular Biology, Shiga University of Medical Science, Otsu, Japan
| | - Akio Shimizu
- Division of Molecular Medical Biochemistry, Department of Biochemistry and Molecular Biology, Shiga University of Medical Science, Otsu, Japan
| | - Akira Sato
- Division of Molecular Medical Biochemistry, Department of Biochemistry and Molecular Biology, Shiga University of Medical Science, Otsu, Japan
| | - Hisakazu Ogita
- Division of Molecular Medical Biochemistry, Department of Biochemistry and Molecular Biology, Shiga University of Medical Science, Otsu, Japan.
| |
Collapse
|
5
|
Zhang B, Hu Q, Li Y, Xu C, Xie X, Liu P, Xu M, Gong S, Wu H. Diaphanous-related formin subfamily: Novel prognostic biomarkers and tumor microenvironment regulators for pancreatic adenocarcinoma. Front Mol Biosci 2022; 9:910950. [PMID: 36589226 PMCID: PMC9797685 DOI: 10.3389/fmolb.2022.910950] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 11/30/2022] [Indexed: 12/23/2022] Open
Abstract
The diaphanous-related formin subfamily includes diaphanous homolog 1 (DIAPH1), DIAPH2, and DIAPH3. DIAPHs play a role in the regulation of actin nucleation and polymerization and in microtubule stability. DIAPH3 also regulates the assembly and bipolarity of mitotic spindles. Accumulating evidence has shown that DIAPHs are anomalously regulated during malignancy. In this study, we reviewed The Cancer Genome Atlas database and found that DIAPHs are abundantly expressed in pancreatic adenocarcinoma (PAAD). Furthermore, we analyzed the gene alteration profiles, protein expression, prognosis, and immune reactivity of DIAPHs in PAAD using data from several well-established databases. In addition, we conducted gene set enrichment analysis to investigate the potential mechanisms underlying the roles of DIAPHs in the carcinogenesis of PAAD. Finally, we performed the experimental validation of DIAPHs expression in several pancreatic cancer cell lines and tissues of patients. This study demonstrated significant correlations between DIAPHs expression and clinical prognosis, oncogenic signature gene sets, T helper 2 cell infiltration, plasmacytoid dendritic cell infiltration, myeloid-derived suppressor cell infiltration, ImmunoScore, and immune checkpoints in PAAD. These data may provide important information regarding the role and mechanisms of DIAPHs in tumorigenesis and PAAD immunotherapy.
Collapse
Affiliation(s)
- Bixi Zhang
- Department of Pathology, Hunan Provincial People’s Hospital, Hunan Normal University, Changsha, China
| | - Qing Hu
- Department of Pathology, Hunan Provincial People’s Hospital, Hunan Normal University, Changsha, China
| | - Yanchun Li
- Department of Pathology, Hunan Provincial People’s Hospital, Hunan Normal University, Changsha, China
| | - Canxia Xu
- Department of Gastroenterology, Third Xiangya Hospital, Central South University, Changsha, China
| | - Xiaoran Xie
- Department of Gastroenterology, Third Xiangya Hospital, Central South University, Changsha, China
| | - Peng Liu
- Department of Gastroenterology, Third Xiangya Hospital, Central South University, Changsha, China
| | - Meihua Xu
- Department of Gastroenterology, Xiangya Hospital, Central South University, Changsha, China
| | - Siming Gong
- Institute of Anatomy, University of Leipzig, Leipzig, Germany
| | - Hao Wu
- Department of Gastroenterology, Third Xiangya Hospital, Central South University, Changsha, China,Center for Precision Medicine, University of Missouri School of Medicine, Columbia, MO, United States,*Correspondence: Hao Wu,
| |
Collapse
|
6
|
Fang X, Svitkina TM. Adenomatous polyposis coli (APC) in cell migration. Eur J Cell Biol 2022; 101:151228. [DOI: 10.1016/j.ejcb.2022.151228] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 04/15/2022] [Accepted: 04/20/2022] [Indexed: 12/22/2022] Open
|
7
|
Rouen A, Rogers E, Kerlan V, Delemer B, Catteau-Jonard S, Reznik Y, Gompel A, Cedrin I, Guedj AM, Grouthier V, Brue T, Pienkowski C, Bachelot A, Chantot-Bastaraud S, Rousseau A, Simon T, Kott E, Siffroi JP, Touraine P, Christin-Maitre S. Whole exome sequencing in a cohort of familial premature ovarian insufficiency cases reveals a broad array of pathogenic or likely pathogenic variants in 50% of families. Fertil Steril 2022; 117:843-853. [PMID: 35115167 DOI: 10.1016/j.fertnstert.2021.12.023] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 12/19/2021] [Accepted: 12/21/2021] [Indexed: 12/22/2022]
Abstract
OBJECTIVE To study the diagnostic yield, including variants in genes yet to be incriminated, of whole exome sequencing (WES) in familial cases of premature ovarian insufficiency (POI). DESIGN Cross-sectional study. SETTING Endocrinology and reproductive medicine teaching hospital departments. PATIENTS Familial POI cases were recruited as part of a nationwide multicentric cohort. A total of 36 index cases in 36 different families were studied. Fifty-two relatives were available, including 25 with POI and 27 affectedwho were nonaffected. Karyotype analysis, FMR1 screening, single nucleotide polymorphism array analysis, and WES were performed in all subjects. INTERVENTIONS None. MAIN OUTCOME MEASURES The primary outcome was a molecular etiology, as diagnosed by karyotype, FMR1 screening, single nucleotide polymorphism array, and WES. RESULTS A likely molecular etiology (pathogenic or likely pathogenic variant) was identified in 18 of 36 index cases (50% diagnostic yield). In 12 families, we found a pathogenic or likely pathogenic variant in a gene previously incriminated in POI, and in 6 families, we found a pathogenic or likely pathogenic variant in new candidate genes. Most of the variants identified were located in genes involved in cell division and meiosis (n = 11) or DNA repair (n = 4). CONCLUSIONS The genetic etiologic diagnosis in POI allows for genetic familial counseling, anticipated pregnancy planning, and ovarian tissue preservation or oocyte preservation. Identifying new genes may lead to future development of therapeutics in reproduction based on disrupted molecular pathways. CLINICAL TRIAL REGISTRATION NUMBER NCT 01177891.
Collapse
Affiliation(s)
- Alexandre Rouen
- Département de Génétique Médicale, Unité INSERM U933, Hôpital Armand-Trousseau, Assistance Publique-Hôpitaux de Paris, Paris, France.
| | - Eli Rogers
- Département de Génétique Médicale, Unité INSERM U933, Hôpital Armand-Trousseau, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Véronique Kerlan
- Service d'Endocrinologie, Centre Hospitalier Universitaire de Brest, Brest, France
| | - Brigitte Delemer
- Service d'Endocrinologie, Diabète, Nutrition, Centre Hospitalier Universitaire de Reims, Reims, France
| | | | - Yves Reznik
- Service d'Endocrinologie, Hôpital Caen, France
| | - Anne Gompel
- Université de Paris, Unité de Gynécologie Médicale, Hôpital Port-Royal, France
| | - Isabelle Cedrin
- Service de Médecine de la Reproduction, Hôpital Jean Verdier, France
| | | | | | - Thierry Brue
- Assistance Publique-Hôpitaux de Marseille, Department of Endocrinology, Hôpital de la Conception, Centre de Référence des Maladies Rares de l'Hypophyse, Marseille, France, and Aix-Marseille Université, Institut National de la Santé et de la Recherche Médicale, Marseille Medical Genetics, Institut Marseille Maladies Rares, Marseille, France
| | | | - Anne Bachelot
- Service d'Endocrinologie et Médecine de la Reproduction, Centre Constitutif des Maladies Endocriniennes Rares de la Croissance et du Développement, Centre Constitutif du Centre des Pathologies Gynécologiques Rares, Sorbonne Université, Hôpital de la Pitié-Salpétrière, Paris, France; Sorbonne Université, Paris, France
| | - Sandra Chantot-Bastaraud
- Département de Génétique Médicale, Unité INSERM U933, Hôpital Armand-Trousseau, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Alexandra Rousseau
- Unité de Recherche Clinique de l'Est Parisien, Hôpital Saint-Antoine, Assistance Publique - Hôpitaux de Paris, France
| | - Tabassome Simon
- Unité de Recherche Clinique de l'Est Parisien, Hôpital Saint-Antoine, Assistance Publique - Hôpitaux de Paris, France
| | - Esther Kott
- Département de Génétique Médicale, Unité INSERM U933, Hôpital Armand-Trousseau, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Jean-Pierre Siffroi
- Département de Génétique Médicale, Unité INSERM U933, Hôpital Armand-Trousseau, Assistance Publique-Hôpitaux de Paris, Paris, France; Sorbonne Université, Paris, France
| | - Philippe Touraine
- Service d'Endocrinologie et Médecine de la Reproduction, Centre Constitutif des Maladies Endocriniennes Rares de la Croissance et du Développement, Centre Constitutif du Centre des Pathologies Gynécologiques Rares, Sorbonne Université, Hôpital de la Pitié-Salpétrière, Paris, France; Sorbonne Université, Paris, France
| | - Sophie Christin-Maitre
- Département de Génétique Médicale, Unité INSERM U933, Hôpital Armand-Trousseau, Assistance Publique-Hôpitaux de Paris, Paris, France; Sorbonne Université, Paris, France; Service d'Endocrinologie, Diabétologie et Médecine de la Reproduction, Centre Constitutif des Maladies Endocriniennes Rares de la Croissance et du Développement, Sorbonne Université, Hôpital Saint-Antoine, Paris, France
| |
Collapse
|
8
|
Abstract
ABSTRACT The Aurora kinases (AURKA and AURKB) have attracted attention as therapeutic targets in head and neck squamous cell carcinomas. Aurora kinases were first defined as regulators of mitosis that localization to the centrosome (AURKA) and centromere (AURKB), governing formation of the mitotic spindle, chromatin condensation, activation of the core mitotic kinase CDK1, alignment of chromosomes at metaphase, and other processes. Subsequently, additional roles for Aurora kinases have been defined in other phases of cell cycle, including regulation of ciliary disassembly and DNA replication. In cancer, elevated expression and activity of Aurora kinases result in enhanced or neomorphic locations and functions that promote aggressive disease, including promotion of MYC expression, oncogenic signaling, stem cell identity, epithelial-mesenchymal transition, and drug resistance. Numerous Aurora-targeted inhibitors have been developed and are being assessed in preclinical and clinical trials, with the goal of improving head and neck squamous cell carcinoma treatment.
Collapse
|
9
|
Zhang L, Smertenko T, Fahy D, Koteyeva N, Moroz N, Kuchařová A, Novák D, Manoilov E, Smertenko P, Galva C, Šamaj J, Kostyukova AS, Sedbrook JC, Smertenko A. Analysis of formin functions during cytokinesis using specific inhibitor SMIFH2. PLANT PHYSIOLOGY 2021; 186:945-963. [PMID: 33620500 PMCID: PMC8195507 DOI: 10.1093/plphys/kiab085] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 02/03/2021] [Indexed: 05/10/2023]
Abstract
The phragmoplast separates daughter cells during cytokinesis by constructing the cell plate, which depends on interaction between cytoskeleton and membrane compartments. Proteins responsible for these interactions remain unknown, but formins can link cytoskeleton with membranes and several members of formin protein family localize to the cell plate. Progress in functional characterization of formins in cytokinesis is hindered by functional redundancies within the large formin gene family. We addressed this limitation by employing Small Molecular Inhibitor of Formin Homology 2 (SMIFH2), a small-molecule inhibitor of formins. Treatment of tobacco (Nicotiana tabacum) tissue culture cells with SMIFH2 perturbed localization of actin at the cell plate; slowed down both microtubule polymerization and phragmoplast expansion; diminished association of dynamin-related proteins with the cell plate independently of actin and microtubules; and caused cell plate swelling. Another impact of SMIFH2 was shortening of the END BINDING1b (EB1b) and EB1c comets on the growing microtubule plus ends in N. tabacum tissue culture cells and Arabidopsis thaliana cotyledon epidermis cells. The shape of the EB1 comets in the SMIFH2-treated cells resembled that of the knockdown mutant of plant Xenopus Microtubule-Associated protein of 215 kDa (XMAP215) homolog MICROTUBULE ORGANIZATION 1/GEMINI 1 (MOR1/GEM1). This outcome suggests that formins promote elongation of tubulin flares on the growing plus ends. Formins AtFH1 (A. thaliana Formin Homology 1) and AtFH8 can also interact with EB1. Besides cytokinesis, formins function in the mitotic spindle assembly and metaphase to anaphase transition. Our data suggest that during cytokinesis formins function in: (1) promoting microtubule polymerization; (2) nucleating F-actin at the cell plate; (3) retaining dynamin-related proteins at the cell plate; and (4) remodeling of the cell plate membrane.
Collapse
Affiliation(s)
- Laining Zhang
- Institute of Biological Chemistry, Washington State University, Pullman, Washington, USA
| | - Tetyana Smertenko
- Institute of Biological Chemistry, Washington State University, Pullman, Washington, USA
| | - Deirdre Fahy
- Institute of Biological Chemistry, Washington State University, Pullman, Washington, USA
| | - Nuria Koteyeva
- Laboratory of Anatomy and Morphology, Komarov Botanical Institute of Russian Academy of Sciences, St. Petersburg 197376, Russia
| | - Natalia Moroz
- The Gene and Linda Voiland School of Chemical Engineering and Bioengineering, Washington State University, Pullman, Washington, USA
| | - Anna Kuchařová
- Centre of the Region Haná for Biotechnological and Agricultural Research, Faculty of Science, Palacký University, 783 71 Olomouc, Czech Republic
| | - Dominik Novák
- Centre of the Region Haná for Biotechnological and Agricultural Research, Faculty of Science, Palacký University, 783 71 Olomouc, Czech Republic
| | - Eduard Manoilov
- V. Lashkaryov Institute of Semiconductor Physics, NAS of Ukraine, Kyiv, Ukraine
| | - Petro Smertenko
- V. Lashkaryov Institute of Semiconductor Physics, NAS of Ukraine, Kyiv, Ukraine
| | - Charitha Galva
- School of Biological Sciences, Illinois State University, Normal, Illinois, USA
| | - Jozef Šamaj
- Centre of the Region Haná for Biotechnological and Agricultural Research, Faculty of Science, Palacký University, 783 71 Olomouc, Czech Republic
| | - Alla S. Kostyukova
- The Gene and Linda Voiland School of Chemical Engineering and Bioengineering, Washington State University, Pullman, Washington, USA
| | - John C. Sedbrook
- School of Biological Sciences, Illinois State University, Normal, Illinois, USA
| | - Andrei Smertenko
- Institute of Biological Chemistry, Washington State University, Pullman, Washington, USA
| |
Collapse
|
10
|
Aurora B kinase: a potential drug target for cancer therapy. J Cancer Res Clin Oncol 2021; 147:2187-2198. [PMID: 34047821 DOI: 10.1007/s00432-021-03669-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Accepted: 05/18/2021] [Indexed: 12/22/2022]
Abstract
BACKGROUND Ensuring genetic integrity is essential during the cell cycle to avoid aneuploidy, one of the underlying causes of malignancies. Aurora kinases are serine/threonine kinase that play a vital role in maintaining the genomic integrity of the cells. There are three forms of aurora kinases in the mammalian cells, which are highly conserved and act together with several other proteins to control chromosome alignment and its equal distribution to daughter cells in mitosis and meiosis. METHODS We provide here a detailed analysis of Aurora B kinase (ABK) in terms of its expression, structure, function, disease association and potential therapeutic implications. RESULTS ABK plays an instrumental in mitotic entry, chromosome condensation, spindle assembly, cytokinesis, and abscission. Small-molecule inhibitors of ABK are designed and synthesized to control cancer progression. A detailed understanding of ABK pathophysiology in different cancers is of great significance in designing and developing effective therapeutic strategies. CONCLUSION In this review, we have discussed the physiological significance of ABK followed by its role in cancer progression. We further highlighted available small-molecule inhibitors to control the tumor proliferation and their mechanistic insights.
Collapse
|
11
|
Loss of DIAPH3, a Formin Family Protein, Leads to Cytokinetic Failure Only under High Temperature Conditions in Mouse FM3A Cells. Int J Mol Sci 2020; 21:ijms21228493. [PMID: 33187357 PMCID: PMC7696919 DOI: 10.3390/ijms21228493] [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: 09/28/2020] [Revised: 11/07/2020] [Accepted: 11/09/2020] [Indexed: 11/17/2022] Open
Abstract
Cell division is essential for the maintenance of life and involves chromosome segregation and subsequent cytokinesis. The processes are tightly regulated at both the spatial and temporal level by various genes, and failures in this regulation are associated with oncogenesis. Here, we investigated the gene responsible for defects in cell division by using murine temperature-sensitive (ts) mutant strains, tsFT101 and tsFT50 cells. The ts mutants normally grow in a low temperature environment (32 °C) but fail to divide in a high temperature environment (39 °C). Exome sequencing and over-expression analyses identified Diaph3, a member of the formin family, as the cause of the temperature sensitivity observed in tsFT101 and tsFT50 cells. Interestingly, Diaph3 knockout cells showed abnormality in cytokinesis at 39 °C, and the phenotype was rescued by re-expression of Diaph3 WT, but not Diaph1 and Diaph2, other members of the formin family. Furthermore, Diaph3 knockout cells cultured at 39 °C showed a significant increase in the level of acetylated α-tubulin, an index of stabilized microtubules, and the level was reduced by Diaph3 expression. These results suggest that Diaph3 is required for cytokinesis only under high temperature conditions. Therefore, our study provides a new insight into the mechanisms by which regulatory factors of cell division function in a temperature-dependent manner.
Collapse
|
12
|
Involvement of Actin and Actin-Binding Proteins in Carcinogenesis. Cells 2020; 9:cells9102245. [PMID: 33036298 PMCID: PMC7600575 DOI: 10.3390/cells9102245] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 09/18/2020] [Accepted: 10/05/2020] [Indexed: 12/13/2022] Open
Abstract
The actin cytoskeleton plays a crucial role in many cellular processes while its reorganization is important in maintaining cell homeostasis. However, in the case of cancer cells, actin and ABPs (actin-binding proteins) are involved in all stages of carcinogenesis. Literature has reported that ABPs such as SATB1 (special AT-rich binding protein 1), WASP (Wiskott-Aldrich syndrome protein), nesprin, and villin take part in the initial step of carcinogenesis by regulating oncogene expression. Additionally, changes in actin localization promote cell proliferation by inhibiting apoptosis (SATB1). In turn, migration and invasion of cancer cells are based on the formation of actin-rich protrusions (Arp2/3 complex, filamin A, fascin, α-actinin, and cofilin). Importantly, more and more scientists suggest that microfilaments together with the associated proteins mediate tumor vascularization. Hence, the presented article aims to summarize literature reports in the context of the potential role of actin and ABPs in all steps of carcinogenesis.
Collapse
|
13
|
Bürgi J, Abrami L, Castanon I, Abriata LA, Kunz B, Yan SE, Lera M, Unger S, Superti-Furga A, Peraro MD, Gaitan MG, van der Goot FG. Ligand Binding to the Collagen VI Receptor Triggers a Talin-to-RhoA Switch that Regulates Receptor Endocytosis. Dev Cell 2020; 53:418-430.e4. [PMID: 32428455 DOI: 10.1016/j.devcel.2020.04.015] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Revised: 01/23/2020] [Accepted: 04/21/2020] [Indexed: 11/23/2022]
Abstract
Capillary morphogenesis gene 2 (CMG2/ANTXR2) is a cell surface receptor for both collagen VI and anthrax toxin. Biallelic loss-of-function mutations in CMG2 lead to a severe condition, hyaline fibromatosis syndrome (HFS). We have here dissected a network of dynamic interactions between CMG2 and various actin interactors and regulators, describing a different behavior from other extracellular matrix receptors. CMG2 binds talin, and thereby the actin cytoskeleton, only in its ligand-free state. Extracellular ligand binding leads to src-dependent talin release and recruitment of the actin cytoskeleton regulator RhoA and its effectors. These sequential interactions of CMG2 are necessary for the control of oriented cell division during fish development. Finally, we demonstrate that effective switching between talin and RhoA binding is required for the intracellular degradation of collagen VI in human fibroblasts, which explains why HFS mutations in the cytoskeleton-binding domain lead to dysregulation of extracellular matrix homeostasis.
Collapse
Affiliation(s)
- Jérôme Bürgi
- Faculty of Life Sciences, Global Health Institute, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland; EMBL Hamburg DESY, 22607 Hamburg, Germany
| | - Laurence Abrami
- Faculty of Life Sciences, Global Health Institute, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Irinka Castanon
- Departments of Biochemistry and of Molecular Biology, Sciences II, 30 Quai Ernest-Ansermet, 1211 Geneva, Switzerland
| | - Luciano Andres Abriata
- Faculty of Life Sciences, Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Beatrice Kunz
- Faculty of Life Sciences, Global Health Institute, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Shixu Emili Yan
- Faculty of Life Sciences, Global Health Institute, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Manuel Lera
- Departments of Biochemistry and of Molecular Biology, Sciences II, 30 Quai Ernest-Ansermet, 1211 Geneva, Switzerland
| | - Sheila Unger
- Division of Genetic Medicine, Centre Hospitalier Universitaire Vaudois, University of Lausanne, 1011 Lausanne, Switzerland
| | - Andrea Superti-Furga
- Division of Genetic Medicine, Centre Hospitalier Universitaire Vaudois, University of Lausanne, 1011 Lausanne, Switzerland
| | - Matteo Dal Peraro
- Faculty of Life Sciences, Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Marcos Gonzalez Gaitan
- Departments of Biochemistry and of Molecular Biology, Sciences II, 30 Quai Ernest-Ansermet, 1211 Geneva, Switzerland
| | - Francoise Gisou van der Goot
- Faculty of Life Sciences, Global Health Institute, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland.
| |
Collapse
|
14
|
The therapeutic potential of Aurora kinases targeting in glioblastoma: from preclinical research to translational oncology. J Mol Med (Berl) 2020; 98:495-512. [PMID: 32219470 DOI: 10.1007/s00109-020-01895-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 02/28/2020] [Accepted: 03/03/2020] [Indexed: 12/21/2022]
Abstract
Glioblastoma is the most common aggressive primary brain tumor. Standard care includes maximal safe surgical resection, radiation, and chemotherapy with temozolomide. However, the impact of this therapeutic approach on patient survival is disappointing and poor outcomes are frequently observed. Therefore, new therapeutic targets are needed to treat this potentially deadly tumor. Aurora kinases are one of today's most sought-after classes of therapeutic targets to glioblastoma therapy. They are a family of proteins composed of three members: Aurora-A, Aurora-B, and Aurora-C that play different roles in the cell division through regulation of chromosome segregation. Deregulation of these genes has been reported in glioblastoma and a progressive number of studies have shown that inhibition of these proteins could be a promising strategy for the treatment of this tumor. This review discusses the preclinical and early clinical findings on the potential use of the Aurora kinases as new targets for the treatment of glioblastoma. KEY MESSAGES: GBM is a very aggressive tumor with limited therapeutic options. Aurora kinases are a family of serine/threonine kinases implicated in GBM pathology. Aurora kinases are critical for glioblastoma cell growth, apoptosis, and chemoresistance. Inhibition of Aurora kinases has a synergistic or sensitizing effect with chemotherapy drugs, radiotherapy, or with other targeted molecules in GBM. Several Aurora kinase inhibitors are currently in clinical trials.
Collapse
|
15
|
Chen A, Arora PD, Lai CC, Copeland JW, Moraes TF, McCulloch CA, Lavoie BD, Wilde A. The scaffold-protein IQGAP1 enhances and spatially restricts the actin-nucleating activity of Diaphanous-related formin 1 (DIAPH1). J Biol Chem 2020; 295:3134-3147. [PMID: 32005666 DOI: 10.1074/jbc.ra119.010476] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 01/24/2020] [Indexed: 11/06/2022] Open
Abstract
The actin cytoskeleton is a dynamic array of filaments that undergoes rapid remodeling to drive many cellular processes. An essential feature of filament remodeling is the spatio-temporal regulation of actin filament nucleation. One family of actin filament nucleators, the Diaphanous-related formins, is activated by the binding of small G-proteins such as RhoA. However, RhoA only partially activates formins, suggesting that additional factors are required to fully activate the formin. Here we identify one such factor, IQ motif containing GTPase activating protein-1 (IQGAP1), which enhances RhoA-mediated activation of the Diaphanous-related formin (DIAPH1) and targets DIAPH1 to the plasma membrane. We find that the inhibitory intramolecular interaction within DIAPH1 is disrupted by the sequential binding of RhoA and IQGAP1. Binding of RhoA and IQGAP1 robustly stimulates DIAPH1-mediated actin filament nucleation in vitro In contrast, the actin capping protein Flightless-I, in conjunction with RhoA, only weakly stimulates DIAPH1 activity. IQGAP1, but not Flightless-I, is required to recruit DIAPH1 to the plasma membrane where actin filaments are generated. These results indicate that IQGAP1 enhances RhoA-mediated activation of DIAPH1 in vivo Collectively these data support a model where the combined action of RhoA and an enhancer ensures the spatio-temporal regulation of actin nucleation to stimulate robust and localized actin filament production in vivo.
Collapse
Affiliation(s)
- Anan Chen
- Department of Biochemistry, University of Toronto, Toronto, Ontario M5G 1M1, Canada
| | - Pam D Arora
- Faculty of Dentistry, University of Toronto, Toronto, Ontario M5G 1M1, Canada
| | - Christine C Lai
- Department of Biochemistry, University of Toronto, Toronto, Ontario M5G 1M1, Canada
| | - John W Copeland
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Trevor F Moraes
- Department of Biochemistry, University of Toronto, Toronto, Ontario M5G 1M1, Canada
| | | | - Brigitte D Lavoie
- Department Molecular Genetics, University of Toronto, Toronto, Ontario M5G 1M1, Canada
| | - Andrew Wilde
- Department of Biochemistry, University of Toronto, Toronto, Ontario M5G 1M1, Canada; Department Molecular Genetics, University of Toronto, Toronto, Ontario M5G 1M1, Canada.
| |
Collapse
|
16
|
Klein O, Krier-Burris RA, Lazki-Hagenbach P, Gorzalczany Y, Mei Y, Ji P, Bochner BS, Sagi-Eisenberg R. Mammalian diaphanous-related formin 1 (mDia1) coordinates mast cell migration and secretion through its actin-nucleating activity. J Allergy Clin Immunol 2019; 144:1074-1090. [PMID: 31279009 PMCID: PMC7278082 DOI: 10.1016/j.jaci.2019.06.028] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Revised: 05/29/2019] [Accepted: 06/19/2019] [Indexed: 12/22/2022]
Abstract
BACKGROUND Actin remodeling is a key regulator of mast cell (MC) migration and secretion. However, the precise mechanism underlying the coordination of these processes has remained obscure. OBJECTIVE We sought to characterize the actin rearrangements that occur during MC secretion or chemotactic migration and identify the underlying mechanism of their coordination. METHODS Using high-resolution microscopy, we analyzed the dynamics of actin rearrangements in MCs triggered to migration by IL-8 or prostaglandin E2 or to FcεRI-stimulated secretion. RESULTS We show that a major feature of the actin skeleton in MCs stimulated to migration is the buildup of pericentral actin clusters that prevent cell flattening and converge the secretory granules (SGs) in the cell center. This migratory phenotype is replaced on encounter of an IgE cross-linking antigen that stimulates secretion through a secretory phenotype characterized by cell flattening, reduction of actin mesh density, ruffling of cortical actin, and mobilization of SGs. Furthermore, we show that knockdown of mammalian diaphanous-related formin 1 (mDia1) inhibits chemotactic migration and its typical actin rearrangements, whereas expression of an active mDia1 mutant recapitulates the migratory actin phenotype and enhances cell migration while inhibiting FcεRI-triggered secretion. However, mice deficient in mDia1 appear to have normal numbers of MCs in various organs at baseline. CONCLUSION Our results demonstrate a unique role of actin rearrangements in clustering the SGs and inhibiting their secretion during MC migration. We identify mDia1 as a novel regulator of MC response that coordinates MC chemotaxis and secretion through its actin-nucleating activity.
Collapse
Affiliation(s)
- Ofir Klein
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Rebecca A Krier-Burris
- Department of Medicine, Division of Allergy-Immunology, Northwestern University Feinberg School of Medicine, Chicago, Ill
| | - Pia Lazki-Hagenbach
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Yaara Gorzalczany
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Yang Mei
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, Ill
| | - Peng Ji
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, Ill
| | - Bruce S Bochner
- Department of Medicine, Division of Allergy-Immunology, Northwestern University Feinberg School of Medicine, Chicago, Ill
| | - Ronit Sagi-Eisenberg
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
17
|
Grueb SS, Muhs S, Popp Y, Schmitt S, Geyer M, Lin YN, Windhorst S. The formin Drosophila homologue of Diaphanous2 (Diaph2) controls microtubule dynamics in colorectal cancer cells independent of its FH2-domain. Sci Rep 2019; 9:5352. [PMID: 30926831 PMCID: PMC6441084 DOI: 10.1038/s41598-019-41731-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Accepted: 03/12/2019] [Indexed: 12/21/2022] Open
Abstract
In this study, we analyzed the functional role of the formin Drosophila Homologue of Diaphanous2 (Diaph2) in colorectal cancer cells. We show that stable down-regulation of Diaph2 expression in HT29 cells decreased chromosome alignment and the velocity of chromosome movement during M-phase, thus reducing the proliferation rate and colony formation. In interphase cells, Diaph2 was diffusely distributed in the cytosol, while in metaphase cells the protein was located to spindle microtubules (MTs). Diaph2-depletion increased the concentration of stable spindle MTs, showing that the formin is required to control spindle MT-dynamics. Our cellular data indicate that Diaph2-controls spindle MT-dynamics independent of Cdc42 activity and our in vitro results reveal that bacterially produced full-length (FL) Diaph2 strongly altered MT-dynamics in absence of Cdc42, where its actin-nucleating activity is auto-inhibited. FL-Diaph2 mediates a 10-fold increase in MT-polymerization compared to the Diaph2-FH2-domain. Interestingly, a Diaph2-mutant lacking the FH2-domain (ΔFH2) increased MT-polymerization to a similar extent as the FH2-domain, indicating the existence of a second MT-binding domain. However, in contrast to FL-Diaph2 and the FH2-domain, ΔFH2 did not alter the density of taxol-stabilized MTs. Thus, the FH2-domain and the second Diaph2-binding domain appear to control MT-dynamics by different mechanisms. In summary, our data indicate that Diaph2 controls M-phase progression under basal conditions by regulating spindle MT-dynamics. In addition, a region outside of the canonical MT-regulating FH2-domain is involved in Diaph2-mediated control of MT-dynamics.
Collapse
Affiliation(s)
- Saskia S Grueb
- Department of Biochemistry and Signal Transduction, University Medical Center Hamburg-Eppendorf Martinistrasse 52, D-20246, Hamburg, Germany
| | - Stefanie Muhs
- Department of Biochemistry and Signal Transduction, University Medical Center Hamburg-Eppendorf Martinistrasse 52, D-20246, Hamburg, Germany
| | - Yannes Popp
- Department of Biochemistry and Signal Transduction, University Medical Center Hamburg-Eppendorf Martinistrasse 52, D-20246, Hamburg, Germany
| | - Sebastian Schmitt
- Institute of Structural Biology, University of Bonn, Sigmund-Freud-Str. 25, D-53127, Bonn, Germany
| | - Matthias Geyer
- Institute of Structural Biology, University of Bonn, Sigmund-Freud-Str. 25, D-53127, Bonn, Germany
| | - Yuan-Na Lin
- Department of Biochemistry and Signal Transduction, University Medical Center Hamburg-Eppendorf Martinistrasse 52, D-20246, Hamburg, Germany
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf Martinistrasse 52, 52 D-20246, Hamburg, Germany
| | - Sabine Windhorst
- Department of Biochemistry and Signal Transduction, University Medical Center Hamburg-Eppendorf Martinistrasse 52, D-20246, Hamburg, Germany.
| |
Collapse
|
18
|
Abstract
Formin homology proteins (formins) are a highly conserved family of cytoskeletal remodeling proteins that are involved in a diverse array of cellular functions. Formins are best known for their ability to regulate actin dynamics, but the same functional domains also govern stability and organization of microtubules. It is thought that this dual activity allows them to coordinate the activity of these two major cytoskeletal networks and thereby influence cellular architecture. Golgi ribbon assembly is dependent upon cooperative interactions between actin filaments and cytoplasmic microtubules originating both at the Golgi itself and from the centrosome. Similarly, centrosome assembly, centriole duplication, and centrosome positioning are also reliant on a dialogue between both cytoskeletal networks. As presented in this chapter, a growing body of evidence suggests that multiple formin proteins play essential roles in these central cellular processes.
Collapse
Affiliation(s)
- John Copeland
- Faculty of Medicine, Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada.
| |
Collapse
|
19
|
Multiple roles of the actin and microtubule-regulating formins in the developing brain. Neurosci Res 2019; 138:59-69. [DOI: 10.1016/j.neures.2018.09.008] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Revised: 08/22/2018] [Accepted: 08/23/2018] [Indexed: 01/08/2023]
|
20
|
Higashi T, Stephenson RE, Miller AL. Comprehensive analysis of formin localization in Xenopus epithelial cells. Mol Biol Cell 2018; 30:82-95. [PMID: 30379611 PMCID: PMC6337911 DOI: 10.1091/mbc.e18-02-0133] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Reorganization of the actin cytoskeleton is crucial for cellular processes, including cytokinesis and cell–cell junction remodeling. Formins are conserved processive actin-polymerizing machines that regulate actin dynamics by nucleating, elongating, and bundling linear actin filaments. Because the formin family is large, with at least 15 members in vertebrates, there have not been any comprehensive studies examining formin localization and function within a common cell type. Here, we characterized the localization of all 15 formins in epithelial cells of Xenopus laevis gastrula-stage embryos. Dia1 and Dia2 localized to tight junctions, while Fhod1 and Fhod3 localized to adherens junctions. Only Dia3 strongly localized at the cytokinetic contractile ring. The Diaphanous inhibitory domain–dimerization domain (DID-DD) region of Dia1 was sufficient for Dia1 localization, and overexpression of a Dia1 DID-DD fragment competitively removed Dia1 and Dia2 from cell–cell junctions. In Dia1 DID-DD–overexpressing cells, Dia1 and Dia2 were mislocalized to the contractile ring, and cells exhibited increased cytokinesis failure. This work provides a comprehensive analysis of the localization of all 15 vertebrate formins in epithelial cells and suggests that misregulated formin localization results in epithelial cytokinesis failure.
Collapse
Affiliation(s)
- Tomohito Higashi
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109
| | - Rachel E Stephenson
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109
| | - Ann L Miller
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109
| |
Collapse
|
21
|
Fernández-Barrera J, Alonso MA. Coordination of microtubule acetylation and the actin cytoskeleton by formins. Cell Mol Life Sci 2018; 75:3181-3191. [PMID: 29947928 PMCID: PMC11105221 DOI: 10.1007/s00018-018-2855-3] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Revised: 05/22/2018] [Accepted: 06/12/2018] [Indexed: 12/12/2022]
Abstract
The acetylation of the lysine 40 residue of α-tubulin was described more than 30 years ago and has been the subject of intense research ever since. Although the exact function of this covalent modification of tubulin in the cell remains unknown, it has been established that tubulin acetylation confers resilience to mechanical stress on the microtubules. Formins have a dual role in the fate of the actin and tubulin cytoskeletons. On the one hand, they catalyze the formation of actin filaments, and on the other, they bind microtubules, act on their stability, and regulate their acetylation and alignment with actin fibers. Recent evidence indicates that formins coordinate the actin cytoskeleton and tubulin acetylation by modulating the levels of free globular actin (G-actin). G-actin, in turn, controls the activity of the myocardin-related transcription factor-serum response factor transcriptional complex that regulates the expression of the α-tubulin acetyltransferase 1 (α-TAT1) gene, which encodes the main enzyme responsible for tubulin acetylation. The effect of formins on tubulin acetylation is the combined result of their ability to activate α-TAT1 gene transcription and of their capacity to regulate microtubule stabilization. The contribution of these two mechanisms in different formins is discussed, particularly with respect to INF2, a formin that is mutated in hereditary human renal and neurodegenerative disorders.
Collapse
Affiliation(s)
- Jaime Fernández-Barrera
- Department of Cell Biology and Immunology, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas and Universidad Autónoma, Madrid, Spain
| | - Miguel A Alonso
- Department of Cell Biology and Immunology, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas and Universidad Autónoma, Madrid, Spain.
| |
Collapse
|
22
|
Miao Y, Tipakornsaowapak T, Zheng L, Mu Y, Lewellyn E. Phospho-regulation of intrinsically disordered proteins for actin assembly and endocytosis. FEBS J 2018; 285:2762-2784. [PMID: 29722136 DOI: 10.1111/febs.14493] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Revised: 04/04/2018] [Accepted: 04/26/2018] [Indexed: 12/13/2022]
Abstract
Actin filament assembly contributes to the endocytic pathway pleiotropically, with active roles in clathrin-dependent and clathrin-independent endocytosis as well as subsequent endosomal trafficking. Endocytosis comprises a series of dynamic events, including the initiation of membrane curvature, bud invagination, vesicle abscission and subsequent vesicular transport. The ultimate success of endocytosis requires the coordinated activities of proteins that trigger actin polymerization, recruit actin-binding proteins (ABPs) and organize endocytic proteins (EPs) that promote membrane curvature through molecular crowding or scaffolding mechanisms. A particularly interesting phenomenon is that multiple EPs and ABPs contain a substantial percentage of intrinsically disordered regions (IDRs), which can contribute to protein coacervation and phase separation. In addition, intrinsically disordered proteins (IDPs) frequently contain sites for post-translational modifications (PTMs) such as phosphorylation, and these modifications exhibit a high preference for IDR residues [Groban ES et al. (2006) PLoS Comput Biol 2, e32]. PTMs are implicated in regulating protein function by modulating the protein conformation, protein-protein interactions and the transition between order and disorder states of IDPs. The molecular mechanisms by which IDRs of ABPs and EPs fine-tune actin assembly and endocytosis remain mostly unexplored and elusive. In this review, we analyze protein sequences of budding yeast EPs and ABPs, and discuss the potential underlying mechanisms for regulating endocytosis and actin assembly through the emerging concept of IDR-mediated protein multivalency, coacervation, and phase transition, with an emphasis on the phospho-regulation of IDRs. Finally, we summarize the current understanding of how these mechanisms coordinate actin cytoskeleton assembly and membrane curvature formation during endocytosis in budding yeast.
Collapse
Affiliation(s)
- Yansong Miao
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore.,School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore, Singapore
| | | | - Liangzhen Zheng
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Yuguang Mu
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Eric Lewellyn
- Department of Biology, Division of Natural Sciences, St Norbert College, De Pere, WI, USA
| |
Collapse
|
23
|
Migh E, Götz T, Földi I, Szikora S, Gombos R, Darula Z, Medzihradszky KF, Maléth J, Hegyi P, Sigrist S, Mihály J. Microtubule organization in presynaptic boutons relies on the formin DAAM. Development 2018; 145:dev158519. [PMID: 29487108 DOI: 10.1242/dev.158519] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Accepted: 02/14/2018] [Indexed: 02/02/2023]
Abstract
Regulation of the cytoskeleton is fundamental to the development and function of synaptic terminals, such as neuromuscular junctions. Despite the identification of numerous proteins that regulate synaptic actin and microtubule dynamics, the mechanisms of cytoskeletal control during terminal arbor formation have remained largely elusive. Here, we show that DAAM, a member of the formin family of cytoskeleton organizing factors, is an important presynaptic regulator of neuromuscular junction development in Drosophila We demonstrate that the actin filament assembly activity of DAAM plays a negligible role in terminal formation; rather, DAAM is necessary for synaptic microtubule organization. Genetic interaction studies consistently link DAAM with the Wg/Ank2/Futsch module of microtubule regulation and bouton formation. Finally, we provide evidence that DAAM is tightly associated with the synaptic active zone scaffold, and electrophysiological data point to a role in the modulation of synaptic vesicle release. Based on these results, we propose that DAAM is an important cytoskeletal effector element of the Wg/Ank2 pathway involved in the determination of basic synaptic structures, and, additionally, that DAAM may couple the active zone scaffold to the presynaptic cytoskeleton.
Collapse
Affiliation(s)
- Ede Migh
- Institute of Genetics, Biological Research Centre, Hungarian Academy of Sciences, MTA-SZBK NAP B Axon Growth and Regeneration Group, Temesvári krt. 62, Szeged H-6726, Hungary
| | - Torsten Götz
- Institut für Biologie/Genetik and NeuroCure, Freie Universitat Berlin, Takustrasse 6, D-14195 Berlin, Germany
| | - István Földi
- Institute of Genetics, Biological Research Centre, Hungarian Academy of Sciences, MTA-SZBK NAP B Axon Growth and Regeneration Group, Temesvári krt. 62, Szeged H-6726, Hungary
| | - Szilárd Szikora
- Institute of Genetics, Biological Research Centre, Hungarian Academy of Sciences, MTA-SZBK NAP B Axon Growth and Regeneration Group, Temesvári krt. 62, Szeged H-6726, Hungary
| | - Rita Gombos
- Institute of Genetics, Biological Research Centre, Hungarian Academy of Sciences, MTA-SZBK NAP B Axon Growth and Regeneration Group, Temesvári krt. 62, Szeged H-6726, Hungary
| | - Zsuzsanna Darula
- Laboratory of Proteomics Research, Biological Research Centre, Hungarian Academy of Sciences, Szeged H-6726, Hungary
| | - Katalin F Medzihradszky
- Laboratory of Proteomics Research, Biological Research Centre, Hungarian Academy of Sciences, Szeged H-6726, Hungary
| | - József Maléth
- MTA-SZTE Translational Gastroenterology Research Group, Szeged H-6725, Hungary
| | - Péter Hegyi
- MTA-SZTE Translational Gastroenterology Research Group, Szeged H-6725, Hungary
- Institute for Translational Medicine, University of Pecs, Pécs H-7624, Hungary
| | - Stephan Sigrist
- Institut für Biologie/Genetik and NeuroCure, Freie Universitat Berlin, Takustrasse 6, D-14195 Berlin, Germany
| | - József Mihály
- Institute of Genetics, Biological Research Centre, Hungarian Academy of Sciences, MTA-SZBK NAP B Axon Growth and Regeneration Group, Temesvári krt. 62, Szeged H-6726, Hungary
| |
Collapse
|
24
|
Tuna M, Amos CI. Next generation sequencing and its applications in HPV-associated cancers. Oncotarget 2018; 8:8877-8889. [PMID: 27784002 PMCID: PMC5352450 DOI: 10.18632/oncotarget.12830] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Accepted: 10/14/2016] [Indexed: 12/18/2022] Open
Abstract
Approximately 18% of all human cancers have a viral etiology, and human papillomavirus (HPV) has been identified as one of the most prevalent viruses that plays causative role in nearly all cervical cancers and, in addition, in subset of head and neck, anal, penile and vulvar cancers. The recent introduction of next generation sequencing (NGS) and other omics approaches have resulted in comprehensive knowledge on the pathogenesis of HPV-driven tumors. Specifically, these approaches have provided detailed information on genomic HPV integration sites, disrupted genes and pathways, and common and distinct genetic and epigenetic alterations in different human HPV-associated cancers. This review focuses on HPV integration sites, its concomitantly disrupted genes and pathways and its functional consequences in both cervical and head and neck cancers. Integration of NGS data with other omics and clinical data is crucial to better understand the pathophysiology of each individual malignancy and, based on this, to select targets and to design effective personalized treatment options.
Collapse
Affiliation(s)
- Musaffe Tuna
- Department of Biomedical Data Science, Geisel School of Medicine, Dartmouth College, Lebanon
| | - Christopher I Amos
- Department of Biomedical Data Science, Geisel School of Medicine, Dartmouth College, Lebanon
| |
Collapse
|
25
|
Fernández-Barrera J, Bernabé-Rubio M, Casares-Arias J, Rangel L, Fernández-Martín L, Correas I, Alonso MA. The actin-MRTF-SRF transcriptional circuit controls tubulin acetylation via α-TAT1 gene expression. J Cell Biol 2018; 217:929-944. [PMID: 29321169 PMCID: PMC5839776 DOI: 10.1083/jcb.201702157] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Revised: 06/28/2017] [Accepted: 12/11/2017] [Indexed: 02/07/2023] Open
Abstract
The role of formins in microtubules is not well understood. In this study, we have investigated the mechanism by which INF2, a formin mutated in degenerative renal and neurological hereditary disorders, controls microtubule acetylation. We found that silencing of INF2 in epithelial RPE-1 cells produced a dramatic drop in tubulin acetylation, increased the G-actin/F-actin ratio, and impaired myocardin-related transcription factor (MRTF)/serum response factor (SRF)-dependent transcription, which is known to be repressed by increased levels of G-actin. The effect on tubulin acetylation was caused by the almost complete absence of α-tubulin acetyltransferase 1 (α-TAT1) messenger RNA (mRNA). Activation of the MRTF-SRF transcriptional complex restored α-TAT1 mRNA levels and tubulin acetylation. Several functional MRTF-SRF-responsive elements were consistently identified in the α-TAT1 gene. The effect of INF2 silencing on microtubule acetylation was also observed in epithelial ECV304 cells, but not in Jurkat T cells. Therefore, the actin-MRTF-SRF circuit controls α-TAT1 transcription. INF2 regulates the circuit, and hence microtubule acetylation, in cell types where it has a prominent role in actin polymerization.
Collapse
Affiliation(s)
- Jaime Fernández-Barrera
- Department of Cell Biology and Immunology, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, Madrid, Spain
| | - Miguel Bernabé-Rubio
- Department of Cell Biology and Immunology, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, Madrid, Spain
| | - Javier Casares-Arias
- Department of Cell Biology and Immunology, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, Madrid, Spain
| | - Laura Rangel
- Department of Cell Biology and Immunology, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, Madrid, Spain.,Department of Molecular Biology, Universidad Autónoma de Madrid, Madrid, Spain
| | - Laura Fernández-Martín
- Department of Cell Biology and Immunology, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, Madrid, Spain
| | - Isabel Correas
- Department of Cell Biology and Immunology, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, Madrid, Spain.,Department of Molecular Biology, Universidad Autónoma de Madrid, Madrid, Spain
| | - Miguel A Alonso
- Department of Cell Biology and Immunology, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, Madrid, Spain
| |
Collapse
|
26
|
Szikora S, Földi I, Tóth K, Migh E, Vig A, Bugyi B, Maléth J, Hegyi P, Kaltenecker P, Sanchez-Soriano N, Mihály J. The formin DAAM is required for coordination of the actin and microtubule cytoskeleton in axonal growth cones. J Cell Sci 2017; 130:2506-2519. [PMID: 28606990 DOI: 10.1242/jcs.203455] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Accepted: 06/05/2017] [Indexed: 01/10/2023] Open
Abstract
Directed axonal growth depends on correct coordination of the actin and microtubule cytoskeleton in the growth cone. However, despite the relatively large number of proteins implicated in actin-microtubule crosstalk, the mechanisms whereby actin polymerization is coupled to microtubule stabilization and advancement in the peripheral growth cone remained largely unclear. Here, we identified the formin Dishevelled-associated activator of morphogenesis (DAAM) as a novel factor playing a role in concerted regulation of actin and microtubule remodeling in Drosophilamelanogaster primary neurons. In vitro, DAAM binds to F-actin as well as to microtubules and has the ability to crosslink the two filament systems. Accordingly, DAAM associates with the neuronal cytoskeleton, and a significant fraction of DAAM accumulates at places where the actin filaments overlap with that of microtubules. Loss of DAAM affects growth cone and microtubule morphology, and several aspects of microtubule dynamics; and biochemical and cellular assays revealed a microtubule stabilization activity and binding to the microtubule tip protein EB1. Together, these data suggest that, besides operating as an actin assembly factor, DAAM is involved in linking actin remodeling in filopodia to microtubule stabilization during axonal growth.
Collapse
Affiliation(s)
- Szilárd Szikora
- Institute of Genetics, Biological Research Centre, Hungarian Academy of Sciences, MTA-SZBK NAP B Axon Growth and Regeneration Group, Temesvári krt. 62, Szeged H-6726, Hungary
| | - István Földi
- Institute of Genetics, Biological Research Centre, Hungarian Academy of Sciences, MTA-SZBK NAP B Axon Growth and Regeneration Group, Temesvári krt. 62, Szeged H-6726, Hungary
| | - Krisztina Tóth
- Institute of Genetics, Biological Research Centre, Hungarian Academy of Sciences, MTA-SZBK NAP B Axon Growth and Regeneration Group, Temesvári krt. 62, Szeged H-6726, Hungary
| | - Ede Migh
- Institute of Genetics, Biological Research Centre, Hungarian Academy of Sciences, MTA-SZBK NAP B Axon Growth and Regeneration Group, Temesvári krt. 62, Szeged H-6726, Hungary
| | - Andrea Vig
- University of Pécs, Medical School, Department of Biophysics, Szigeti str. 12, Pécs H-7624, Hungary
| | - Beáta Bugyi
- University of Pécs, Medical School, Department of Biophysics, Szigeti str. 12, Pécs H-7624, Hungary
- Szentágothai Research Center, Ifjúság str. 34, Pécs H-7624, Hungary
| | - József Maléth
- MTA-SZTE Translational Gastroenterology Research Group, First Department of Internal Medicine, Szeged H-6720, Hungary
| | - Péter Hegyi
- MTA-SZTE Translational Gastroenterology Research Group, First Department of Internal Medicine, Szeged H-6720, Hungary
- Institute for Translational Medicine, Department of Pathophysiology, University of Pécs, Pécs H-7624, Hungary
| | - Péter Kaltenecker
- Institute for Translational Medicine, Department of Cellular and Molecular Physiology, University of Liverpool, Liverpool L69 3BX, UK
| | - Natalia Sanchez-Soriano
- Institute for Translational Medicine, Department of Cellular and Molecular Physiology, University of Liverpool, Liverpool L69 3BX, UK
| | - József Mihály
- Institute of Genetics, Biological Research Centre, Hungarian Academy of Sciences, MTA-SZBK NAP B Axon Growth and Regeneration Group, Temesvári krt. 62, Szeged H-6726, Hungary
| |
Collapse
|
27
|
Diaphanous-related formin 1 as a target for tumor therapy. Biochem Soc Trans 2017; 44:1289-1293. [PMID: 27911711 DOI: 10.1042/bst20160120] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Revised: 08/01/2016] [Accepted: 08/17/2016] [Indexed: 12/25/2022]
Abstract
Formins nucleate actin and stabilize microtubules (MTs). Expression of the formin Diaphanous homolog 1 (DIAPH1) is increased in malignant colon carcinoma cells, while expression of DIAPH3 is up-regulated in breast and prostate carcinoma cells. Both DIAPH1 isoforms are required to stabilize interphase MTs of cancer cells, and it has been shown that loss of this function decreases the metastatic potential of these cells. Moreover, depletion of DIAPH3 increases the sensitivity of breast and prostate carcinoma cells to taxanes. In contrast with DIAPH1 + 3, DIAPH2 regulates metaphase MTs of tumor cells by stabilizing binding of kinetochore MTs to chromosomes. Depletion of DIAPH2 impairs chromosome alignment, thus proper chromosome segregation during mitosis. In summary, expression of DIAPH formins in tumor cells is essential for stabilizing interphase or metaphase MTs, respectively. Thus, it would be very interesting to analyze if tumor cells exhibiting low DIAPH expression are more sensitive to taxanes than those with high DIAPH expression.
Collapse
|
28
|
Guo A, Huang S, Yu J, Wang H, Li H, Pei G, Shen L. Single-Cell Dynamic Analysis of Mitosis in Haploid Embryonic Stem Cells Shows the Prolonged Metaphase and Its Association with Self-diploidization. Stem Cell Reports 2017; 8:1124-1134. [PMID: 28457886 PMCID: PMC5425685 DOI: 10.1016/j.stemcr.2017.03.025] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2016] [Revised: 03/30/2017] [Accepted: 03/31/2017] [Indexed: 12/30/2022] Open
Abstract
The recent establishment of mammalian haploid embryonic stem cells (ESCs) provides new possibilities for genetic screening and for understanding genome evolution and function. However, the dynamics of mitosis in haploid ESCs is still unclear. Here, we report that the duration of mitosis in haploid ESCs, especially the metaphase, is significantly longer than that in diploid ESCs. Delaying mitosis by chemicals increased self-diploidization of haploid ESCs, while shortening mitosis stabilized haploid ESCs. Taken together, our study suggests that the delayed mitosis of haploid ESCs is associated with self-diploidization. Mitosis is prolonged in haploid ESCs, especially in metaphase Self-diploidization might be associated with the prolonged mitosis of haploid ESCs
Collapse
Affiliation(s)
- Ao Guo
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Shichao Huang
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai 200031, China; Shanghai Key Laboratory of Signaling and Disease Research, Laboratory of Receptor-based Bio-medicine, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Jiali Yu
- Life Sciences Institute, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Huihan Wang
- Life Sciences Institute, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Haisen Li
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Gang Pei
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai 200031, China; Shanghai Key Laboratory of Signaling and Disease Research, Laboratory of Receptor-based Bio-medicine, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China.
| | - Li Shen
- Life Sciences Institute, Zhejiang University, Hangzhou, Zhejiang 310058, China.
| |
Collapse
|
29
|
Wheelock MS, Wynne DJ, Tseng BS, Funabiki H. Dual recognition of chromatin and microtubules by INCENP is important for mitotic progression. J Cell Biol 2017; 216:925-941. [PMID: 28314740 PMCID: PMC5379950 DOI: 10.1083/jcb.201609061] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Revised: 01/11/2017] [Accepted: 02/07/2017] [Indexed: 12/23/2022] Open
Abstract
The chromosomal passenger complex (CPC), composed of inner centromere protein (INCENP), Survivin, Borealin, and the kinase Aurora B, contributes to the activation of the mitotic checkpoint. The regulation of CPC function remains unclear. Here, we reveal that in addition to Survivin and Borealin, the single α-helix (SAH) domain of INCENP supports CPC localization to chromatin and the mitotic checkpoint. The INCENP SAH domain also mediates INCENP's microtubule binding, which is negatively regulated by Cyclin-dependent kinase-mediated phosphorylation of segments flanking the SAH domain. The microtubule-binding capacity of the SAH domain is important for mitotic arrest in conditions of suppressed microtubule dynamics, and the duration of mitotic arrest dictates the probability, but not the timing, of cell death. Although independent targeting of INCENP to microtubules or the kinetochore/centromere promotes the mitotic checkpoint, it is insufficient for a robust mitotic arrest. Altogether, our results demonstrate that dual recognition of chromatin and microtubules by CPC is important for checkpoint maintenance and determination of cell fate in mitosis.
Collapse
Affiliation(s)
- Michael S Wheelock
- Laboratory of Chromosome and Cell Biology, The Rockefeller University, New York, NY 10065
| | - David J Wynne
- Laboratory of Chromosome and Cell Biology, The Rockefeller University, New York, NY 10065.,Department of Biology, The College of New Jersey, Ewing, NJ 08628
| | - Boo Shan Tseng
- Laboratory of Chromosome and Cell Biology, The Rockefeller University, New York, NY 10065.,The School of Life Sciences, The University of Nevada Las Vegas, Las Vegas, NV 89154
| | - Hironori Funabiki
- Laboratory of Chromosome and Cell Biology, The Rockefeller University, New York, NY 10065
| |
Collapse
|
30
|
Disruptions in asymmetric centrosome inheritance and WDR62-Aurora kinase B interactions in primary microcephaly. Sci Rep 2017; 7:43708. [PMID: 28272472 PMCID: PMC5341122 DOI: 10.1038/srep43708] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Accepted: 01/26/2017] [Indexed: 12/11/2022] Open
Abstract
Recessive mutations in WD repeat domain 62 (WDR62) cause microcephaly and a wide spectrum of severe brain malformations. Disruption of the mouse ortholog results in microcephaly underlain by reduced proliferation of neocortical progenitors during late neurogenesis, abnormalities in asymmetric centrosome inheritance leading to neuronal migration delays, and altered neuronal differentiation. Spindle pole localization of WDR62 and mitotic progression are defective in patient-derived fibroblasts, which, similar to mouse neocortical progenitors, transiently arrest at prometaphase. Expression of WDR62 is closely correlated with components of the chromosome passenger complex (CPC), a key regulator of mitosis. Wild type WDR62, but not disease-associated mutant forms, interacts with the CPC core enzyme Aurora kinase B and staining of CPC components at centromeres is altered in patient-derived fibroblasts. Our findings demonstrate critical and diverse functions of WDR62 in neocortical development and provide insight into the mechanisms by which its disruption leads to a plethora of structural abnormalities.
Collapse
|
31
|
Abstract
Accurate chromosome segregation in mammalian cells is guided by the centromere, a specialized chromosome region defined by the histone H3 variant centromere protein A (CENP-A). It is not well understood how cells maintain CENP-A levels at centromeres while continuously going through genome replications and cell divisions. A MgcRacGAP-dependent small GTPase molecular switch has been shown as essential for centromeric CENP-A maintenance. By using quantitative imaging, pulse-chase and live cell analysis, a recent work has suggested that the diaphanous formin mDia2, a well-established small GTPase effector, functions downstream of this small GTPase pathway to maintain CENP-A levels at centromeres. A constitutively active mDia2 construct is able to rescue the CENP-A loading defect caused by MgcRacGAP depletion. This study has uncovered an unsuspected role of the cytoskeleton protein mDia2 as an effector of the MgcRacGAP-dependent small GTPase signaling inside the nucleus to participate in the epigenetic regulation of centromere maintenance during cell cycle.
Collapse
Affiliation(s)
- Chenshu Liu
- a Department of Pathology and Cell Biology , Columbia University Medical Center , New York , NY , USA
| | - Yinghui Mao
- a Department of Pathology and Cell Biology , Columbia University Medical Center , New York , NY , USA
| |
Collapse
|
32
|
Rao MV, Zaidel-Bar R. Formin-mediated actin polymerization at cell-cell junctions stabilizes E-cadherin and maintains monolayer integrity during wound repair. Mol Biol Cell 2016; 27:2844-56. [PMID: 27440924 PMCID: PMC5025271 DOI: 10.1091/mbc.e16-06-0429] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Accepted: 07/12/2016] [Indexed: 02/05/2023] Open
Abstract
Cadherin-mediated cell–cell adhesion is required for epithelial tissue integrity in homeostasis, during development, and in tissue repair. Fmnl3 and mDia1 cooperate in stabilizing E-cadherin at cell–cell junctions and facilitate strong cell adhesion and monolayer cohesion during collective cell migration. Cadherin-mediated cell–cell adhesion is required for epithelial tissue integrity in homeostasis, during development, and in tissue repair. E-cadherin stability depends on F-actin, but the mechanisms regulating actin polymerization at cell–cell junctions remain poorly understood. Here we investigated a role for formin-mediated actin polymerization at cell–cell junctions. We identify mDia1 and Fmnl3 as major factors enhancing actin polymerization and stabilizing E-cadherin at epithelial junctions. Fmnl3 localizes to adherens junctions downstream of Src and Cdc42 and its depletion leads to a reduction in F-actin and E-cadherin at junctions and a weakening of cell–cell adhesion. Of importance, Fmnl3 expression is up-regulated and junctional localization increases during collective cell migration. Depletion of Fmnl3 or mDia1 in migrating monolayers results in dissociation of leader cells and impaired wound repair. In summary, our results show that formin activity at epithelial cell–cell junctions is important for adhesion and the maintenance of epithelial cohesion during dynamic processes, such as wound repair.
Collapse
Affiliation(s)
- Megha Vaman Rao
- Mechanobiology Institute, National University of Singapore, Singapore 117411
| | - Ronen Zaidel-Bar
- Mechanobiology Institute, National University of Singapore, Singapore 117411 Department of Biomedical Engineering, National University of Singapore, Singapore 117575
| |
Collapse
|
33
|
Liu C, Mao Y. Diaphanous formin mDia2 regulates CENP-A levels at centromeres. J Cell Biol 2016; 213:415-24. [PMID: 27185834 PMCID: PMC4878093 DOI: 10.1083/jcb.201512034] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Accepted: 04/20/2016] [Indexed: 12/05/2022] Open
Abstract
The diaphanous formin mDia2, a protein involved in cytoskeletal control, is required for new CENP-A loading at centromeres during the cell cycle to maintain epigenetic markers. Centromeres of higher eukaryotes are epigenetically defined by centromere protein A (CENP-A), a centromere-specific histone H3 variant. The incorporation of new CENP-A into centromeres to maintain the epigenetic marker after genome replication in S phase occurs in G1 phase; however, how new CENP-A is loaded and stabilized remains poorly understood. Here, we identify the formin mDia2 as essential for stable replenishment of new CENP-A at centromeres. Quantitative imaging, pulse-chase analysis, and high-resolution ratiometric live-cell studies demonstrate that mDia2 and its nuclear localization are required to maintain CENP-A levels at centromeres. Depletion of mDia2 results in a prolonged centromere association of holiday junction recognition protein (HJURP), the chaperone required for CENP-A loading. A constitutively active form of mDia2 rescues the defect in new CENP-A loading caused by depletion of male germ cell Rac GTPase-activating protein (MgcRacGAP), a component of the small GTPase pathway essential for CENP-A maintenance. Thus, the formin mDia2 functions downstream of the MgcRacGAP-dependent pathway in regulating assembly of new CENP-A containing nucleosomes at centromeres.
Collapse
Affiliation(s)
- Chenshu Liu
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY 10032
| | - Yinghui Mao
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY 10032
| |
Collapse
|
34
|
Miao Y, Han X, Zheng L, Xie Y, Mu Y, Yates JR, Drubin DG. Fimbrin phosphorylation by metaphase Cdk1 regulates actin cable dynamics in budding yeast. Nat Commun 2016; 7:11265. [PMID: 27068241 PMCID: PMC4832064 DOI: 10.1038/ncomms11265] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Accepted: 03/07/2016] [Indexed: 12/15/2022] Open
Abstract
Actin cables, composed of actin filament bundles nucleated by formins, mediate intracellular transport for cell polarity establishment and maintenance. We previously observed that metaphase cells preferentially promote actin cable assembly through cyclin-dependent kinase 1 (Cdk1) activity. However, the relevant metaphase Cdk1 targets were not known. Here we show that the highly conserved actin filament crosslinking protein fimbrin is a critical Cdk1 target for actin cable assembly regulation in budding yeast. Fimbrin is specifically phosphorylated on threonine 103 by the metaphase cyclin–Cdk1 complex, in vivo and in vitro. On the basis of conformational simulations, we suggest that this phosphorylation stabilizes fimbrin's N-terminal domain, and modulates actin filament binding to regulate actin cable assembly and stability in cells. Overall, this work identifies fimbrin as a key target for cell cycle regulation of actin cable assembly in budding yeast, and suggests an underlying mechanism. Metaphase cells preferentially promote actin cable assembly through cyclin-dependent kinase 1 (Cdk1) activity. Here the authors identify fimbrin as one of the main metaphase Cdk1 targets for cell cycle regulation of actin cable assembly in budding yeast.
Collapse
Affiliation(s)
- Yansong Miao
- Department of Molecular and Cell Biology, University of California, Berkeley, California 94720-3202, USA.,School of Biological Sciences, Nanyang Technological University, Singapore 637551, Singapore.,School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore 637459, Singapore
| | - Xuemei Han
- Department of Chemical Physiology, The Scripps Research Institute, La Jolla, California 92037, USA
| | - Liangzhen Zheng
- School of Biological Sciences, Nanyang Technological University, Singapore 637551, Singapore
| | - Ying Xie
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore 637459, Singapore
| | - Yuguang Mu
- School of Biological Sciences, Nanyang Technological University, Singapore 637551, Singapore
| | - John R Yates
- Department of Chemical Physiology, The Scripps Research Institute, La Jolla, California 92037, USA
| | - David G Drubin
- Department of Molecular and Cell Biology, University of California, Berkeley, California 94720-3202, USA
| |
Collapse
|
35
|
Bartolini F, Andres-Delgado L, Qu X, Nik S, Ramalingam N, Kremer L, Alonso MA, Gundersen GG. An mDia1-INF2 formin activation cascade facilitated by IQGAP1 regulates stable microtubules in migrating cells. Mol Biol Cell 2016; 27:1797-808. [PMID: 27030671 PMCID: PMC4884070 DOI: 10.1091/mbc.e15-07-0489] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Accepted: 03/25/2016] [Indexed: 01/08/2023] Open
Abstract
The formin INF2 is required for stable Glu microtubule formation and inhibition of microtubule dynamics in NIH3T3 cells downstream of mDia1 and LPA. Evidence also shows that the formation of an mDia1/INF2 complex is necessary for microtubule stabilization stimulated by LPA and is regulated by IQGAP1. Multiple formins regulate microtubule (MT) arrays, but whether they function individually or in a common pathway is unknown. Lysophosphatidic acid (LPA) stimulates the formation of stabilized detyrosinated MTs (Glu MTs) in NIH3T3 fibroblasts through RhoA and the formin mDia1. Here we show that another formin, INF2, is necessary for mDia1-mediated induction of Glu MTs and regulation of MT dynamics and that mDia1 can be bypassed by activating INF2. INF2 localized to MTs after LPA treatment in an mDia1-dependent manner, suggesting that mDia1 regulates INF2. Mutants of either formin that disrupt their interaction failed to rescue MT stability in cells depleted of the respective formin, and the mDia1-interacting protein IQGAP1 regulated INF2’s localization to MTs and the induction of Glu MTs by either formin. The N-terminus of IQGAP1 associated with the C-terminus of INF2 directly, suggesting the possibility of a tripartite complex stimulated by LPA. Supporting this, the interaction of mDia1 and INF2 was induced by LPA and dependent on IQGAP1. Our data highlight a unique mechanism of formin action in which mDia1 and INF2 function in series to stabilize MTs and point to IQGAP1 as a scaffold that facilitates the activation of one formin by another.
Collapse
Affiliation(s)
- Francesca Bartolini
- Department of Pathology and Cell Biology, Columbia University, New York, NY 10032
| | - Laura Andres-Delgado
- Centro de Biologia Molecular Severo Ochoa, Consejo Superior de Investigaciones Cientificas and Universidad Autonoma de Madrid, 28049 Madrid, Spain
| | - Xiaoyi Qu
- Department of Pathology and Cell Biology, Columbia University, New York, NY 10032
| | - Sara Nik
- Department of Pathology and Cell Biology, Columbia University, New York, NY 10032
| | - Nagendran Ramalingam
- Department of Pathology and Cell Biology, Columbia University, New York, NY 10032
| | - Leonor Kremer
- Centro de Biologia Molecular Severo Ochoa, Consejo Superior de Investigaciones Cientificas and Universidad Autonoma de Madrid, 28049 Madrid, Spain
| | - Miguel A Alonso
- Centro de Biologia Molecular Severo Ochoa, Consejo Superior de Investigaciones Cientificas and Universidad Autonoma de Madrid, 28049 Madrid, Spain
| | - Gregg G Gundersen
- Department of Pathology and Cell Biology, Columbia University, New York, NY 10032
| |
Collapse
|
36
|
Abstract
Dynein light chains are accessory subunits of the cytoplasmic dynein complex, a minus-end directed microtubule motor. Here, we demonstrate that the dynein light chain Tctex-1 associates with unattached kinetochores and is essential for accurate chromosome segregation. Tctex-1 knockdown in cells does not affect the localization and function of dynein at the kinetochore, but produces a prolonged mitotic arrest with a few misaligned chromosomes, which are subsequently missegregated during anaphase. This function is independent of Tctex-1's association with dynein. The kinetochore localization of Tctex-1 is independent of the ZW10-dynein pathway, but requires the Ndc80 complex. Thus, our findings reveal a dynein independent role of Tctex-1 at the kinetochore to enhance the stability of kinetochore-microtubule attachment.
Collapse
Affiliation(s)
- Chenshu Liu
- a Department of Pathology and Cell Biology ; Columbia University College of Physicians and Surgeons ; New York , NY USA
| | | | | | | |
Collapse
|
37
|
Human phosphatase CDC14A is recruited to the cell leading edge to regulate cell migration and adhesion. Proc Natl Acad Sci U S A 2016; 113:990-5. [PMID: 26747605 DOI: 10.1073/pnas.1515605113] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Cell adhesion and migration are highly dynamic biological processes that play important roles in organ development and cancer metastasis. Their tight regulation by small GTPases and protein phosphorylation make interrogation of these key processes of great importance. We now show that the conserved dual-specificity phosphatase human cell-division cycle 14A (hCDC14A) associates with the actin cytoskeleton of human cells. To understand hCDC14A function at this location, we manipulated native loci to ablate hCDC14A phosphatase activity (hCDC14A(PD)) in untransformed hTERT-RPE1 and colorectal cancer (HCT116) cell lines and expressed the phosphatase in HeLa FRT T-Rex cells. Ectopic expression of hCDC14A induced stress fiber formation, whereas stress fibers were diminished in hCDC14A(PD) cells. hCDC14A(PD) cells displayed faster cell migration and less adhesion than wild-type controls. hCDC14A colocalized with the hCDC14A substrate kidney- and brain-expressed protein (KIBRA) at the cell leading edge and overexpression of KIBRA was able to reverse the phenotypes of hCDC14A(PD) cells. Finally, we show that ablation of hCDC14A activity increased the aggressive nature of cells in an in vitro tumor formation assay. Consistently, hCDC14A is down-regulated in many tumor tissues and reduced hCDC14A expression is correlated with poorer survival of patients with cancer, to suggest that hCDC14A may directly contribute to the metastatic potential of tumors. Thus, we have uncovered an unanticipated role for hCDC14A in cell migration and adhesion that is clearly distinct from the mitotic and cytokinesis functions of Cdc14/Flp1 in budding and fission yeast.
Collapse
|
38
|
Formins at the Junction. Trends Biochem Sci 2015; 41:148-159. [PMID: 26732401 DOI: 10.1016/j.tibs.2015.12.002] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Revised: 12/01/2015] [Accepted: 12/04/2015] [Indexed: 12/21/2022]
Abstract
The actin cytoskeleton and adhesion junctions are physically and functionally coupled at the cell-cell interface between epithelial cells. The actin regulatory complex Arp2/3 has an established role in the turnover of junctional actin; however, the role of formins, the largest group of actin regulators, is less clear. Formins dynamically shape the actin cytoskeleton and have various functions within cells. In this review we describe recent progress on how formins regulate actin dynamics at cell-cell contacts and highlight formin functions during polarized protein traffic necessary for epithelialization.
Collapse
|
39
|
Lenhart KF, DiNardo S. Somatic cell encystment promotes abscission in germline stem cells following a regulated block in cytokinesis. Dev Cell 2015; 34:192-205. [PMID: 26143993 DOI: 10.1016/j.devcel.2015.05.003] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2014] [Revised: 03/17/2015] [Accepted: 05/07/2015] [Indexed: 02/05/2023]
Abstract
In many tissues, the stem cell niche must coordinate behavior across multiple stem cell lineages. How this is achieved is largely unknown. We have identified delayed completion of cytokinesis in germline stem cells (GSCs) as a mechanism that regulates the production of stem cell daughters in the Drosophila testis. Through live imaging, we show that a secondary F-actin ring is formed through regulation of Cofilin activity to block cytokinesis progress after contractile ring disassembly. The duration of this block is controlled by Aurora B kinase. Additionally, we have identified a requirement for somatic cell encystment of the germline in promoting GSC abscission. We suggest that this non-autonomous role promotes coordination between stem cell lineages. These findings reveal the mechanisms by which cytokinesis is inhibited and reinitiated in GSCs and why such complex regulation exists within the stem cell niche.
Collapse
Affiliation(s)
- Kari F Lenhart
- Department of Cell and Developmental Biology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104-6058, USA
| | - Stephen DiNardo
- Department of Cell and Developmental Biology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104-6058, USA; Institute for Regenerative Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104-6058, USA.
| |
Collapse
|
40
|
Li N, Mruk DD, Tang EI, Wong CK, Lee WM, Silvestrini B, Cheng CY. Formins: Actin nucleators that regulate cytoskeletal dynamics during spermatogenesis. SPERMATOGENESIS 2015; 5:e1066476. [PMID: 26413414 DOI: 10.1080/21565562.2015.1066476] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2015] [Revised: 06/18/2015] [Accepted: 06/18/2015] [Indexed: 12/21/2022]
Abstract
Formins are a growing class of actin nucleation proteins that promote the polymerization of actin microfilaments, forming long stretches of actin microfilaments to confer actin filament bundling in mammalian cells. As such, microfilament bundles can be formed in specific cellular domains, in particular in motile mammalian cells, such as filopodia. Since ectoplasmic specialization (ES), a testis-specific adherens junction (AJ), at the Sertoli cell-cell and Sertoli-spermatid interface is constituted by arrays of actin microfilament bundles, it is likely that formins are playing a significant physiological role on the homeostasis of ES during the epithelial cycle of spermatogenesis. In this Commentary, we provide a timely discussion on formin 1 which was recently shown to be a crucial regulator of actin microfilaments at the ES in the rat testis (Li N et al. Endocrinology, 2015, in press; DOI: 10.1210/en.2015-1161, PMID:25901598). We also highlight research that is needed to unravel the functional significance of formins in spermatogenesis.
Collapse
Affiliation(s)
- Nan Li
- The Mary M. Wohlford Laboratory for Male Contraceptive Research; Center for Biomedical Research; Population Council ; New York, NY USA
| | - Dolores D Mruk
- The Mary M. Wohlford Laboratory for Male Contraceptive Research; Center for Biomedical Research; Population Council ; New York, NY USA
| | - Elizabeth I Tang
- The Mary M. Wohlford Laboratory for Male Contraceptive Research; Center for Biomedical Research; Population Council ; New York, NY USA
| | - Chris Kc Wong
- Department of Biology; Hong Kong Baptist University ; Hong Kong, China
| | - Will M Lee
- School of Biological Sciences; University of Hong Kong ; Hong Kong, China
| | | | - C Yan Cheng
- The Mary M. Wohlford Laboratory for Male Contraceptive Research; Center for Biomedical Research; Population Council ; New York, NY USA
| |
Collapse
|
41
|
Goins LM, Mullins RD. A novel tropomyosin isoform functions at the mitotic spindle and Golgi in Drosophila. Mol Biol Cell 2015; 26:2491-504. [PMID: 25971803 PMCID: PMC4571303 DOI: 10.1091/mbc.e14-12-1619] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Accepted: 05/05/2015] [Indexed: 12/28/2022] Open
Abstract
Most eukaryotic cells express multiple isoforms of the actin-binding protein tropomyosin that help construct a variety of cytoskeletal networks. Only one nonmuscle tropomyosin (Tm1A) has previously been described in Drosophila, but developmental defects caused by insertion of P-elements near tropomyosin genes imply the existence of additional, nonmuscle isoforms. Using biochemical and molecular genetic approaches, we identified three tropomyosins expressed in Drosophila S2 cells: Tm1A, Tm1J, and Tm2A. The Tm1A isoform localizes to the cell cortex, lamellar actin networks, and the cleavage furrow of dividing cells--always together with myosin-II. Isoforms Tm1J and Tm2A colocalize around the Golgi apparatus with the formin-family protein Diaphanous, and loss of either isoform perturbs cell cycle progression. During mitosis, Tm1J localizes to the mitotic spindle, where it promotes chromosome segregation. Using chimeras, we identified the determinants of tropomyosin localization near the C-terminus. This work 1) identifies and characterizes previously unknown nonmuscle tropomyosins in Drosophila, 2) reveals a function for tropomyosin in the mitotic spindle, and 3) uncovers sequence elements that specify isoform-specific localizations and functions of tropomyosin.
Collapse
Affiliation(s)
- Lauren M Goins
- Department of Cellular and Molecular Pharmacology, School of Medicine, University of California, San Francisco, San Francisco, CA 94158
| | - R Dyche Mullins
- Department of Cellular and Molecular Pharmacology, School of Medicine, University of California, San Francisco, San Francisco, CA 94158
| |
Collapse
|
42
|
SMIFH2 has effects on Formins and p53 that perturb the cell cytoskeleton. Sci Rep 2015; 5:9802. [PMID: 25925024 PMCID: PMC5386218 DOI: 10.1038/srep09802] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2014] [Accepted: 03/19/2015] [Indexed: 01/08/2023] Open
Abstract
Formin proteins are key regulators of the cytoskeleton involved in developmental and homeostatic programs, and human disease. For these reasons, small molecules interfering with Formins' activity have gained increasing attention. Among them, small molecule inhibitor of Formin Homology 2 domains (SMIFH2) is often used as a pharmacological Formin blocker. Although SMIFH2 inhibits actin polymerization by Formins and affects the actin cytoskeleton, its cellular mechanism of action and target specificity remain unclear. Here we show that SMIFH2 induces remodelling of actin filaments, microtubules and the Golgi complex as a result of its effects on Formins and p53. We found that SMIFH2 triggers alternated depolymerization-repolymerization cycles of actin and tubulin, increases cell migration, causes scattering of the Golgi complex, and also cytotoxicity at high dose. Moreover, SMIFH2 reduces expression and activity of p53 through a post-transcriptional, proteasome-independent mechanism that influences remodelling of the cytoskeleton. As the action of SMIFH2 may go beyond Formin inhibition, only short-term and low-dose SMIFH2 treatments minimize confounding effects induced by loss of p53 and cytotoxicity.
Collapse
|
43
|
Yoo H, Roth-Johnson EA, Bor B, Quinlan ME. Drosophila Cappuccino alleles provide insight into formin mechanism and role in oogenesis. Mol Biol Cell 2015; 26:1875-86. [PMID: 25788286 PMCID: PMC4436832 DOI: 10.1091/mbc.e14-11-1558] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2014] [Accepted: 03/10/2015] [Indexed: 11/11/2022] Open
Abstract
During Drosophila development, the formin actin nucleator Cappuccino (Capu) helps build a cytoplasmic actin mesh throughout the oocyte. Loss of Capu leads to female sterility, presumably because polarity determinants fail to localize properly in the absence of the mesh. To gain deeper insight into how Capu builds this actin mesh, we systematically characterized seven capu alleles, which have missense mutations in Capu's formin homology 2 (FH2) domain. We report that all seven alleles have deleterious effects on fly fertility and the actin mesh in vivo but have strikingly different effects on Capu's biochemical activity in vitro. Using a combination of bulk and single- filament actin-assembly assays, we find that the alleles differentially affect Capu's ability to nucleate and processively elongate actin filaments. We also identify a unique "loop" in the lasso region of Capu's FH2 domain. Removing this loop enhances Capu's nucleation, elongation, and F-actin-bundling activities in vitro. Together our results on the loop and the seven missense mutations provides mechanistic insight into formin function in general and Capu's role in the Drosophila oocyte in particular.
Collapse
Affiliation(s)
- Haneul Yoo
- Department of Chemistry and Biochemistry
| | - Elizabeth A Roth-Johnson
- Molecular Biology Interdepartmental PhD Program, University of California, Los Angeles, Los Angeles, CA 90095
| | - Batbileg Bor
- Molecular Biology Interdepartmental PhD Program, University of California, Los Angeles, Los Angeles, CA 90095
| | - Margot E Quinlan
- Department of Chemistry and Biochemistry Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095
| |
Collapse
|
44
|
The tumour suppressor DLC2 ensures mitotic fidelity by coordinating spindle positioning and cell-cell adhesion. Nat Commun 2014; 5:5826. [PMID: 25518808 PMCID: PMC4284802 DOI: 10.1038/ncomms6826] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2014] [Accepted: 11/07/2014] [Indexed: 02/07/2023] Open
Abstract
Dividing epithelial cells need to coordinate spindle positioning with shape changes to maintain cell–cell adhesion. Microtubule interactions with the cell cortex regulate mitotic spindle positioning within the plane of division. How the spindle crosstalks with the actin cytoskeleton to ensure faithful mitosis and spindle positioning is unclear. Here we demonstrate that the tumour suppressor DLC2, a negative regulator of Cdc42, and the interacting kinesin Kif1B coordinate cell junction maintenance and planar spindle positioning by regulating microtubule growth and crosstalk with the actin cytoskeleton. Loss of DLC2 induces the mislocalization of Kif1B, increased Cdc42 activity and cortical recruitment of the Cdc42 effector mDia3, a microtubule stabilizer and promoter of actin dynamics. Accordingly, DLC2 or Kif1B depletion promotes microtubule stabilization, defective spindle positioning, chromosome misalignment and aneuploidy. The tumour suppressor DLC2 and Kif1B are thus central components of a signalling network that guides spindle positioning, cell–cell adhesion and mitotic fidelity. Epithelial cells must position their mitotic spindle correctly to maintain cell–cell adhesion. Here Vitiello et al. show that the tumour suppressor DLC2 and the mitotic kinesin Kif1b coordinate microtubule–actin interactions upstream of mDia3, guiding spindle positioning and mitotic fidelity.
Collapse
|
45
|
|
46
|
Guo Y, Cheng L, Ahmad S, Mao Y. Formin mDia3: A novel target for Aurora B kinase. BIOARCHITECTURE 2014; 1:88-90. [PMID: 21866269 DOI: 10.4161/bioa.1.2.16240] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/21/2011] [Accepted: 04/29/2011] [Indexed: 02/01/2023]
Abstract
Stable attachment of kinetochores to spindle microtubules is essential for accurate chromosome segregation. We have shown that a kinetochore-associated formin protein, mDia3, contributes to the generation of stable kinetochore-microtubule attachment. The published report reviewed here shows an essential role of mDia3 in achieving metaphase chromosome alignment, and this function is directly regulated by Aurora B phosphorylation. Aurora B is a central component during the capture of spindle microtubules by kinetochores, in which it selectively eliminates incorrect attachments by phosphorylating a group of microtubule binding proteins at kinetochores to reduce their microtubule binding affinity. Here, we discuss the roles of Aurora B kinase and its substrates in achieving proper kinetochore-microtubule attachment.
Collapse
Affiliation(s)
- Yige Guo
- Department of Pathology and Cell Biology; Columbia University College of Physicians and Surgeons; New York, NY USA
| | | | | | | |
Collapse
|
47
|
The formin DIAPH1 (mDia1) regulates megakaryocyte proplatelet formation by remodeling the actin and microtubule cytoskeletons. Blood 2014; 124:3967-77. [PMID: 25298036 DOI: 10.1182/blood-2013-12-544924] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Megakaryocytes are highly specialized precursor cells that produce platelets via cytoplasmic extensions called proplatelets. Proplatelet formation (PPF) requires profound changes in microtubule and actin organization. In this work, we demonstrated that DIAPH1 (mDia1), a mammalian homolog of Drosophila diaphanous that works as an effector of the small GTPase Rho, negatively regulates PPF by controlling the dynamics of the actin and microtubule cytoskeletons. Moreover, we showed that inhibition of both DIAPH1 and the Rho-associated protein kinase (Rock)/myosin pathway increased PPF via coordination of both cytoskeletons. We provide evidence that 2 major effectors of the Rho GTPase pathway (DIAPH1 and Rock/myosin II) are involved not only in Rho-mediated stress fibers assembly, but also in the regulation of microtubule stability and dynamics during PPF.
Collapse
|
48
|
Abstract
Formin proteins were recognized as effectors of Rho GTPases some 15 years ago. They contribute to different cellular actin cytoskeleton structures by their ability to polymerize straight actin filaments at the barbed end. While not all formins necessarily interact with Rho GTPases, a subgroup of mammalian formins, termed Diaphanous-related formins or DRFs, were shown to be activated by small GTPases of the Rho superfamily. DRFs are autoinhibited in the resting state by an N- to C-terminal interaction that renders the central actin polymerization domain inactive. Upon the interaction with a GTP-bound Rho, Rac, or Cdc42 GTPase, the C-terminal autoregulation domain is displaced from its N-terminal recognition site and the formin becomes active to polymerize actin filaments. In this review we discuss the current knowledge on the structure, activation, and function of formin-GTPase interactions for the mammalian formin families Dia, Daam, FMNL, and FHOD. We describe both direct and indirect interactions of formins with GTPases, which lead to formin activation and cytoskeletal rearrangements. The multifaceted function of formins as effector proteins of Rho GTPases thus reflects the diversity of the actin cytoskeleton in cells.
Collapse
Affiliation(s)
- Sonja Kühn
- Center of Advanced European Studies and Research (caesar); Group Physical Biochemistry; Bonn, Germany
| | - Matthias Geyer
- Center of Advanced European Studies and Research (caesar); Group Physical Biochemistry; Bonn, Germany
| |
Collapse
|
49
|
Truong D, Copeland JW, Brumell JH. Bacterial subversion of host cytoskeletal machinery: hijacking formins and the Arp2/3 complex. Bioessays 2014; 36:687-96. [PMID: 24849003 DOI: 10.1002/bies.201400038] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The host actin nucleation machinery is subverted by many bacterial pathogens to facilitate their entry, motility, replication, and survival. The majority of research conducted in the past primarily focused on exploitation of a host actin nucleator, the Arp2/3 complex, by bacterial pathogens. Recently, new studies have begun to explore the role of formins, another family of host actin nucleators, in bacterial pathogenesis. This review provides an overview of recent advances in the study of the exploitation of the Arp2/3 complex and formins by bacterial pathogens. Secreted bacterial effector proteins seem to manipulate the regulation of these actin nucleators or functionally mimic them to drive bacterial entry, motility and survival within host cells. An enhanced understanding of how formins are exploited will provide us with greater insight into how a fundamental eurkaryotic cellular process is utilized by bacteria and will also advance our knowledge of host-pathogen interactions.
Collapse
Affiliation(s)
- Dorothy Truong
- Cell Biology Program, The Hospital for Sick Children, Toronto, ON, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | | | | |
Collapse
|
50
|
Pardo I, Lillemoe HA, Blosser RJ, Choi M, Sauder CAM, Doxey DK, Mathieson T, Hancock BA, Baptiste D, Atale R, Hickenbotham M, Zhu J, Glasscock J, Storniolo AMV, Zheng F, Doerge RW, Liu Y, Badve S, Radovich M, Clare SE. Next-generation transcriptome sequencing of the premenopausal breast epithelium using specimens from a normal human breast tissue bank. Breast Cancer Res 2014; 16:R26. [PMID: 24636070 PMCID: PMC4053088 DOI: 10.1186/bcr3627] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2013] [Accepted: 03/10/2014] [Indexed: 12/12/2022] Open
Abstract
Introduction Our efforts to prevent and treat breast cancer are significantly impeded by a lack of knowledge of the biology and developmental genetics of the normal mammary gland. In order to provide the specimens that will facilitate such an understanding, The Susan G. Komen for the Cure Tissue Bank at the IU Simon Cancer Center (KTB) was established. The KTB is, to our knowledge, the only biorepository in the world prospectively established to collect normal, healthy breast tissue from volunteer donors. As a first initiative toward a molecular understanding of the biology and developmental genetics of the normal mammary gland, the effect of the menstrual cycle and hormonal contraceptives on DNA expression in the normal breast epithelium was examined. Methods Using normal breast tissue from 20 premenopausal donors to KTB, the changes in the mRNA of the normal breast epithelium as a function of phase of the menstrual cycle and hormonal contraception were assayed using next-generation whole transcriptome sequencing (RNA-Seq). Results In total, 255 genes representing 1.4% of all genes were deemed to have statistically significant differential expression between the two phases of the menstrual cycle. The overwhelming majority (221; 87%) of the genes have higher expression during the luteal phase. These data provide important insights into the processes occurring during each phase of the menstrual cycle. There was only a single gene significantly differentially expressed when comparing the epithelium of women using hormonal contraception to those in the luteal phase. Conclusions We have taken advantage of a unique research resource, the KTB, to complete the first-ever next-generation transcriptome sequencing of the epithelial compartment of 20 normal human breast specimens. This work has produced a comprehensive catalog of the differences in the expression of protein-coding genes as a function of the phase of the menstrual cycle. These data constitute the beginning of a reference data set of the normal mammary gland, which can be consulted for comparison with data developed from malignant specimens, or to mine the effects of the hormonal flux that occurs during the menstrual cycle.
Collapse
|