1
|
Lemay SE, Montesinos MS, Grobs Y, Yokokawa T, Shimauchi T, Mougin M, Romanet C, Sauvaget M, Breuils-Bonnet S, Bourgeois A, Théberge C, Pelletier A, El Kabbout R, Martineau S, Yamamoto K, Akram M, Ray AS, Lippa B, Goodwin B, Lin FY, Wang H, Dowling JE, Lu M, Qiao Q, McTeague TA, Moy TI, Potus F, Provencher S, Boucherat O, Bonnet S. Exploring Integrin α5β1 as a Potential Therapeutic Target for Pulmonary Arterial Hypertension: Insights From Comprehensive Multicenter Preclinical Studies. Circulation 2025; 151:1162-1183. [PMID: 39829438 PMCID: PMC12011439 DOI: 10.1161/circulationaha.124.070693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 12/17/2024] [Indexed: 01/22/2025]
Abstract
BACKGROUND Pulmonary arterial hypertension (PAH) is characterized by obliterative vascular remodeling of the small pulmonary arteries (PAs) and progressive increase in pulmonary vascular resistance leading to right ventricular failure. Although several drugs are approved for the treatment of PAH, mortality rates remain high. Accumulating evidence supports a pathological function of integrins in vessel remodeling, which are gaining renewed interest as drug targets. However, their role in PAH remains largely unexplored. METHODS The expression of the RGD (arginylglycylaspartic acid)-binding integrin α5β1 was assessed in PAs, PA smooth muscle cells, and PA endothelial cells from patients with PAH and controls using NanoString, immunoblotting, and Mesoscale Discovery assays. RNA sequencing was conducted to identify gene networks regulated by α5β1 inhibition in PAH PA smooth muscle cells. The therapeutic efficacy of α5β1 inhibition was evaluated using a novel small molecule inhibitor and selective neutralizing antibodies in Sugen/hypoxia and monocrotaline rat models, with validation by an external contract research organization. Comparisons were made against standard-of-care therapies (ie, macitentan, tadalafil) and sotatercept and efficacy was assessed using echocardiographic, hemodynamic, and histological assessments. Ex vivo studies using human precision-cut lung slices were performed to further assess the effects of α5β1 inhibition on pulmonary vascular remodeling. RESULTS We found that the arginine-glycine-aspartate RGD-binding integrin α5β1 is upregulated in PA endothelial cells and PA smooth muscle cells from patients with PAH and remodeled PAs from animal models. Blockade of the integrin α5β1 or depletion of the α5 subunit downregulated FOXM1 (forkhead box protein M1)-regulated gene networks, resulting in mitotic defects and inhibition of the pro-proliferative and apoptosis-resistant phenotype of PAH cells. We demonstrated that α5β1 integrin blockade safely attenuates pulmonary vascular remodeling and improves hemodynamics and right ventricular function and matched or exceeded the efficacy of standard of care and sotatercept in multiple preclinical models. Ex vivo studies further validated its potential in reversing advanced remodeling in human precision-cut lung slices. CONCLUSIONS These findings establish α5β1 integrin as a pivotal driver of PAH pathology and we propose its inhibition as a novel, safe, and effective therapeutic strategy for PAH.
Collapse
Affiliation(s)
- Sarah-Eve Lemay
- Pulmonary Hypertension Research Group, Québec Heart and Lung Institute Research Center, Quebec City, QC, Canada (S.-E.L., Y.G., T.Y., T.S., M.M., C.R., M.S., S.B.-B., A.B., C.T., A.P., R.E.K., S.M., K.Y., F.P., S.P., O.B., S.B.)
| | - Mónica S. Montesinos
- Morphic Therapeutic, Inc, Waltham, MA (M.S.M., M.A., A.S.R., B.L., B.G., F.-Y.L., H.W., J.E.D., M.L., Q.Q., T.A.M., T.I.M.)
| | - Yann Grobs
- Pulmonary Hypertension Research Group, Québec Heart and Lung Institute Research Center, Quebec City, QC, Canada (S.-E.L., Y.G., T.Y., T.S., M.M., C.R., M.S., S.B.-B., A.B., C.T., A.P., R.E.K., S.M., K.Y., F.P., S.P., O.B., S.B.)
| | - Tetsuro Yokokawa
- Pulmonary Hypertension Research Group, Québec Heart and Lung Institute Research Center, Quebec City, QC, Canada (S.-E.L., Y.G., T.Y., T.S., M.M., C.R., M.S., S.B.-B., A.B., C.T., A.P., R.E.K., S.M., K.Y., F.P., S.P., O.B., S.B.)
- Department of Cardiovascular Medicine, Fukushima Medical University, Japan (T.Y.)
| | - Tsukasa Shimauchi
- Pulmonary Hypertension Research Group, Québec Heart and Lung Institute Research Center, Quebec City, QC, Canada (S.-E.L., Y.G., T.Y., T.S., M.M., C.R., M.S., S.B.-B., A.B., C.T., A.P., R.E.K., S.M., K.Y., F.P., S.P., O.B., S.B.)
- Department of Anesthesiology, St Mary’s Hospital, Kurume, Fukuoka, Japan (T.S.)
| | - Manon Mougin
- Pulmonary Hypertension Research Group, Québec Heart and Lung Institute Research Center, Quebec City, QC, Canada (S.-E.L., Y.G., T.Y., T.S., M.M., C.R., M.S., S.B.-B., A.B., C.T., A.P., R.E.K., S.M., K.Y., F.P., S.P., O.B., S.B.)
| | - Charlotte Romanet
- Pulmonary Hypertension Research Group, Québec Heart and Lung Institute Research Center, Quebec City, QC, Canada (S.-E.L., Y.G., T.Y., T.S., M.M., C.R., M.S., S.B.-B., A.B., C.T., A.P., R.E.K., S.M., K.Y., F.P., S.P., O.B., S.B.)
| | - Mélanie Sauvaget
- Pulmonary Hypertension Research Group, Québec Heart and Lung Institute Research Center, Quebec City, QC, Canada (S.-E.L., Y.G., T.Y., T.S., M.M., C.R., M.S., S.B.-B., A.B., C.T., A.P., R.E.K., S.M., K.Y., F.P., S.P., O.B., S.B.)
| | - Sandra Breuils-Bonnet
- Pulmonary Hypertension Research Group, Québec Heart and Lung Institute Research Center, Quebec City, QC, Canada (S.-E.L., Y.G., T.Y., T.S., M.M., C.R., M.S., S.B.-B., A.B., C.T., A.P., R.E.K., S.M., K.Y., F.P., S.P., O.B., S.B.)
| | - Alice Bourgeois
- Pulmonary Hypertension Research Group, Québec Heart and Lung Institute Research Center, Quebec City, QC, Canada (S.-E.L., Y.G., T.Y., T.S., M.M., C.R., M.S., S.B.-B., A.B., C.T., A.P., R.E.K., S.M., K.Y., F.P., S.P., O.B., S.B.)
| | - Charlie Théberge
- Pulmonary Hypertension Research Group, Québec Heart and Lung Institute Research Center, Quebec City, QC, Canada (S.-E.L., Y.G., T.Y., T.S., M.M., C.R., M.S., S.B.-B., A.B., C.T., A.P., R.E.K., S.M., K.Y., F.P., S.P., O.B., S.B.)
| | - Andréanne Pelletier
- Pulmonary Hypertension Research Group, Québec Heart and Lung Institute Research Center, Quebec City, QC, Canada (S.-E.L., Y.G., T.Y., T.S., M.M., C.R., M.S., S.B.-B., A.B., C.T., A.P., R.E.K., S.M., K.Y., F.P., S.P., O.B., S.B.)
| | - Reem El Kabbout
- Pulmonary Hypertension Research Group, Québec Heart and Lung Institute Research Center, Quebec City, QC, Canada (S.-E.L., Y.G., T.Y., T.S., M.M., C.R., M.S., S.B.-B., A.B., C.T., A.P., R.E.K., S.M., K.Y., F.P., S.P., O.B., S.B.)
| | - Sandra Martineau
- Pulmonary Hypertension Research Group, Québec Heart and Lung Institute Research Center, Quebec City, QC, Canada (S.-E.L., Y.G., T.Y., T.S., M.M., C.R., M.S., S.B.-B., A.B., C.T., A.P., R.E.K., S.M., K.Y., F.P., S.P., O.B., S.B.)
| | - Keiko Yamamoto
- Pulmonary Hypertension Research Group, Québec Heart and Lung Institute Research Center, Quebec City, QC, Canada (S.-E.L., Y.G., T.Y., T.S., M.M., C.R., M.S., S.B.-B., A.B., C.T., A.P., R.E.K., S.M., K.Y., F.P., S.P., O.B., S.B.)
| | - Muzaffar Akram
- Morphic Therapeutic, Inc, Waltham, MA (M.S.M., M.A., A.S.R., B.L., B.G., F.-Y.L., H.W., J.E.D., M.L., Q.Q., T.A.M., T.I.M.)
| | - Adrian S. Ray
- Morphic Therapeutic, Inc, Waltham, MA (M.S.M., M.A., A.S.R., B.L., B.G., F.-Y.L., H.W., J.E.D., M.L., Q.Q., T.A.M., T.I.M.)
| | - Blaise Lippa
- Morphic Therapeutic, Inc, Waltham, MA (M.S.M., M.A., A.S.R., B.L., B.G., F.-Y.L., H.W., J.E.D., M.L., Q.Q., T.A.M., T.I.M.)
| | - Bryan Goodwin
- Morphic Therapeutic, Inc, Waltham, MA (M.S.M., M.A., A.S.R., B.L., B.G., F.-Y.L., H.W., J.E.D., M.L., Q.Q., T.A.M., T.I.M.)
| | - Fu-Yang Lin
- Morphic Therapeutic, Inc, Waltham, MA (M.S.M., M.A., A.S.R., B.L., B.G., F.-Y.L., H.W., J.E.D., M.L., Q.Q., T.A.M., T.I.M.)
| | - Hua Wang
- Morphic Therapeutic, Inc, Waltham, MA (M.S.M., M.A., A.S.R., B.L., B.G., F.-Y.L., H.W., J.E.D., M.L., Q.Q., T.A.M., T.I.M.)
| | - James E. Dowling
- Morphic Therapeutic, Inc, Waltham, MA (M.S.M., M.A., A.S.R., B.L., B.G., F.-Y.L., H.W., J.E.D., M.L., Q.Q., T.A.M., T.I.M.)
| | - Min Lu
- Morphic Therapeutic, Inc, Waltham, MA (M.S.M., M.A., A.S.R., B.L., B.G., F.-Y.L., H.W., J.E.D., M.L., Q.Q., T.A.M., T.I.M.)
| | - Qi Qiao
- Morphic Therapeutic, Inc, Waltham, MA (M.S.M., M.A., A.S.R., B.L., B.G., F.-Y.L., H.W., J.E.D., M.L., Q.Q., T.A.M., T.I.M.)
| | - T. Andrew McTeague
- Morphic Therapeutic, Inc, Waltham, MA (M.S.M., M.A., A.S.R., B.L., B.G., F.-Y.L., H.W., J.E.D., M.L., Q.Q., T.A.M., T.I.M.)
| | - Terence I. Moy
- Morphic Therapeutic, Inc, Waltham, MA (M.S.M., M.A., A.S.R., B.L., B.G., F.-Y.L., H.W., J.E.D., M.L., Q.Q., T.A.M., T.I.M.)
| | - François Potus
- Pulmonary Hypertension Research Group, Québec Heart and Lung Institute Research Center, Quebec City, QC, Canada (S.-E.L., Y.G., T.Y., T.S., M.M., C.R., M.S., S.B.-B., A.B., C.T., A.P., R.E.K., S.M., K.Y., F.P., S.P., O.B., S.B.)
- Department of Medicine, Laval University, Quebec City, QC, Canada (F.P., S.P., O.B., S.B.)
| | - Steeve Provencher
- Pulmonary Hypertension Research Group, Québec Heart and Lung Institute Research Center, Quebec City, QC, Canada (S.-E.L., Y.G., T.Y., T.S., M.M., C.R., M.S., S.B.-B., A.B., C.T., A.P., R.E.K., S.M., K.Y., F.P., S.P., O.B., S.B.)
- Department of Medicine, Laval University, Quebec City, QC, Canada (F.P., S.P., O.B., S.B.)
| | - Olivier Boucherat
- Pulmonary Hypertension Research Group, Québec Heart and Lung Institute Research Center, Quebec City, QC, Canada (S.-E.L., Y.G., T.Y., T.S., M.M., C.R., M.S., S.B.-B., A.B., C.T., A.P., R.E.K., S.M., K.Y., F.P., S.P., O.B., S.B.)
- Department of Medicine, Laval University, Quebec City, QC, Canada (F.P., S.P., O.B., S.B.)
| | - Sébastien Bonnet
- Pulmonary Hypertension Research Group, Québec Heart and Lung Institute Research Center, Quebec City, QC, Canada (S.-E.L., Y.G., T.Y., T.S., M.M., C.R., M.S., S.B.-B., A.B., C.T., A.P., R.E.K., S.M., K.Y., F.P., S.P., O.B., S.B.)
- Department of Medicine, Laval University, Quebec City, QC, Canada (F.P., S.P., O.B., S.B.)
| |
Collapse
|
2
|
Valls A, Ruiz-Roldán C, Immanuel J, Alonso-Martín S, Gallardo E, Fernández-Torrón R, Bonilla M, Lersundi A, Hernández-Laín A, Domínguez-González C, Vílchez JJ, Iruzubieta P, López de Munain A, Sáenz A. The Role of Integrin β1D Mislocalization in the Pathophysiology of Calpain 3-Related Limb-Girdle Muscular Dystrophy. Cells 2025; 14:446. [PMID: 40136695 PMCID: PMC11941428 DOI: 10.3390/cells14060446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2025] [Revised: 03/06/2025] [Accepted: 03/10/2025] [Indexed: 03/27/2025] Open
Abstract
Limb-girdle muscular dystrophy R1 (LGMDR1) is characterized by progressive proximal muscle weakness due to mutations in the CAPN3 gene. Little is known about CAPN3's function in muscle, but its loss results in aberrant sarcomere formation. Human muscle structure was analyzed in this study, with observations including integrin β1D isoform (ITGβ1D) mislocalization, a lack of Talin-1 (TLN1) in the sarcolemma and the irregular expression of focal adhesion kinase (FAK) in LGMDR1 muscles, suggesting a lack of integrin activation with an altered sarcolemma, extracellular matrix (ECM) assembly and signaling pathway deregulation, which may cause frailty in LGMDR1 muscle fibers. Additionally, altered nuclear morphology, centrosome distribution and microtubule organization have been found in muscle cells derived from LGMDR1 patients.
Collapse
Affiliation(s)
- Andrea Valls
- Neuromuscular Diseases Group, Neurosciences Area, Biogipuzkoa Health Research Institute, 20014 San Sebastian, Spain
- Center for Biomedical Network Research on Neurodegenerative Diseases (CIBERNED), Spanish Ministry of Science & Innovation, Carlos III Health Institute, 28029 Madrid, Spain
| | - Cristina Ruiz-Roldán
- Neuromuscular Diseases Group, Neurosciences Area, Biogipuzkoa Health Research Institute, 20014 San Sebastian, Spain
| | - Jenita Immanuel
- Neuromuscular Diseases Group, Neurosciences Area, Biogipuzkoa Health Research Institute, 20014 San Sebastian, Spain
- Center for Biomedical Network Research on Neurodegenerative Diseases (CIBERNED), Spanish Ministry of Science & Innovation, Carlos III Health Institute, 28029 Madrid, Spain
| | - Sonia Alonso-Martín
- Center for Biomedical Network Research on Neurodegenerative Diseases (CIBERNED), Spanish Ministry of Science & Innovation, Carlos III Health Institute, 28029 Madrid, Spain
- Stem Cells and Aging Group, Bioengineering Area, Biogipuzkoa Health Research Institute, 20014 San Sebastian, Spain
| | - Eduard Gallardo
- Neuromuscular Diseases Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain
- Institut de Recerca Sant Pau, IR-SantPau, 08041 Barcelona, Spain
- Center for Biomedical Network Research on Rare Diseases (CIBERER), Spanish Ministry of Science & Innovation, Carlos III Health Institute, 28029 Madrid, Spain
| | - Roberto Fernández-Torrón
- Neuromuscular Diseases Group, Neurosciences Area, Biogipuzkoa Health Research Institute, 20014 San Sebastian, Spain
- Center for Biomedical Network Research on Neurodegenerative Diseases (CIBERNED), Spanish Ministry of Science & Innovation, Carlos III Health Institute, 28029 Madrid, Spain
- Department of Neurology, Hospital Universitario Donostia, Osakidetza, 20014 San Sebastian, Spain
| | - Mario Bonilla
- Stem Cells and Aging Group, Bioengineering Area, Biogipuzkoa Health Research Institute, 20014 San Sebastian, Spain
- Department of Traumatology, Donostialdea Integrated Health Organisation, Osakidetza, 20014 San Sebastian, Spain
| | - Ana Lersundi
- Department of Traumatology, Donostialdea Integrated Health Organisation, Osakidetza, 20014 San Sebastian, Spain
- Department of Surgery, University of the Basque Country UPV/EHU, 20014 San Sebastian, Spain
| | - Aurelio Hernández-Laín
- Department of Neuropathology, Hospital Universitario 12 de Octubre, 28041 Madrid, Spain
- Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), 28041 Madrid, Spain
- Department of Pathology, Faculty of Medicine, Complutense University of Madrid (UCM), 28040 Madrid, Spain
| | - Cristina Domínguez-González
- Center for Biomedical Network Research on Rare Diseases (CIBERER), Spanish Ministry of Science & Innovation, Carlos III Health Institute, 28029 Madrid, Spain
- Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), 28041 Madrid, Spain
- Neuromuscular Unit, Department of Neurology, Hospital 12 de Octubre, 28041 Madrid, Spain
| | - Juan Jesús Vílchez
- Center for Biomedical Network Research on Rare Diseases (CIBERER), Spanish Ministry of Science & Innovation, Carlos III Health Institute, 28029 Madrid, Spain
- Neuromuscular and Ataxias Research Group, Instituto de Investigación Sanitaria La Fe, 46026 Valencia, Spain
- Neuromuscular Diseases Unit, Neurology Department, Hospital Universitari I Politècnic La Fe, 46026 Valencia, Spain
| | - Pablo Iruzubieta
- Center for Biomedical Network Research on Neurodegenerative Diseases (CIBERNED), Spanish Ministry of Science & Innovation, Carlos III Health Institute, 28029 Madrid, Spain
- Neurogenetics, RNA Biology and Therapies Group, Neurosciences Area, Biogipuzkoa Health Research Institute, 20014 San Sebastian, Spain
- Department of Neurology and Neurosurgery, Montreal Neurological Hospital and Institute, McGill University, Montreal, QC H3A 2B4, Canada
| | - Adolfo López de Munain
- Neuromuscular Diseases Group, Neurosciences Area, Biogipuzkoa Health Research Institute, 20014 San Sebastian, Spain
- Center for Biomedical Network Research on Neurodegenerative Diseases (CIBERNED), Spanish Ministry of Science & Innovation, Carlos III Health Institute, 28029 Madrid, Spain
- Department of Neurology, Hospital Universitario Donostia, Osakidetza, 20014 San Sebastian, Spain
- Department of Neurosciences, University of the Basque Country UPV-EHU, 20014 San Sebastian, Spain
- Faculty of Medicine, University of Deusto, 48007 Bilbao, Spain
| | - Amets Sáenz
- Neuromuscular Diseases Group, Neurosciences Area, Biogipuzkoa Health Research Institute, 20014 San Sebastian, Spain
- Center for Biomedical Network Research on Neurodegenerative Diseases (CIBERNED), Spanish Ministry of Science & Innovation, Carlos III Health Institute, 28029 Madrid, Spain
| |
Collapse
|
3
|
Lemay SE, Montesinos MS, Grobs Y, Yokokawa T, Shimauchi T, Romanet C, Sauvaget M, Breuils-Bonnet S, Bourgeois A, Théberge C, Pelletier A, El Kabbout R, Martineau S, Yamamoto K, Ray AS, Lippa B, Goodwin B, Lin FY, Wang H, Dowling JE, Lu M, Qiao Q, McTeague TA, Moy TI, Potus F, Provencher S, Boucherat O, Bonnet S. Exploring Integrin α5β1 as a Potential Therapeutic Target for Pulmonary Arterial Hypertension: Insights from Comprehensive Multicenter Preclinical Studies. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.27.596052. [PMID: 38854025 PMCID: PMC11160677 DOI: 10.1101/2024.05.27.596052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
Pulmonary arterial hypertension (PAH) is characterized by obliterative vascular remodeling of the small pulmonary arteries (PA) and progressive increase in pulmonary vascular resistance (PVR) leading to right ventricular (RV) failure. Although several drugs are approved for the treatment of PAH, mortality remains high. Accumulating evidence supports a pathological function of integrins in vessel remodeling, which are gaining renewed interest as drug targets. However, their role in PAH remains largely unexplored. We found that the arginine-glycine-aspartate (RGD)-binding integrin α5β1 is upregulated in PA endothelial cells (PAEC) and PA smooth muscle cells (PASMC) from PAH patients and remodeled PAs from animal models. Blockade of the integrin α5β1 or depletion of the α5 subunit resulted in mitotic defects and inhibition of the pro-proliferative and apoptosis-resistant phenotype of PAH cells. Using a novel small molecule integrin inhibitor and neutralizing antibodies, we demonstrated that α5β1 integrin blockade attenuates pulmonary vascular remodeling and improves hemodynamics and RV function in multiple preclinical models. Our results provide converging evidence to consider α5β1 integrin inhibition as a promising therapy for pulmonary hypertension. One sentence summary The α5β1 integrin plays a crucial role in pulmonary vascular remodeling.
Collapse
|
4
|
Rani B, Gupta DK, Johansson S, Kamranvar SA. Contribution of integrin adhesion to cytokinetic abscission and genomic integrity. Front Cell Dev Biol 2022; 10:1048717. [PMID: 36578785 PMCID: PMC9791049 DOI: 10.3389/fcell.2022.1048717] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 11/28/2022] [Indexed: 12/14/2022] Open
Abstract
Recent research shows that integrin-mediated adhesion contributes to the regulation of cell division at two key steps: the formation of the mitotic spindle at the mitotic entry and the final cytokinetic abscission at the mitotic exit. Failure in either of these processes will have a direct impact on the other in each round of the cell cycle and on the genomic integrity. This review aims to present how integrin signals are involved at these cell cycle stages under normal conditions and some safety mechanisms that may counteract the generation of aneuploid cells in cases of defective integrin signals.
Collapse
Affiliation(s)
- Bhavna Rani
- Department of Medical Biochemistry and Microbiology (IMBIM), Biomedical Center, Uppsala University, Uppsala, Sweden
| | - Deepesh K. Gupta
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, United States
| | - Staffan Johansson
- Department of Medical Biochemistry and Microbiology (IMBIM), Biomedical Center, Uppsala University, Uppsala, Sweden,*Correspondence: Staffan Johansson, ; Siamak A. Kamranvar,
| | - Siamak A. Kamranvar
- Department of Medical Biochemistry and Microbiology (IMBIM), Biomedical Center, Uppsala University, Uppsala, Sweden,*Correspondence: Staffan Johansson, ; Siamak A. Kamranvar,
| |
Collapse
|
5
|
Gugnoni M, Manzotti G, Vitale E, Sauta E, Torricelli F, Reggiani F, Pistoni M, Piana S, Ciarrocchi A. OVOL2 impairs RHO GTPase signaling to restrain mitosis and aggressiveness of Anaplastic Thyroid Cancer. J Exp Clin Cancer Res 2022; 41:108. [PMID: 35337349 PMCID: PMC8957195 DOI: 10.1186/s13046-022-02316-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 03/08/2022] [Indexed: 11/10/2022] Open
Abstract
Background Anaplastic Thyroid Cancer (ATC) is an undifferentiated and aggressive tumor that often originates from well-Differentiated Thyroid Carcinoma (DTC) through a trans-differentiation process. Epithelial-to-Mesenchymal Transition (EMT) is recognized as one of the major players of this process. OVOL2 is a transcription factor (TF) that promotes epithelial differentiation and restrains EMT during embryonic development. OVOL2 loss in some types of cancers is linked to aggressiveness and poor prognosis. Here, we aim to clarify the unexplored role of OVOL2 in ATC. Methods Gene expression analysis in thyroid cancer patients and cell lines showed that OVOL2 is mainly associated with epithelial features and its expression is deeply impaired in ATC. To assess OVOL2 function, we established an OVOL2-overexpression model in ATC cell lines and evaluated its effects by analyzing gene expression, proliferation, invasion and migration abilities, cell cycle, specific protein localization through immunofluorescence staining. RNA-seq profiling showed that OVOL2 controls a complex network of genes converging on cell cycle and mitosis regulation and Chromatin Immunoprecipitation identified new OVOL2 target genes. Results Coherently with its reported function, OVOL2 re-expression restrained EMT and aggressiveness in ATC cells. Unexpectedly, we observed that it caused G2/M block, a consequent reduction in cell proliferation and an increase in cell death. This phenotype was associated to generalized abnormalities in the mitotic spindle structure and cytoskeletal organization. By RNA-seq experiments, we showed that many pathways related to cytoskeleton and migration, cell cycle and mitosis are profoundly affected by OVOL2 expression, in particular the RHO-GTPase pathway resulted as the most interesting. We demonstrated that RHO GTPase pathway is the central hub of OVOL2-mediated program in ATC and that OVOL2 transcriptionally inhibits RhoU and RhoJ. Silencing of RhoU recapitulated the OVOL2-driven phenotype pointing to this protein as a crucial target of OVOL2 in ATC. Conclusions Collectively, these data describe the role of OVOL2 in ATC and uncover a novel function of this TF in inhibiting the RHO GTPase pathway interlacing its effects on EMT, cytoskeleton dynamics and mitosis. Supplementary Information The online version contains supplementary material available at 10.1186/s13046-022-02316-2.
Collapse
|
6
|
Gumina DL, Ji S, Flockton A, McPeak K, Stich D, Moldovan R, Su EJ. Dysregulation of integrin αvβ3 and α5β1 impedes migration of placental endothelial cells in fetal growth restriction. Development 2022; 149:dev200717. [PMID: 36193846 PMCID: PMC9641665 DOI: 10.1242/dev.200717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 08/23/2022] [Indexed: 11/06/2022]
Abstract
Placentas from pregnancies complicated by severe early-onset fetal growth restriction (FGR) exhibit diminished vascular development mediated by impaired angiogenesis, but underlying mechanisms remain unknown. In this study, we show that FGR endothelial cells demonstrate inherently reduced migratory capacity despite the presence of fibronectin, a matrix protein abundant in placental stroma that displays abnormal organization in FGR placentas. Thus, we hypothesized that aberrant endothelial-fibronectin interactions in FGR are a key mechanism underlying impaired FGR endothelial migration. Using human fetoplacental endothelial cells isolated from uncomplicated term control and FGR pregnancies, we assessed integrin α5β1 and αvβ3 regulation during cell migration. We show that endothelial integrin α5β1 and αvβ3 interactions with fibronectin are required for migration and that FGR endothelial cells responded differentially to integrin inhibition, indicating integrin dysregulation in FGR. Whole-cell expression was not different between groups. However, there were significantly more integrins in focal adhesions and reduced intracellular trafficking in FGR. These newly identified changes in FGR endothelial cellular processes represent previously unidentified mechanisms contributing to persistent angiogenic deficiencies in FGR.
Collapse
Affiliation(s)
- Diane L. Gumina
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Shuhan Ji
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Amanda Flockton
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Kathryn McPeak
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Dominik Stich
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Radu Moldovan
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Emily J. Su
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado School of Medicine, Aurora, CO 80045, USA
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, University of Colorado School of Medicine, Aurora, CO 80045, USA
| |
Collapse
|
7
|
Cell Cycle Regulation by Integrin-Mediated Adhesion. Cells 2022; 11:cells11162521. [PMID: 36010598 PMCID: PMC9406542 DOI: 10.3390/cells11162521] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 08/08/2022] [Accepted: 08/11/2022] [Indexed: 11/23/2022] Open
Abstract
Cell cycle and cell adhesion are two interdependent cellular processes regulating each other, reciprocally, in every cell cycle phase. The cell adhesion to the extracellular matrix (ECM) via integrin receptors triggers signaling pathways required for the cell cycle progression; the passage from the G1 to S phase and the completion of cytokinesis are the best-understood events. Growing evidence, however, suggests more adhesion-dependent regulatory aspects of the cell cycle, particularly during G2 to M transition and early mitosis. Conversely, the cell cycle machinery regulates cell adhesion in manners recently shown driven mainly by cyclin-dependent kinase 1 (CDK1). This review summarizes the recent findings regarding the role of integrin-mediated cell adhesion and its downstream signaling components in regulating the cell cycle, emphasizing the cell cycle progression through the G2 and early M phases. Further investigations are required to raise our knowledge about the molecular mechanisms of crosstalk between cell adhesion and the cell cycle in detail.
Collapse
|
8
|
Integrin-Mediated Adhesion Promotes Centrosome Separation in Early Mitosis. Cells 2022; 11:cells11081360. [PMID: 35456039 PMCID: PMC9030014 DOI: 10.3390/cells11081360] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 10/29/2021] [Accepted: 04/11/2022] [Indexed: 01/17/2023] Open
Abstract
Integrin-mediated adhesion to the extracellular matrix is a key regulator of the cell cycle, as demonstrated for the passage of the G1/S checkpoint and the completion of cytokinetic abscission. Here, integrin-dependent regulation of the cell cycle in G2 and early M phases was investigated. The progression through the G2 and M phases was monitored by live-cell imaging and immunofluorescence staining in adherent and non-adherent fibroblast cells. Non-adherent cells, as well as adherent cells lacking FAK activity due to suppressed expression or pharmacological inhibition, exhibited a prolonged G2 phase and severely defect centrosome separation, resulting in delayed progress through the early mitotic stages. The activation of the critical mitotic regulator PLK1 and its indirect target Eg5, a kinesin-family motor protein driving the centrosome separation, were reduced in the cells lacking FAK activity. Furthermore, the absence of integrin adhesion or FAK activity destabilized the structural integrity of centrosomes and often caused detachment of pericentriolar material from the centrioles. These data identify a novel adhesion-dependent mechanism by which integrins via FAK and PLK1 contribute to the regulation of the cell cycle in the G2 and early M phases, and to the maintenance of genome integrity.
Collapse
|
9
|
Cell stretchers and the LINC complex in mechanotransduction. Arch Biochem Biophys 2021; 702:108829. [PMID: 33716002 DOI: 10.1016/j.abb.2021.108829] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 02/23/2021] [Accepted: 03/07/2021] [Indexed: 02/07/2023]
Abstract
How cells respond to mechanical forces from the surrounding environment is critical for cell survival and function. The LINC complex is a central component in the mechanotransduction pathway that transmits mechanical information from the cell surface to the nucleus. Through LINC complex functionality, the nucleus is able to respond to mechanical stress by altering nuclear structure, chromatin organization, and gene expression. The use of specialized devices that apply mechanical strain to cells have been central to investigating how mechanotransduction occurs, how cells respond to mechanical stress, and the role of the LINC complexes in these processes. A large variety of designs have been reported for these devices, with the most common type being cell stretchers. Here we highlight some of the salient features of cell stretchers and suggest some key parameters that should be considered when using these devices. We provide a brief overview of how the LINC complexes contribute to the cellular responses to mechanical strain. And finally, we suggest that stretchers may be a useful tool to study aging.
Collapse
|
10
|
Tan HF, Tan SM. The focal adhesion protein kindlin-2 controls mitotic spindle assembly by inhibiting histone deacetylase 6 and maintaining α-tubulin acetylation. J Biol Chem 2020; 295:5928-5943. [PMID: 32169902 DOI: 10.1074/jbc.ra120.012954] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 03/09/2020] [Indexed: 02/06/2023] Open
Abstract
Kindlins are focal adhesion proteins that regulate integrin activation and outside-in signaling. The kindlin family consists of three members, kindlin-1, -2, and -3. Kindlin-2 is widely expressed in multiple cell types, except those from the hematopoietic lineage. A previous study has reported that the Drosophila Fit1 protein (an ortholog of kindlin-2) prevents abnormal spindle assembly; however, the mechanism remains unknown. Here, we show that kindlin-2 maintains spindle integrity in mitotic human cells. The human neuroblastoma SH-SY5Y cell line expresses only kindlin-2, and we found that when SH-SY5Y cells are depleted of kindlin-2, they exhibit pronounced spindle abnormalities and delayed mitosis. Of note, acetylation of α-tubulin, which maintains microtubule flexibility and stability, was diminished in the kindlin-2-depleted cells. Mechanistically, we found that kindlin-2 maintains α-tubulin acetylation by inhibiting the microtubule-associated deacetylase histone deacetylase 6 (HDAC6) via a signaling pathway involving AKT Ser/Thr kinase (AKT)/glycogen synthase kinase 3β (GSK3β) or paxillin. We also provide evidence that prolonged hypoxia down-regulates kindlin-2 expression, leading to spindle abnormalities not only in the SH-SY5Y cell line, but also cell lines derived from colon and breast tissues. The findings of our study highlight that kindlin-2 regulates mitotic spindle assembly and that this process is perturbed in cancer cells in a hypoxic environment.
Collapse
Affiliation(s)
- Hui-Foon Tan
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore
| | - Suet-Mien Tan
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore.
| |
Collapse
|
11
|
Jones MC, Zha J, Humphries MJ. Connections between the cell cycle, cell adhesion and the cytoskeleton. Philos Trans R Soc Lond B Biol Sci 2019; 374:20180227. [PMID: 31431178 PMCID: PMC6627016 DOI: 10.1098/rstb.2018.0227] [Citation(s) in RCA: 101] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/26/2018] [Indexed: 12/18/2022] Open
Abstract
Cell division, the purpose of which is to enable cell replication, and in particular to distribute complete, accurate copies of genetic material to daughter cells, is essential for the propagation of life. At a morphological level, division not only necessitates duplication of cellular structures, but it also relies on polar segregation of this material followed by physical scission of the parent cell. For these fundamental changes in cell shape and positioning to be achieved, mechanisms are required to link the cell cycle to the modulation of cytoarchitecture. Outside of mitosis, the three main cytoskeletal networks not only endow cells with a physical cytoplasmic skeleton, but they also provide a mechanism for spatio-temporal sensing via integrin-associated adhesion complexes and site-directed delivery of cargoes. During mitosis, some interphase functions are retained, but the architecture of the cytoskeleton changes dramatically, and there is a need to generate a mitotic spindle for chromosome segregation. An economical solution is to re-use existing cytoskeletal molecules: transcellular actin stress fibres remodel to create a rigid cortex and a cytokinetic furrow, while unipolar radial microtubules become the primary components of the bipolar spindle. This remodelling implies the existence of specific mechanisms that link the cell-cycle machinery to the control of adhesion and the cytoskeleton. In this article, we review the intimate three-way connection between microenvironmental sensing, adhesion signalling and cell proliferation, particularly in the contexts of normal growth control and aberrant tumour progression. As the morphological changes that occur during mitosis are ancient, the mechanisms linking the cell cycle to the cytoskeleton/adhesion signalling network are likely to be primordial in nature and we discuss recent advances that have elucidated elements of this link. A particular focus is the connection between CDK1 and cell adhesion. This article is part of a discussion meeting issue 'Forces in cancer: interdisciplinary approaches in tumour mechanobiology'.
Collapse
Affiliation(s)
| | | | - Martin J. Humphries
- Wellcome Trust Centre for Cell-Matrix Research, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PT, UK
| |
Collapse
|
12
|
Bazzoun D, Adissu HA, Wang L, Urazaev A, Tenvooren I, Fostok SF, Chittiboyina S, Sturgis J, Hodges K, Chandramouly G, Vidi PA, Talhouk RS, Lelièvre SA. Connexin 43 maintains tissue polarity and regulates mitotic spindle orientation in the breast epithelium. J Cell Sci 2019; 132:jcs.223313. [PMID: 30992345 DOI: 10.1242/jcs.223313] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Accepted: 04/08/2019] [Indexed: 12/11/2022] Open
Abstract
Cell-cell communication is essential for tissue homeostasis, but its contribution to disease prevention remains to be understood. We demonstrate the involvement of connexin 43 (Cx43, also known as GJA1) and related gap junction in epithelial homeostasis, illustrated by polarity-mediated cell cycle entry and mitotic spindle orientation (MSO). Cx43 localization is restricted to the apicolateral membrane of phenotypically normal breast luminal epithelial cells in 3D culture and in vivo Chemically induced blockade of gap junction intercellular communication (GJIC), as well as the absence of Cx43, disrupt the apicolateral distribution of polarity determinant tight junction marker ZO-1 (also known as TJP1) and lead to random MSO and cell multilayering. Induced expression of Cx43 in cells that normally lack this protein reestablishes polarity and proper MSO in 3D culture. Cx43-directed MSO implicates PI3K-aPKC signaling, and Cx43 co-precipitates with signaling node proteins β-catenin (CTNNB1) and ZO-2 (also known as TJP2) in the polarized epithelium. The distribution of Cx43 is altered by pro-inflammatory breast cancer risk factors such as leptin and high-fat diet, as shown in cell culture and on tissue biopsy sections. The control of polarity-mediated quiescence and MSO may contribute to the tumor-suppressive role of Cx43.
Collapse
Affiliation(s)
- D Bazzoun
- Basic Medical Sciences, Purdue University, West Lafayette, IN 47907, USA.,Biology Department, Faculty of Arts and Sciences, American University of Beirut, 11-0236 Beirut, Lebanon
| | - H A Adissu
- Basic Medical Sciences, Purdue University, West Lafayette, IN 47907, USA
| | - L Wang
- Basic Medical Sciences, Purdue University, West Lafayette, IN 47907, USA
| | - A Urazaev
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907, USA
| | - I Tenvooren
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - S F Fostok
- Biology Department, Faculty of Arts and Sciences, American University of Beirut, 11-0236 Beirut, Lebanon
| | - S Chittiboyina
- Basic Medical Sciences, Purdue University, West Lafayette, IN 47907, USA
| | - J Sturgis
- Basic Medical Sciences, Purdue University, West Lafayette, IN 47907, USA
| | - K Hodges
- Basic Medical Sciences, Purdue University, West Lafayette, IN 47907, USA
| | - G Chandramouly
- Basic Medical Sciences, Purdue University, West Lafayette, IN 47907, USA
| | - P-A Vidi
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - R S Talhouk
- Biology Department, Faculty of Arts and Sciences, American University of Beirut, 11-0236 Beirut, Lebanon
| | - S A Lelièvre
- Basic Medical Sciences, Purdue University, West Lafayette, IN 47907, USA .,Purdue University Center for Cancer Research, Purdue University, West Lafayette, IN 47907, USA
| |
Collapse
|
13
|
Humphries JD, Chastney MR, Askari JA, Humphries MJ. Signal transduction via integrin adhesion complexes. Curr Opin Cell Biol 2019; 56:14-21. [PMID: 30195153 DOI: 10.1016/j.ceb.2018.08.004] [Citation(s) in RCA: 211] [Impact Index Per Article: 35.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Revised: 08/13/2018] [Accepted: 08/14/2018] [Indexed: 12/19/2022]
Abstract
Integrin adhesion complexes (IACs) have evolved over millions of years to integrate metazoan cells physically with their microenvironment. It is presumed that the simultaneous interaction of thousands of integrin receptors to binding sites in anisotropic extracellular matrix (ECM) networks enables cells to assemble a topological description of the chemical and mechanical properties of their surroundings. This information is then converted into intracellular signals that influence cell positioning, differentiation and growth, but may also influence other fundamental processes, such as protein synthesis and energy regulation. In this way, changes in the microenvironment can influence all aspects of cell phenotype. Current concepts envisage cell fate decisions being controlled by the integrated signalling output of myriad receptor clusters, but the mechanisms are not understood. Analyses of the adhesome, the complement of proteins attracted to the vicinity of IACs, are now providing insights into some of the primordial links connecting these processes. This article reviews recent advances in our understanding of the composition of IACs, the mechanisms used to transduce signals through these junctions, and the links between IACs and cell phenotype.
Collapse
Affiliation(s)
- Jonathan D Humphries
- Wellcome Trust Centre for Cell-Matrix Research, Faculty of Biology, Medicine & Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PT, UK
| | - Megan R Chastney
- Wellcome Trust Centre for Cell-Matrix Research, Faculty of Biology, Medicine & Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PT, UK
| | - Janet A Askari
- Wellcome Trust Centre for Cell-Matrix Research, Faculty of Biology, Medicine & Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PT, UK
| | - Martin J Humphries
- Wellcome Trust Centre for Cell-Matrix Research, Faculty of Biology, Medicine & Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PT, UK.
| |
Collapse
|
14
|
LaFlamme SE, Mathew-Steiner S, Singh N, Colello-Borges D, Nieves B. Integrin and microtubule crosstalk in the regulation of cellular processes. Cell Mol Life Sci 2018; 75:4177-4185. [PMID: 30206641 PMCID: PMC6182340 DOI: 10.1007/s00018-018-2913-x] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2018] [Revised: 08/14/2018] [Accepted: 08/27/2018] [Indexed: 11/25/2022]
Abstract
Integrins engage components of the extracellular matrix, and in collaboration with other receptors, regulate signaling cascades that impact cell behavior in part by modulating the cell's cytoskeleton. Integrins have long been known to function together with the actin cytoskeleton to promote cell adhesion, migration, and invasion, and with the intermediate filament cytoskeleton to mediate the strong adhesion needed for the maintenance and integrity of epithelial tissues. Recent studies have shed light on the crosstalk between integrin and the microtubule cytoskeleton. Integrins promote microtubule nucleation, growth, and stabilization at the cell cortex, whereas microtubules regulate integrin activity and remodeling of adhesion sites. Integrin-dependent stabilization of microtubules at the cell cortex is critical to the establishment of apical-basal polarity required for the formation of epithelial tissues. During cell migration, integrin-dependent microtubule stabilization contributes to front-rear polarity, whereas microtubules promote the turnover of integrin-mediated adhesions. This review focuses on this interdependent relationship and its impact on cell behavior and function.
Collapse
Affiliation(s)
- Susan E LaFlamme
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, 47 New Scotland Avenue, Albany, NY, 12208, USA.
| | - Shomita Mathew-Steiner
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, 47 New Scotland Avenue, Albany, NY, 12208, USA
- Indiana University, 975 W. Walnut Street, Indianapolis, IN, 46202, USA
| | - Neetu Singh
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, 47 New Scotland Avenue, Albany, NY, 12208, USA
| | - Diane Colello-Borges
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, 47 New Scotland Avenue, Albany, NY, 12208, USA
| | - Bethsaida Nieves
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, 47 New Scotland Avenue, Albany, NY, 12208, USA
| |
Collapse
|
15
|
Lock JG, Jones MC, Askari JA, Gong X, Oddone A, Olofsson H, Göransson S, Lakadamyali M, Humphries MJ, Strömblad S. Reticular adhesions are a distinct class of cell-matrix adhesions that mediate attachment during mitosis. Nat Cell Biol 2018; 20:1290-1302. [PMID: 30361699 DOI: 10.1038/s41556-018-0220-2] [Citation(s) in RCA: 98] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Accepted: 09/21/2018] [Indexed: 12/13/2022]
Abstract
Adhesion to the extracellular matrix persists during mitosis in most cell types. However, while classical adhesion complexes, such as focal adhesions, do and must disassemble to enable mitotic rounding, the mechanisms of residual mitotic cell-extracellular matrix adhesion remain undefined. Here, we identify 'reticular adhesions', a class of adhesion complex that is mediated by integrin αvβ5, formed during interphase, and preserved at cell-extracellular matrix attachment sites throughout cell division. Consistent with this role, integrin β5 depletion perturbs mitosis and disrupts spatial memory transmission between cell generations. Reticular adhesions are morphologically and dynamically distinct from classical focal adhesions. Mass spectrometry defines their unique composition, enriched in phosphatidylinositol-4,5-bisphosphate (PtdIns(4,5)P2)-binding proteins but lacking virtually all consensus adhesome components. Indeed, reticular adhesions are promoted by PtdIns(4,5)P2, and form independently of talin and F-actin. The distinct characteristics of reticular adhesions provide a solution to the problem of maintaining cell-extracellular matrix attachment during mitotic rounding and division.
Collapse
Affiliation(s)
- John G Lock
- Department of Pathology, School of Medical Sciences, University of New South Wales, Sydney, Australia.
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden.
| | - Matthew C Jones
- Wellcome Trust Centre for Cell-Matrix Research, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Janet A Askari
- Wellcome Trust Centre for Cell-Matrix Research, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Xiaowei Gong
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
| | - Anna Oddone
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain
- ICFO, Institut de Ciencies Fotoniques, Mediterranean Technology Park, The Barcelona Institute of Science and Technology, Castelldefels, Barcelona, Spain
| | - Helene Olofsson
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
| | - Sara Göransson
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
| | - Melike Lakadamyali
- ICFO, Institut de Ciencies Fotoniques, Mediterranean Technology Park, The Barcelona Institute of Science and Technology, Castelldefels, Barcelona, Spain
- Perelman School of Medicine, Department of Physiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Martin J Humphries
- Wellcome Trust Centre for Cell-Matrix Research, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Staffan Strömblad
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden.
| |
Collapse
|
16
|
Jones MC, Askari JA, Humphries JD, Humphries MJ. Cell adhesion is regulated by CDK1 during the cell cycle. J Cell Biol 2018; 217:3203-3218. [PMID: 29930204 PMCID: PMC6122981 DOI: 10.1083/jcb.201802088] [Citation(s) in RCA: 96] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Revised: 05/08/2018] [Accepted: 05/29/2018] [Indexed: 12/15/2022] Open
Abstract
In most tissues, anchorage-dependent growth and cell cycle progression are dependent on cells engaging extracellular matrices (ECMs) via integrin-receptor adhesion complexes. In a highly conserved manner, cells disassemble adhesion complexes, round up, and retract from their surroundings before division, suggestive of a primordial link between the cell cycle machinery and the regulation of cell adhesion to the ECM. In this study, we demonstrate that cyclin-dependent kinase 1 (CDK1) mediates this link. CDK1, in complex with cyclin A2, promotes adhesion complex and actin cytoskeleton organization during interphase and mediates a large increase in adhesion complex area as cells transition from G1 into S. Adhesion complex area decreases in G2, and disassembly occurs several hours before mitosis. This loss requires elevated cyclin B1 levels and is caused by inhibitory phosphorylation of CDK1-cyclin complexes. The inactivation of CDK1 is therefore the trigger that initiates remodeling of adhesion complexes and the actin cytoskeleton in preparation for rapid entry into mitosis.
Collapse
Affiliation(s)
- Matthew C Jones
- Wellcome Trust Centre for Cell-Matrix Research, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, England, UK
| | - Janet A Askari
- Wellcome Trust Centre for Cell-Matrix Research, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, England, UK
| | - Jonathan D Humphries
- Wellcome Trust Centre for Cell-Matrix Research, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, England, UK
| | - Martin J Humphries
- Wellcome Trust Centre for Cell-Matrix Research, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, England, UK
| |
Collapse
|
17
|
Laminin β2 Chain Regulates Retinal Progenitor Cell Mitotic Spindle Orientation via Dystroglycan. J Neurosci 2018; 38:5996-6010. [PMID: 29853630 DOI: 10.1523/jneurosci.0551-18.2018] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Revised: 05/10/2018] [Accepted: 05/18/2018] [Indexed: 01/27/2023] Open
Abstract
Vertebrate retinal development follows a pattern during which retinal progenitor cells (RPCs) give rise to all retinal cell types in a highly conserved temporal sequence. RPC proliferation and cell cycle exit are tightly coordinated to ensure proper and timely production of each of the retinal cell types. Extracellular matrix (ECM) plays an important role in eye development, influencing RPC proliferation and differentiation. In this study, we demonstrate that laminins, key ECM components, in the inner limiting membrane, control mitotic spindle orientation by providing environmental cues to the RPCs. In vivo deletion of laminin β2 in mice of both sexes results in a loss RPC basal processes and contact with the ECM, leading to a shift of the mitotic spindle pole orientation toward asymmetric cell divisions. This leads to decreased proliferation and premature RPC pool depletion, resulting in overproduction of rod photoreceptors at the expense of bipolar cells and Müller glia. Moreover, we show that deletion of laminin β2 leads to disruption and mislocalization of its receptors: dystroglycan and β1-integrin. Addition of exogenous β2-containing laminins to laminin β2-deficient retinal explants stabilizes the RPC basal processes and directs their mitotic spindle orientation toward symmetric divisions, leading to increased RPC proliferation, as well as restores proper receptor localization at the retinal surface. Finally, functional blocking of dystroglycan in wild-type retinal explants phenocopies laminin β2 ablation. Our data suggest that dystroglycan-mediated signaling between RPCs and the ECM is of key importance in controlling critical developmental events during retinogenesis.SIGNIFICANCE STATEMENT The mechanisms governing retinogenesis are subject to both intrinsic and extrinsic signaling cues. Although the role of intrinsic signaling has been the subject of many studies, our understanding of the role of the microenvironment in retinal development remains unclear. Using a combination of in vivo and ex vivo approaches, we demonstrate that laminins, key extracellular matrix components, provide signaling cues that control retinal progenitor cell attachment to the basement membrane, mitotic axis, proliferation, and fate adoption. Moreover, we identify, for the first time, dystroglycan as the receptor responsible for directing retinal progenitor cell mitotic spindle orientation. Our data suggest a mechanism where dystroglycan-mediated signaling between the cell and the extracellular matrix controls the proliferative potential of progenitors in the developing CNS.
Collapse
|
18
|
De Santis Puzzonia M, Gonzalez L, Ascenzi S, Cundari E, Degrassi F. Tetraploid cells produced by absence of substrate adhesion during cytokinesis are limited in their proliferation and enter senescence after DNA replication. Cell Cycle 2016; 15:274-82. [PMID: 26693937 DOI: 10.1080/15384101.2015.1127469] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
Abstract
Tetraploidy has been proposed as an intermediate state in neoplastic transformation due to the intrinsic chromosome instability of tetraploid cells. Despite the identification of p53 as a major factor in growth arrest of tetraploid cells, it is still unclear whether the p53-dependent mechanism for proliferation restriction is intrinsic to the tetraploid status or dependent on the origin of tetraploidy. Substrate adherence is fundamental for cytokinesis completion in adherent untransformed cells. Here we show that untransformed fibroblast cells undergoing mitosis in suspension produce binucleated tetraploid cells due to defective cleavage furrow constriction that leads to incomplete cell abscission. Binucleated cells obtained after loss of substrate adhesion maintain an inactive p53 status and are able to progress into G1 and S phase. However, binucleated cells arrest in G2, accumulate p53 and are not able to enter mitosis as no tetraploid metaphases were recorded after one cell cycle time. In contrast, tetraploid metaphases were found following pharmacological inhibition of Chk1 kinase, suggesting the involvement of the ATR/Chk1 pathway in the G2 arrest of binucleated cells. Interestingly, after persistence in the G2 phase of the cell cycle, a large fraction of binucleated cells become senescent. These findings identify a new pathway of proliferation restriction for tetraploid untransformed cells that seems to be specific for loss of adhesion-dependent cytokinesis failure. This involves Chk1 and p53 activation during G2. Inhibition of growth and entrance into senescence after cytokinesis in suspension may represent an important mechanism to control tumor growth. In fact, anchorage independent growth is a hallmark of cancer and it has been demonstrated that binucleated transformed cells can enter a cycle of anchorage independent growth.
Collapse
Affiliation(s)
- Marco De Santis Puzzonia
- a Institute of Molecular Biology and Pathology, CNR National Research Council , Rome , Italy.,b Sapienza University , Department of Cellular Biotechnology and Hematology , Rome , Italy
| | - Laetitia Gonzalez
- c Vrije Universiteit Brussel, Laboratory of Cell Genetics , Brussels , Belgium
| | - Sonia Ascenzi
- a Institute of Molecular Biology and Pathology, CNR National Research Council , Rome , Italy
| | - Enrico Cundari
- a Institute of Molecular Biology and Pathology, CNR National Research Council , Rome , Italy
| | - Francesca Degrassi
- a Institute of Molecular Biology and Pathology, CNR National Research Council , Rome , Italy
| |
Collapse
|
19
|
Kamranvar SA, Gupta DK, Huang Y, Gupta RK, Johansson S. Integrin signaling via FAK-Src controls cytokinetic abscission by decelerating PLK1 degradation and subsequent recruitment of CEP55 at the midbody. Oncotarget 2016; 7:30820-30. [PMID: 27127172 PMCID: PMC5058720 DOI: 10.18632/oncotarget.9003] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2015] [Accepted: 04/09/2016] [Indexed: 01/08/2023] Open
Abstract
Adhesion to extracellular matrix is required for cell cycle progression through the G1 phase and for the completion of cytokinesis in normal adherent cells. Cancer cells acquire the ability to proliferate anchorage-independently, a characteristic feature of malignantly transformed cells. However, the molecular mechanisms underlying this escape of the normal control mechanisms remain unclear. The current study aimed to identify adhesion-induced reactions regulating the cytokinesis of non-transformed human fibroblasts.The adhesion-dependent control of cytokinesis was found to occur at a late stage close to the abscission, during which the endosomal sorting complex required for transport (ESCRT) severs the thin intercellular bridge connecting two nascent daughter cells. CEP55, a key protein involved in the abscission process, was localized at the midbody in both adherent and non-adherent fibroblasts, but it was unable to efficiently recruit ALIX, TSG101, and consequently the ESCRT-III subunit CHMP4B was missing in the non-adherent cells. PLK1, a kinase that prevents premature recruitment of CEP55 to the midbody, disappeared from this site more rapidly in the non-adherent cells. A FAK-Src signaling pathway downstream of integrin-mediated cell adhesion was found to decelerate both PLK1 degradation and CEP55 accumulation at the midbody. These data identify the regulation of PLK1 and CEP55 as steps where integrins exert control over the cytokinetic abscission.
Collapse
Affiliation(s)
- Siamak A. Kamranvar
- Department of Medical Biochemistry and Microbiology, Biomedical Center, Uppsala University, Uppsala, Sweden
| | - Deepesh Kumar Gupta
- Department of Medical Biochemistry and Microbiology, Biomedical Center, Uppsala University, Uppsala, Sweden
| | - Ying Huang
- Department of Medical Biochemistry and Microbiology, Biomedical Center, Uppsala University, Uppsala, Sweden
| | - Rajesh Kumar Gupta
- Department of Immunology, Genetics and Pathology, The Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Staffan Johansson
- Department of Medical Biochemistry and Microbiology, Biomedical Center, Uppsala University, Uppsala, Sweden
| |
Collapse
|
20
|
Hoon JL, Tan MH, Koh CG. The Regulation of Cellular Responses to Mechanical Cues by Rho GTPases. Cells 2016; 5:cells5020017. [PMID: 27058559 PMCID: PMC4931666 DOI: 10.3390/cells5020017] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Revised: 03/29/2016] [Accepted: 03/30/2016] [Indexed: 12/21/2022] Open
Abstract
The Rho GTPases regulate many cellular signaling cascades that modulate cell motility, migration, morphology and cell division. A large body of work has now delineated the biochemical cues and pathways, which stimulate the GTPases and their downstream effectors. However, cells also respond exquisitely to biophysical and mechanical cues such as stiffness and topography of the extracellular matrix that profoundly influence cell migration, proliferation and differentiation. As these cellular responses are mediated by the actin cytoskeleton, an involvement of Rho GTPases in the transduction of such cues is not unexpected. In this review, we discuss an emerging role of Rho GTPase proteins in the regulation of the responses elicited by biophysical and mechanical stimuli.
Collapse
Affiliation(s)
- Jing Ling Hoon
- School of Biological Sciences, Nanyang Technological University, Singapore 637551, Singapore.
| | - Mei Hua Tan
- School of Biological Sciences, Nanyang Technological University, Singapore 637551, Singapore.
| | - Cheng-Gee Koh
- School of Biological Sciences, Nanyang Technological University, Singapore 637551, Singapore.
- Mechanobiology Institute, Singapore 117411, Singapore.
| |
Collapse
|
21
|
Sambandamoorthy S, Mathew-Steiner S, Varney S, Zuidema JM, Gilbert RJ, Van De Water L, LaFlamme SE. Matrix compliance and the regulation of cytokinesis. Biol Open 2015; 4:885-92. [PMID: 26002930 PMCID: PMC4571092 DOI: 10.1242/bio.011825] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Integrin-mediated cell adhesion to the ECM regulates many physiological processes in part by controlling cell proliferation. It is well established that many normal cells require integrin-mediated adhesion to enter S phase of the cell cycle. Recent evidence indicates that integrins also regulate cytokinesis. Mechanical properties of the ECM can dictate entry into S phase; however, it is not known whether they also can affect the successful completion of cell division. To address this issue, we modulated substrate compliance using fibronectin-coated acrylamide-based hydrogels. Soft and hard substrates were generated with approximate elastic moduli of 1600 and 34,000 Pascals (Pa) respectively. Our results indicate that dermal fibroblasts successfully complete cytokinesis on hard substrates, whereas on soft substrates, a significant number fail and become binucleated. Cytokinesis failure occurs at a step following the formation of the intercellular bridge connecting presumptive daughter cells, suggesting a defect in abscission. Like dermal fibroblasts, mesenchymal stem cells require cell-matrix adhesion for successful cytokinesis. However, in contrast to dermal fibroblasts, they are able to complete cytokinesis on both hard and soft substrates. These results indicate that matrix stiffness regulates the successful completion of cytokinesis, and does so in a cell-type specific manner. To our knowledge, our study is the first to demonstrate that matrix stiffness can affect cytokinesis. Understanding the cell-type specific contribution of matrix compliance to the regulation of cytokinesis will provide new insights important for development, as well as tissue homeostasis and regeneration.
Collapse
Affiliation(s)
| | - Shomita Mathew-Steiner
- Center for Cell Biology and Cancer Research, Albany Medical College, Albany, NY 12208, USA
| | - Scott Varney
- Center for Cell Biology and Cancer Research, Albany Medical College, Albany, NY 12208, USA
| | - Jonathan M Zuidema
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
| | - Ryan J Gilbert
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
| | | | - Susan E LaFlamme
- Center for Cell Biology and Cancer Research, Albany Medical College, Albany, NY 12208, USA
| |
Collapse
|
22
|
Brownlow N, Pike T, Crossland V, Claus J, Parker P. Regulation of the cytokinesis cleavage furrow by PKCε. Biochem Soc Trans 2014; 42:1534-7. [PMID: 25399566 DOI: 10.1042/bst20140240] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Cytokinesis is the final act of the cell cycle where the replicated DNA and cellular contents are finally split into two daughter cells. This process is very tightly controlled as DNA segregation errors and cytokinesis failure is commonly associated with aneuploidy and aggressive tumours. Protein kinase Cε (PKCε) is a lipid-activated serine/threonine kinase that is part of the PKC superfamily. PKCε plays a complex role in the regulation of migration, adhesion and cytokinesis and in the present article we discuss the interplay between these processes. Integrin-mediated interaction with the actin cytoskeleton is a known regulator of cell adhesion and migration and there is emerging evidence that this pathway may also be essential for cytokinesis. We discuss evidence that a known actin-binding region in PKCε is involved in PKCε-mediated regulation of cytokinesis, providing a link between integrin-mediated stabilization of the cytokinesis furrow and PKCε recruitment.
Collapse
Affiliation(s)
- Nicola Brownlow
- *Protein Phosphorylation Laboratory, Cancer Research UK London Research Institute, London WC2A 3LY, U.K
| | - Tanya Pike
- *Protein Phosphorylation Laboratory, Cancer Research UK London Research Institute, London WC2A 3LY, U.K
| | - Victoria Crossland
- *Protein Phosphorylation Laboratory, Cancer Research UK London Research Institute, London WC2A 3LY, U.K
| | - Jeroen Claus
- *Protein Phosphorylation Laboratory, Cancer Research UK London Research Institute, London WC2A 3LY, U.K
| | | |
Collapse
|
23
|
Elia N, Ott C, Lippincott-Schwartz J. Incisive imaging and computation for cellular mysteries: lessons from abscission. Cell 2014; 155:1220-31. [PMID: 24315094 DOI: 10.1016/j.cell.2013.11.011] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2013] [Indexed: 02/06/2023]
Abstract
The final cleavage event that terminates cell division, abscission of the small, dense intercellular bridge, has been particularly challenging to resolve. Here, we describe imaging innovations that helped answer long-standing questions about the mechanism of abscission. We further explain how computational modeling of high-resolution data was employed to test hypotheses and generate additional insights. We present the model that emerges from application of these complimentary approaches. Similar experimental strategies will undoubtedly reveal exciting details about other underresolved cellular structures.
Collapse
Affiliation(s)
- Natalie Elia
- Department of Life Sciences and the NIBN, Ben Gurion University of the Negev, Beer Sheva 84105, Israel.
| | | | | |
Collapse
|
24
|
Fan Y, Li R, Huang J, Zhao HC, Ding T, Sun X, Yu Y, Qiao J. Improved efficiency of microsurgical enucleated tripronuclear zygotes development and embryonic stem cell derivation by supplementing epidermal growth factor, brain-derived neurotrophic factor, and insulin-like growth factor-1. Stem Cells Dev 2014; 23:563-75. [PMID: 24261581 DOI: 10.1089/scd.2013.0420] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Human embryonic stem cells (hESCs) hold great promise for future clinical cell therapies because of their unique potential to differentiate into all human cell types. However, the destruction of normal fertilized embryos and the derivation of hESCs for research has resulted in polarized ethical debates, with most of the controversy centered on embryo destruction. Therefore, due to less ethical controversy surrounding them, abnormal fertilized zygotes that are usually discarded are a potential feasible resource for the derivation of hESCs. Microsurgery on human polyspermic zygotes can contribute to the derivation of hESCs, but the efficiency is much lower. Here, we reported a culture system to enhance the developmental competence of such microsurgical human polyspermic zygotes by EGF-BDNF-IGF-1 combination, which eventually resulted in the increased derivation efficiency of hESCs from them. We found that the developmental efficiency of microsurgical enucleated tripronuclear (3PN) embryos cultured with the EGF-BDNF-IGF-1 combination was significantly increased compared with the control group. More importantly, when the microsurgical enucleated 3PN embryos were cultured in medium supplemented with EGF-BDNF-IGF-1, the frequency ratio of chromosome abnormality was reduced. Our present study will facilitate the development of hESC line derivation in subsequent studies and also provide an additional choice for infertile couples.
Collapse
Affiliation(s)
- Yong Fan
- 1 Key Laboratory for Major Obstetric Diseases of Guangdong Province, The Third Affiliated Hospital of Guangzhou Medical University , Guangzhou, People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Mathew SS, Nieves B, Sequeira S, Sambandamoorthy S, Pumiglia K, Larsen M, Laflamme SE. Integrins promote cytokinesis through the RSK signaling axis. J Cell Sci 2013; 127:534-45. [PMID: 24284076 DOI: 10.1242/jcs.133280] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Cytokinesis is the final stage in cell division. Although integrins can regulate cytokinesis, the mechanisms involved are not fully understood. In this study, we demonstrate that integrin-regulated ERK (extracellular signal-related kinase) and RSK (p90 ribosomal S6 kinase) signaling promotes successful cytokinesis. Inhibiting the activation of ERK and RSK in CHO cells by a mutation in the integrin β1 cytoplasmic tail or with pharmacological inhibitors results in the accumulation of cells with midbodies and the formation of binucleated cells. Activation of ERK and RSK signaling by the expression of constitutively active RAF1 suppresses the mutant phenotype in a RSK-dependent manner. Constitutively active RSK2 also restores cytokinesis inhibited by the mutant integrin. Importantly, the regulatory role of the RSK pathway is not specific to CHO cells. MCF-10A human mammary epithelial cells and HPNE human pancreatic ductal epithelial cells exhibit a similar dependence on RSK for successful cytokinesis. In addition, depriving mitotic MCF10A cells of integrin-mediated adhesion by incubating them in suspension suppressed ERK and RSK activation and resulted in a failure of cytokinesis. Furthermore, inhibition of RSK or integrins within the 3D context of a developing salivary gland organ explant also leads to an accumulation of epithelial cells with midbodies, suggesting a similar defect in cytokinesis. Interestingly, neither ERK nor RSK regulates cytokinesis in human fibroblasts, suggesting cell-type specificity. Taken together, our results identify the integrin-RSK signaling axis as an important regulator of cytokinesis in epithelial cells. We propose that the proper interaction of cells with their microenvironment through integrins contributes to the maintenance of genomic stability by promoting the successful completion of cytokinesis.
Collapse
Affiliation(s)
- Shomita S Mathew
- Center for Cell Biology and Cancer Research, Albany Medical College, Albany, NY 12208, USA
| | | | | | | | | | | | | |
Collapse
|
26
|
Rabineau M, Kocgozlu L, Dujardin D, Senger B, Haikel Y, Voegel JC, Freund JN, Schaaf P, Lavalle P, Vautier D. Contribution of soft substrates to malignancy and tumor suppression during colon cancer cell division. PLoS One 2013; 8:e78468. [PMID: 24167628 PMCID: PMC3805547 DOI: 10.1371/journal.pone.0078468] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2013] [Accepted: 09/13/2013] [Indexed: 01/05/2023] Open
Abstract
In colon cancer, a highly aggressive disease, progression through the malignant sequence is accompanied by increasingly numerous chromosomal rearrangements. To colonize target organs, invasive cells cross several tissues of various elastic moduli. Whether soft tissue increases malignancy or in contrast limits invasive colon cell spreading remains an open question. Using polyelectrolyte multilayer films mimicking microenvironments of various elastic moduli, we revealed that human SW480 colon cancer cells displayed increasing frequency in chromosomal segregation abnormalities when cultured on substrates with decreasing stiffness. Our results show that, although decreasing stiffness correlates with increased cell lethality, a significant proportion of SW480 cancer cells did escape from the very soft substrates, even when bearing abnormal chromosome segregation, achieve mitosis and undergo a new cycle of replication in contrast to human colonic HCoEpiC cells which died on soft substrates. This observation opens the possibility that the ability of cancer cells to overcome defects in chromosome segregation on very soft substrates could contribute to increasing chromosomal rearrangements and tumor cell aggressiveness.
Collapse
Affiliation(s)
- Morgane Rabineau
- Inserm UMR 1121, Strasbourg, France
- Université de Strasbourg, Faculté de Chirurgie Dentaire, Strasbourg, France
- Fédération de Médecine Translationnelle, Strasbourg, France
| | - Leyla Kocgozlu
- Inserm UMR 1121, Strasbourg, France
- Université de Strasbourg, Faculté de Chirurgie Dentaire, Strasbourg, France
| | | | - Bernard Senger
- Inserm UMR 1121, Strasbourg, France
- Université de Strasbourg, Faculté de Chirurgie Dentaire, Strasbourg, France
- Fédération de Médecine Translationnelle, Strasbourg, France
| | - Youssef Haikel
- Inserm UMR 1121, Strasbourg, France
- Université de Strasbourg, Faculté de Chirurgie Dentaire, Strasbourg, France
- Fédération de Médecine Translationnelle, Strasbourg, France
| | - Jean-Claude Voegel
- Inserm UMR 1121, Strasbourg, France
- Université de Strasbourg, Faculté de Chirurgie Dentaire, Strasbourg, France
- Fédération de Médecine Translationnelle, Strasbourg, France
| | - Jean-Noel Freund
- Inserm UMR S1113, Université de Strasbourg, Strasbourg, France
- Université de Strasbourg, Faculté de Médecine, Strasbourg, France
- Fédération de Médecine Translationnelle, Strasbourg, France
| | - Pierre Schaaf
- Inserm UMR 1121, Strasbourg, France
- Université de Strasbourg, Faculté de Chirurgie Dentaire, Strasbourg, France
- CNRS, UPR 22, Institut Charles Sadron, Strasbourg, France
- Fédération de Médecine Translationnelle, Strasbourg, France
| | - Philippe Lavalle
- Inserm UMR 1121, Strasbourg, France
- Université de Strasbourg, Faculté de Chirurgie Dentaire, Strasbourg, France
- Fédération de Médecine Translationnelle, Strasbourg, France
| | - Dominique Vautier
- Inserm UMR 1121, Strasbourg, France
- Université de Strasbourg, Faculté de Chirurgie Dentaire, Strasbourg, France
- Fédération de Médecine Translationnelle, Strasbourg, France
| |
Collapse
|
27
|
Peng H, Ong YM, Shah WA, Holland PC, Carbonetto S. Integrins regulate centrosome integrity and astrocyte polarization following a wound. Dev Neurobiol 2013; 73:333-53. [PMID: 22949126 DOI: 10.1002/dneu.22055] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2012] [Revised: 08/14/2012] [Accepted: 08/27/2012] [Indexed: 12/31/2022]
Abstract
In response to a wound, astrocytes in culture extend microtubule-rich processes and polarize, orienting their centrosomes and Golgi apparatus woundside. β1 Integrin null astrocytes fail to extend processes toward the wound, and are disoriented, and often migrate away orthogonal, to the wound. The centrosome is unusually fragmented in β1 integrin null astrocytes. Expression of a β1 integrin cDNA in the null background yields cells with intact centrosomes that polarize and extend processes normally. Fragmented centrosomes rapidly assemble following integrin ligation and cell attachment. However, several experiments indicated that cell adhesion is not necessary. For example, astrocytes in suspension expressing a chimeric β1 subunit that can be activated by an antibody assemble centrosomes suggesting that β1 activation is sufficient to cause centrosome assembly in the absence of cell adhesion. siRNA knockdown of PCM1, a major centrosomal protein, inhibits cell polarization, consistent with the notion that centrosomes are necessary for polarity and that integrins regulate polarity via centrosome integrity. Screening inhibitors of molecules downstream of integrins indicate that neither FAK nor ILK is involved in regulation of centrosome integrity. In contrast, blebbistatin, a specific inhibitor of non-muscle myosin II (NMII), mimics the response of β1 integrin null astrocytes by disrupting centrosome integrity and cell polarization. Blebbistatin also inhibits integrin-mediated centrosome assembly in astrocytes attaching to fibronectin, consistent with the hypothesis that NMII functions downstream of integrins in regulating centrosome integrity.
Collapse
Affiliation(s)
- Huashan Peng
- Centre for Research in Neuroscience, McGill University Health Centre, Montreal, Quebec, H3G 1A4, Canada
| | | | | | | | | |
Collapse
|
28
|
Patel H, Zich J, Serrels B, Rickman C, Hardwick KG, Frame MC, Brunton VG. Kindlin-1 regulates mitotic spindle formation by interacting with integrins and Plk-1. Nat Commun 2013; 4:2056. [PMID: 23804033 DOI: 10.1038/ncomms3056] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2012] [Accepted: 05/24/2013] [Indexed: 11/08/2022] Open
Abstract
Kindlin-1 binds to integrins and regulates integrin activation at cell adhesions. Here we report a new function of Kindlin-1 in regulating spindle assembly. We show that Kindlin-1 localizes to centrosomes, its concentration peaking during G2/M, where it associates with various pericentriolar material proteins, including Polo-like kinase 1. Short interfering RNA-mediated depletion of Kindlin-1 increases formation of abnormal mitotic spindles and decreases cellular survival. This effect is dependent not only on the ability of Kindlin-1 to bind integrins but also on Polo-like kinase 1-mediated Kindlin-1 phosphorylation. We demonstrate that a subcellular pool of phosphorylated Kindlin-1 is located exclusively at centrosomes. Our work identifies a novel cellular role for Kindlin-1 in ensuring mitotic spindle assembly and cellular survival that is controlled by phosphorylation via Polo-like kinase 1.
Collapse
Affiliation(s)
- Hitesh Patel
- Edinburgh Cancer Research UK Centre, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh EH4 2XR, UK
| | | | | | | | | | | | | |
Collapse
|
29
|
Lessons from the embryonic neural stem cell niche for neural lineage differentiation of pluripotent stem cells. Stem Cell Rev Rep 2012; 8:813-29. [PMID: 22628111 PMCID: PMC3412081 DOI: 10.1007/s12015-012-9381-8] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Pluripotent stem cells offer an abundant and malleable source for the generation of differentiated cells for transplantation as well as for in vitro screens. Patterning and differentiation protocols have been developed to generate neural progeny from human embryonic or induced pluripotent stem cells. However, continued refinement is required to enhance efficiency and to prevent the generation of unwanted cell types. We summarize and interpret insights gained from studies of embryonic neuroepithelium. A multitude of factors including soluble molecules, interactions with the extracellular matrix and neighboring cells cooperate to control neural stem cell self-renewal versus differentiation. Applying these findings and concepts to human stem cell systems in vitro may yield more appropriately patterned cell types for biomedical applications.
Collapse
|
30
|
Matthews HK, Baum B. The metastatic cancer cell cortex: An adaptation to enhance robust cell division in novel environments? Bioessays 2012; 34:1017-20. [DOI: 10.1002/bies.201200109] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
31
|
Brizzi MF, Tarone G, Defilippi P. Extracellular matrix, integrins, and growth factors as tailors of the stem cell niche. Curr Opin Cell Biol 2012; 24:645-51. [PMID: 22898530 DOI: 10.1016/j.ceb.2012.07.001] [Citation(s) in RCA: 282] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2012] [Revised: 06/13/2012] [Accepted: 07/09/2012] [Indexed: 12/21/2022]
Abstract
It is widely acknowledged that integrins, the major receptors for the extracellular matrix (ECM) proteins, exert an extensive crosstalk with many growth factor and cytokine receptors. Among them, growth factor receptors, such as the EGFR, MET, PDGFR and VEGFR, and the IL-3 receptor have been shown to be physically and functionally associated to integrins. The connection between integrins and other transmembrane receptors is bidirectional, integrins being essential for receptor signalling, and receptors being involved in regulation of integrin expression or activation. Moreover, there is accumulating evidence for direct binding of specific growth factors and morphogens to the ECM proteins, suggesting that ECM might spatially integrate different types of signals in a specific microenvironment, facilitating integrin/transmembrane receptors connection. These interactions are crucial in controlling a variety of cell behaviours including proliferation, survival and differentiation. The increasing interest for cell therapy in regenerative medicine has recently emphasized the role of cell-ECM adhesion as stem cell determinant. The relevance of ECM, integrins and growth factor receptor network in the establishment of stem cell niche, in maintenance of stem cells and in their differentiation will be analyzed in the present review.
Collapse
Affiliation(s)
- Maria Felice Brizzi
- Università degli Studi di Torino, Department of Medical Sciences, Torino, Italy
| | | | | |
Collapse
|
32
|
Laminin/β1 integrin signal triggers axon formation by promoting microtubule assembly and stabilization. Cell Res 2012; 22:954-72. [PMID: 22430151 DOI: 10.1038/cr.2012.40] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Axon specification during neuronal polarization is closely associated with increased microtubule stabilization in one of the neurites of unpolarized neuron, but how this increased microtubule stability is achieved is unclear. Here, we show that extracellular matrix (ECM) component laminin promotes neuronal polarization via regulating directional microtubule assembly through β1 integrin (Itgb1). Contact with laminin coated on culture substrate or polystyrene beads was sufficient for axon specification of undifferentiated neurites in cultured hippocampal neurons and cortical slices. Active Itgb1 was found to be concentrated in laminin-contacting neurites. Axon formation was promoted and abolished by enhancing and attenuating Itgb1 signaling, respectively. Interestingly, laminin contact promoted plus-end microtubule assembly in a manner that required Itgb1. Moreover, stabilizing microtubules partially prevented polarization defects caused by Itgb1 downregulation. Finally, genetic ablation of Itgb1 in dorsal telencephalic progenitors caused deficits in axon development of cortical pyramidal neurons. Thus, laminin/Itgb1 signaling plays an instructive role in axon initiation and growth, both in vitro and in vivo, through the regulation of microtubule assembly. This study has established a linkage between an extrinsic factor and intrinsic cytoskeletal dynamics during neuronal polarization.
Collapse
|
33
|
Fan Y, Li R, Huang J, Yu Y, Qiao J. Diploid, but not haploid, human embryonic stem cells can be derived from microsurgically repaired tripronuclear human zygotes. Cell Cycle 2012; 12:302-11. [PMID: 23255130 DOI: 10.4161/cc.23103] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Human embryonic stem cells have shown tremendous potential in regenerative medicine, and the recent progress in haploid embryonic stem cells provides new insights for future applications of embryonic stem cells. Disruption of normal fertilized embryos remains controversial; thus, the development of a new source for human embryonic stem cells is important for their usefulness. Here, we investigated the feasibility of haploid and diploid embryo reconstruction and embryonic stem cell derivation using microsurgically repaired tripronuclear human zygotes. Diploid and haploid zygotes were successfully reconstructed, but a large proportion of them still had a tripolar spindle assembly. The reconstructed embryos developed to the blastocyst stage, although the loss of chromosomes was observed in these zygotes. Finally, triploid and diploid human embryonic stem cells were derived from tripronuclear and reconstructed zygotes (from which only one pronucleus was removed), but haploid human embryonic stem cells were not successfully derived from the reconstructed zygotes when two pronuclei were removed. Both triploid and diploid human embryonic stem cells showed the general characteristics of human embryonic stem cells. These results indicate that the lower embryo quality resulting from abnormal spindle assembly contributed to the failure of the haploid embryonic stem cell derivation. However, the successful derivation of diploid embryonic stem cells demonstrated that microsurgical tripronuclear zygotes are an alternative source of human embryonic stem cells. In the future, improving spindle assembly will facilitate the application of triploid zygotes to the field of haploid embryonic stem cells.
Collapse
Affiliation(s)
- Yong Fan
- Key Laboratory for Major Obstetric Diseases of Guangdong Province, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | | | | | | | | |
Collapse
|
34
|
Cytokinesis failure due to derailed integrin traffic induces aneuploidy and oncogenic transformation in vitro and in vivo. Oncogene 2011; 31:3597-606. [PMID: 22120710 PMCID: PMC3419982 DOI: 10.1038/onc.2011.527] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Aneuploidy is frequently detected in solid tumors but the mechanisms regulating the generation of aneuploidy and their relevance in cancer initiation remain under debate and are incompletely characterized. Spatial and temporal regulation of integrin traffic is critical for cell migration and cytokinesis. Impaired integrin endocytosis, because of the loss of Rab21 small GTPase or mutations in the integrin β-subunit cytoplasmic tail, induces failure of cytokinesis in vitro. Here, we describe that repeatedly failed cytokinesis, because of impaired traffic, is sufficient to trigger the generation of aneuploid cells, which display characteristics of oncogenic transformation in vitro and are tumorigenic in vivo. Furthermore, in an in vivo mouse xenograft model, non-transformed cells with impaired integrin traffic formed tumors with a long latency. More detailed investigation of these tumors revealed that the tumor cells were aneuploid. Therefore, abnormal integrin traffic was linked with generation of aneuploidy and cell transformation also in vivo. In human prostate and ovarian cancer samples, downregulation of Rab21 correlates with increased malignancy. Loss-of-function experiments demonstrate that long-term depletion of Rab21 is sufficient to induce chromosome number aberrations in normal human epithelial cells. These data are the first to demonstrate that impaired integrin traffic is sufficient to induce conversion of non-transformed cells to tumorigenic cells in vitro and in vivo.
Collapse
|
35
|
de Andrea CE, Hogendoorn PCW. Epiphyseal growth plate and secondary peripheral chondrosarcoma: the neighbours matter. J Pathol 2011; 226:219-28. [PMID: 21956842 DOI: 10.1002/path.3003] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2011] [Revised: 09/20/2011] [Accepted: 09/22/2011] [Indexed: 12/16/2022]
Abstract
Chondrocytes interact with their neighbours through their cartilaginous extracellular matrix (ECM). Chondrocyte-matrix interactions compensate the lack of cell-cell contact and are modulated by proteoglycans and other molecules. The epiphyseal growth plate is a highly organized tissue responsible for long bone elongation. The growth plate is regulated by gradients of morphogens that are established by proteoglycans. Morphogens diffuse across the ECM, creating short- and long-range signalling that lead to the formation of a polarized tissue. Mutations affecting genes that modulate cell-matrix interactions are linked to several human disorders. Homozygous mutations of EXT1/EXT2 result in reduced synthesis and shortened heparan sulphate chains on both cell surface and matrix proteoglycans. This disrupts the diffusion gradients of morphogens and signal transduction in the epiphyseal growth plate, contributing to loss of cell polarity and osteochondroma formation. Osteochondromas are cartilage-capped bony projections arising from the metaphyses of endochondral bones adjacent to the growth plate. The osteochondroma cap is formed by cells with homozygous mutation of EXT1/EXT2 and committed stem cells/wild-type chondrocytes. Osteochondroma serves as a niche (a permissive environment), which facilitates the committed stem cells/wild-type chondrocytes to acquire secondary genetic changes to form a secondary peripheral chondrosarcoma. In such a scenario, the micro-environment is the site of the initiating processes that ultimately lead to cancer.
Collapse
Affiliation(s)
- Carlos E de Andrea
- Department of Pathology, Leiden University Medical Centre, Leiden, The Netherlands
| | | |
Collapse
|
36
|
Colello D, Mathew S, Ward R, Pumiglia K, LaFlamme SE. Integrins regulate microtubule nucleating activity of centrosome through mitogen-activated protein kinase/extracellular signal-regulated kinase kinase/extracellular signal-regulated kinase (MEK/ERK) signaling. J Biol Chem 2011; 287:2520-30. [PMID: 22117069 DOI: 10.1074/jbc.m111.254128] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Microtubule nucleation is an essential step in the formation of the microtubule cytoskeleton. We recently showed that androgen and Src promote microtubule nucleation and γ-tubulin accumulation at the centrosome. Here, we explore the mechanisms by which androgen and Src regulate these processes and ask whether integrins play a role. We perturb integrin function by a tyrosine-to-alanine substitution in membrane-proximal NPIY motif in the integrin β1 tail and show that this mutant substantially decreases microtubule nucleation and γ-tubulin accumulation at the centrosome. Because androgen stimulation promotes the interaction of the androgen receptor with Src, resulting in PI3K/AKT and MEK/ERK signaling, we asked whether these pathways are inhibited by the mutant integrin and whether they regulate microtubule nucleation. Our results indicate that the formation of the androgen receptor-Src complex and the activation of downstream pathways are significantly suppressed when cells are adhered by the mutant integrin. Inhibitor studies indicate that microtubule nucleation requires MEK/ERK but not PI3K/AKT signaling. Importantly, the expression of activated RAF-1 is sufficient to rescue microtubule nucleation inhibited by the mutant integrin by promoting the centrosomal accumulation of γ-tubulin. Our data define a novel paradigm of integrin signaling, where integrins regulate microtubule nucleation by promoting the formation of androgen receptor-Src signaling complexes to activate the MEK/ERK signaling pathway.
Collapse
Affiliation(s)
- Diane Colello
- Center for Cell Biology and Cancer Research, Albany Medical College, Albany, New York 12208, USA
| | | | | | | | | |
Collapse
|
37
|
Kocgozlu L, Rabineau M, Koenig G, Haikel Y, Schaaf P, Freund JN, Voegel JC, Lavalle P, Vautier D. The control of chromosome segregation during mitosis in epithelial cells by substrate elasticity. Biomaterials 2011; 33:798-809. [PMID: 22041225 DOI: 10.1016/j.biomaterials.2011.10.024] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2011] [Accepted: 10/10/2011] [Indexed: 11/19/2022]
Abstract
Materials of defined elasticity, including synthetic material scaffolds and tissue-derived matrices, can regulate biological responses of cells and in particular adhesion, migration, growth and differentiation which are essential parameters for tissue integration. These responses have been extensively investigated in interphase cells, but little is known whether and how material elasticity affects mitotic cells. We used polyelectrolyte multilayer films as model substrates with elastic modulus ranging from Eap = 0 up to Eap = 500 kPa and mitotic PtK2 epithelial cells to address these important questions. Soft substrates (Eap < 50 kPa) led to abnormal morphology in chromosome segregation, materialized by chromatin bridges and chromosome lagging. Frequency of these damages increased with decreasing substrate stiffness and was correlated with a pro-apoptotic phenotype. Mitotic spindle was not observed on soft substrates where formation of chromatin damages is due to low β1-integrin engagement and decrease of Rac1 activities. This work constitutes the first evidence that soft substrates hinder epithelial cell division. In perspective, our findings emphasize the prime incidence of the material elasticity on the fate of the phenotype, especially of stem cells in the mitotic phase.
Collapse
Affiliation(s)
- Leyla Kocgozlu
- Institut National de la Santé et de la Recherche Médicale, INSERM Unité 977, 11 rue Humann, 67085 Strasbourg Cedex, France
| | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Xu X, Vogel BE. A new job for ancient extracellular matrix proteins: Hemicentins stabilize cleavage furrows. Commun Integr Biol 2011; 4:433-5. [PMID: 21966563 DOI: 10.4161/cib.4.4.15324] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2011] [Accepted: 03/01/2011] [Indexed: 01/21/2023] Open
Abstract
Interactions between extracellular matrix (ECM) proteins and transmembrane receptors mediate changes in cell shape during cell migration, adhesion, differentiation and polarization. Cytokinesis is the final step in cell division as cells employ a contractile ring composed of actin and myosin to partition one cell into two. During the partition process, an invagination in nascent membrane forms a new extracellular space called the cleavage furrow. Despite the dramatic changes in cell shape during cytokinesis, existing models include no role for the ECM. In a recent paper, we show that hemicentins assemble in the cleavage furrow of C. elegans germ cells and mouse embryo blastomeres. Hemicentin depletion results in membrane destabilization, cleavage furrow retraction and cytokinesis failure. The data suggest that hemicentins and other ECM proteins stabilize the cleavage furrow during cytokinesis of multiple cell types.
Collapse
Affiliation(s)
- Xuehong Xu
- Center for Biomedical Engineering and Technology and Department of Physiology; University of Maryland Baltimore; Baltimore, MD USA
| | | |
Collapse
|
39
|
Abstract
How might the extracellular matrix contribute to cytokinesis? In a recent report, evidence is presented that the conserved extracellular matrix protein hemicentin(HIM-4) is required for cytokinesis in worms and mice.
Collapse
Affiliation(s)
- Shawn N Jordan
- Columbia University, Department of Pathology and Cell Biology, New York, NY 10032, USA
| | | | | |
Collapse
|
40
|
|
41
|
Holubcová Z, Matula P, Sedláčková M, Vinarský V, Doležalová D, Bárta T, Dvořák P, Hampl A. Human embryonic stem cells suffer from centrosomal amplification. Stem Cells 2011; 29:46-56. [PMID: 20960514 DOI: 10.1002/stem.549] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Propagation of human embryonic stem cells (hESCs) in culture tends to alter karyotype, potentially limiting the prospective use of these cells in patients. The chromosomal instability of some malignancies is considered to be driven, at least in part, by centrosomal overamplification, perturbing balanced chromosome segregation. Here, we report, for the first time, that very high percentage of cultured hESCs has supernumerary centrosomes during mitosis. Supernumerary centrosomes were strictly associated with an undifferentiated hESC state and progressively disappeared on prolonged propagation in culture. Improved attachment to culture substratum and inhibition of CDK2 and Aurora A (key regulators of centrosomal metabolism) diminished the frequency of multicentrosomal mitoses. Thus, both attenuated cell attachment and deregulation of machinery controlling centrosome number contribute to centrosomal overamplification in hESCs. Linking the excessive number of centrosomes in mitoses to the ploidy indicated that both overduplication within a single cell cycle and mitotic failure contributed to generation of numerical centrosomal abnormalities in hESCs. Collectively, our data indicate that supernumerary centrosomes are a significant risk factor for chromosome instability in cultured hESCs and should be evaluated when new culture conditions are being implemented.
Collapse
Affiliation(s)
- Zuzana Holubcová
- Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Marthiens V, Kazanis I, Moss L, Long K, Ffrench-Constant C. Adhesion molecules in the stem cell niche--more than just staying in shape? J Cell Sci 2010; 123:1613-22. [PMID: 20445012 DOI: 10.1242/jcs.054312] [Citation(s) in RCA: 118] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The expression of adhesion molecules by stem cells within their niches is well described, but what is their function? A conventional view is that these adhesion molecules simply retain stem cells in the niche and thereby maintain its architecture and shape. Here, we review recent literature showing that this is but one of their roles, and that they have essential functions in all aspects of the stem cell-niche interaction--retention, division and exit. We also highlight from this literature evidence supporting a simple model whereby the regulation of centrosome positioning and spindle angle is regulated by both cadherins and integrins, and the differential activity of these two adhesion molecules enables the fundamental stem cell property of switching between asymmetrical and symmetrical divisions.
Collapse
|
43
|
LFA-1 activity state on dendritic cells regulates contact duration with T cells and promotes T-cell priming. Blood 2010; 116:1885-94. [PMID: 20530790 DOI: 10.1182/blood-2009-05-224428] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
A key event in the successful induction of adaptive immune responses is the antigen-specific activation of T cells by dendritic cells (DCs). Although LFA-1 (lymphocyte function-associated antigen 1) on T cells is considered to be important for antigen-specific T-cell activation, the role for LFA-1 on DCs remains elusive. Using 2 different approaches to activate LFA-1 on DCs, either by deletion of the αL-integrin cytoplasmic GFFKR sequence or by silencing cytohesin-1-interacting protein, we now provide evidence that DCs are able to make use of active LFA-1 and can thereby control the contact duration with naive T cells. Enhanced duration of DC/T-cell interaction correlates inversely with antigen-specific T-cell proliferation, generation of T-helper 1 cells, and immune responses leading to delayed-type hypersensitivity. We could revert normal interaction time and T-cell proliferation to wild-type levels by inhibition of active LFA-1 on DCs. Our data further suggest that cytohesin-1-interacting protein might be responsible for controlling LFA-1 deactivation on mature DCs. In summary, our findings indicate that LFA-1 on DCs needs to be in an inactive state to ensure optimal T-cell activation and suggest that regulation of LFA-1 activity allows DCs to actively control antigen-driven T-cell proliferation and effective immune responses.
Collapse
|
44
|
Colello D, Reverte CG, Ward R, Jones CW, Magidson V, Khodjakov A, LaFlamme SE. Androgen and Src signaling regulate centrosome activity. J Cell Sci 2010; 123:2094-102. [PMID: 20501699 DOI: 10.1242/jcs.057505] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Microtubules nucleated from gamma-tubulin ring complexes located at the centrosome regulate the localization of organelles, promote vesicular transport and direct cell migration. Although several signaling mechanisms have been identified that regulate microtubule dynamics during interphase, signaling pathways that promote microtubule nucleation remain elusive. We assayed microtubule regrowth following nocodazole washout in human fibroblasts and CHO-K1 cells adhered to fibronectin in either normal serum-free medium or the serum-free, growth-promoting medium, CCM1, which contains IGF1 and androgen, as well as other nutrients. The results indicate that integrin-mediated adhesion is not sufficient to promote rapid microtubule regrowth in either cell type. The addition of androgen, but not IGF1, for 5 minutes was sufficient to promote rapid regrowth and this occurred by a mechanism requiring the androgen receptor. Since Src is a component of the cytoplasmic androgen-receptor-signaling complex, we examined its role using Src siRNA, the Src kinase inhibitor SU6656, and the expression of a constitutively active Src mutant. The data show that Src signaling is both required and sufficient to promote rapid microtubule regrowth in cells adhered to fibronectin. Measurement of the density of microtubules close to the centrosome and the rates of GFP-EB1-labeled microtubules emanating from the centrosome indicated that Src signaling promotes microtubule nucleation. Furthermore, recovery of GFP-gamma-tubulin at the centrosome following photobleaching and measurements of endogenous gamma-tubulin levels at the centrosome showed that androgen and Src signaling regulate the levels of centrosomal gamma-tubulin. Thus, we propose that androgen and Src signaling regulate microtubule nucleation during interphase by promoting the centrosomal localization of gamma-tubulin.
Collapse
Affiliation(s)
- Diane Colello
- Center for Cell Biology and Cancer Research, Albany Medical College, Albany, NY 12208, USA
| | | | | | | | | | | | | |
Collapse
|
45
|
Heng YW, Koh CG. Actin cytoskeleton dynamics and the cell division cycle. Int J Biochem Cell Biol 2010; 42:1622-33. [PMID: 20412868 DOI: 10.1016/j.biocel.2010.04.007] [Citation(s) in RCA: 202] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2010] [Revised: 04/12/2010] [Accepted: 04/14/2010] [Indexed: 11/29/2022]
Abstract
The network of actin filaments is one of the crucial cytoskeletal structures contributing to the morphological framework of a cell and which participates in the dynamic regulation of cellular functions. In adherent cell types, cells adhere to the substratum during interphase and spread to assume their characteristic shape supported by the actin cytoskeleton. This actin cytoskeleton is reorganized during mitosis to form rounded cells with increased cortical rigidity. The actin cytoskeleton is re-established after mitosis, allowing cells to regain their extended shape and attachment to the substratum. The modulation of such drastic changes in cell shape in coordination with cell cycle progression suggests a tight regulatory interaction between cytoskeleton signalling, cell-cell/cell-matrix adhesions and mitotic events. Here, we review the contribution of the actin cytoskeleton to cell cycle progression with an emphasis on the effectors responsible for the regulation of the actin cytoskeleton and integration of their activities with the cell cycle machinery.
Collapse
Affiliation(s)
- Yi-Wen Heng
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | | |
Collapse
|
46
|
Nieves B, Jones CW, Ward R, Ohta Y, Reverte CG, LaFlamme SE. The NPIY motif in the integrin beta1 tail dictates the requirement for talin-1 in outside-in signaling. J Cell Sci 2010; 123:1216-26. [PMID: 20332112 DOI: 10.1242/jcs.056549] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Protein interactions with the integrin beta-subunit cytoplasmic domain (beta-tail) are essential for adhesion-dependent processes, including cell spreading and the connection of integrins with actin filaments at adhesion sites. Talin-1 binds to the conserved membrane-proximal NPxY motif of beta-tails (NPIY in beta1 integrin) promoting the inside-out activation of integrins and providing a linkage between integrins and the actin cytoskeleton. Here, we characterize the role of interactions between talin-1 and beta-tail downstream of integrin activation, in the context of recombinant integrins containing either the wild type (WT) or the (YA) mutant beta1A tail, with a tyrosine to alanine substitution in the NPIY motif. In addition to inhibiting integrin activation, the YA mutation suppresses cell spreading, integrin signaling, focal adhesion and stress-fiber formation, as well as microtubule assembly. Constitutive activation of the mutant integrin restores these integrin-dependent processes, bringing into question the importance of the NPIY motif downstream of integrin activation. Depletion of talin-1 using TLN1 siRNA demonstrated that talin-1 is required for cell spreading, focal adhesion and stress-fiber formation, as well as microtubule assembly, even when cells are adhered by constitutively activated WT integrins. Depletion of talin-1 does not inhibit these processes when cells are adhered by constitutively activated mutant integrins, suggesting that the binding of an inhibitory protein to the NPIY motif negatively regulates integrin function when talin-1 is depleted. We identified filamin A (FLNa) as this inhibitory protein; it binds to the beta1A tail in an NPIY-dependent manner and inhibition of FLNa expression in talin-1-depleted cells restores integrin function when cells are adhered by constitutively activated WT integrins. FLNa binds FilGAP, which is a negative regulator of Rac activation. Expression of the dominant inhibitory mutant, FilGAP(DeltaGAP), which lacks GAP activity restores spreading in cells adhered by constitutively activated integrins containing the beta1A tail, but not by integrins containing the beta1D tail, which is known to bind poorly to FLNa. Together, these results suggest that the binding of talin-1 to the NPIY motif is required downstream of integrin activation to promote cell spreading by preventing the inappropriate recruitment of FLNa and FilGAP to the beta1A tail. Our studies emphasize the importance of understanding the mechanisms that regulate the differential binding FLNa and talin-1 to the beta1 tail downstream of integrin activation in promoting integrin function.
Collapse
Affiliation(s)
- Bethsaida Nieves
- Center for Cell Biology and Cancer Research, Albany Medical College, 47 New Scotland Avenue, Albany, New York 12208, USA
| | | | | | | | | | | |
Collapse
|
47
|
Shi F, Harman J, Fujiwara K, Sottile J. Collagen I matrix turnover is regulated by fibronectin polymerization. Am J Physiol Cell Physiol 2010; 298:C1265-75. [PMID: 20107040 DOI: 10.1152/ajpcell.00341.2009] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Extracellular matrix (ECM) remodeling occurs during normal homeostasis and also plays an important role during development, tissue repair, and in various disease processes. ECM remodeling involves changes in the synthesis, deposition, and degradation of ECM molecules. ECM molecules can be degraded extracellularly, as well as intracellularly following endocytosis. Our data show that the ECM protein fibronectin is an important regulator of ECM remodeling. We previously showed that agents that inhibit the polymerization of fibronectin into ECM fibrils promote the loss of preexisting fibronectin matrix and accelerate fibronectin endocytosis and degradation. In this paper we show that inhibition of fibronectin polymerization leads to the loss of collagen I matrix fibrils and a corresponding increase in the levels of endocytosed collagen I. In contrast, manipulations that stabilize fibronectin matrix fibrils, such as caveolin-1 depletion, stabilize collagen I matrix fibrils and cause a decrease in ECM collagen I endocytosis. Our data also show that endocytosis of ECM collagen I is regulated by both beta1 integrins and Endo180/urokinase plasminogen activator associated protein (uPARAP). Unexpectedly, Endo180/uPARAP was also shown to promote the endocytosis of fibronectin from the ECM. These data demonstrate that fibronectin polymerization regulates the remodeling of ECM collagen I, in part, by regulating collagen I endocytosis. Furthermore, these data show that processes that regulate ECM deposition coordinately regulate the removal of proteins from the ECM. These data highlight the complexity of ECM remodeling. This multifaceted regulatory process may be important to ensure tight regulation of ECM fibronectin and collagen I levels.
Collapse
Affiliation(s)
- Feng Shi
- Aab Cardiovascular Research Institute, Univ. of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA
| | | | | | | |
Collapse
|
48
|
Humphries JD, Byron A, Bass MD, Craig SE, Pinney JW, Knight D, Humphries MJ. Proteomic analysis of integrin-associated complexes identifies RCC2 as a dual regulator of Rac1 and Arf6. Sci Signal 2009; 2:ra51. [PMID: 19738201 PMCID: PMC2857963 DOI: 10.1126/scisignal.2000396] [Citation(s) in RCA: 195] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The binding of integrin adhesion receptors to their extracellular matrix ligands controls cell morphology, movement, survival, and differentiation in various developmental, homeostatic, and disease processes. Here, we report a methodology to isolate complexes associated with integrin adhesion receptors, which, like other receptor-associated signaling complexes, have been refractory to proteomic analysis. Quantitative, comparative analyses of the proteomes of two receptor-ligand pairs, alpha(4)beta(1)-vascular cell adhesion molecule-1 and alpha(5)beta(1)-fibronectin, defined both core and receptor-specific components. Regulator of chromosome condensation-2 (RCC2) was detected in the alpha(5)beta(1)-fibronectin signaling network at an intersection between the Rac1 and adenosine 5'-diphosphate ribosylation factor 6 (Arf6) subnetworks. RCC2 knockdown enhanced fibronectin-induced activation of both Rac1 and Arf6 and accelerated cell spreading, suggesting that RCC2 limits the signaling required for membrane protrusion and delivery. Dysregulation of Rac1 and Arf6 function by RCC2 knockdown also abolished persistent migration along fibronectin fibers, indicating a functional role for RCC2 in directional cell movement. This proteomics workflow now opens the way to further dissection and systems-level analyses of adhesion signaling.
Collapse
Affiliation(s)
- Jonathan D. Humphries
- Wellcome Trust Centre for Cell-Matrix Research, University of Manchester, Manchester M13 9PT, UK
- Faculty of Life Sciences, University of Manchester, Manchester M13 9PT, UK
| | - Adam Byron
- Wellcome Trust Centre for Cell-Matrix Research, University of Manchester, Manchester M13 9PT, UK
- Faculty of Life Sciences, University of Manchester, Manchester M13 9PT, UK
| | - Mark D. Bass
- Wellcome Trust Centre for Cell-Matrix Research, University of Manchester, Manchester M13 9PT, UK
- Faculty of Life Sciences, University of Manchester, Manchester M13 9PT, UK
| | - Sue E. Craig
- Wellcome Trust Centre for Cell-Matrix Research, University of Manchester, Manchester M13 9PT, UK
- Faculty of Life Sciences, University of Manchester, Manchester M13 9PT, UK
| | - John W. Pinney
- Faculty of Life Sciences, University of Manchester, Manchester M13 9PT, UK
| | - David Knight
- Faculty of Life Sciences, University of Manchester, Manchester M13 9PT, UK
| | - Martin J. Humphries
- Wellcome Trust Centre for Cell-Matrix Research, University of Manchester, Manchester M13 9PT, UK
- Faculty of Life Sciences, University of Manchester, Manchester M13 9PT, UK
| |
Collapse
|
49
|
Abstract
Precise control of centrosome number is crucial for bipolar spindle assembly and accurate transmission of genetic material to daughter cells. Failure to properly control centrosome number results in supernumerary centrosomes, which are frequently found in cancer cells. This presents a paradox: during mitosis, cells with more than two centrosomes are prone to multipolar mitoses and cell death, however, cancer cells possessing extra centrosomes usually divide successfully. One mechanism frequently utilized by cancer cells to escape death caused by multipolar mitoses is the clustering of supernumerary centrosomes into bipolar arrays. An understanding of the molecular mechanisms by which cancer cells can suppress multipolar mitoses is beginning to emerge. Here, we review what's currently known about centrosome clustering mechanisms and discuss potential strategies to target these mechanisms for the selective killing of cancer cells.
Collapse
|
50
|
Loulier K, Lathia JD, Marthiens V, Relucio J, Mughal MR, Tang SC, Coksaygan T, Hall PE, Chigurupati S, Patton B, Colognato H, Rao MS, Mattson MP, Haydar TF, ffrench-Constant C. beta1 integrin maintains integrity of the embryonic neocortical stem cell niche. PLoS Biol 2009; 7:e1000176. [PMID: 19688041 PMCID: PMC2720642 DOI: 10.1371/journal.pbio.1000176] [Citation(s) in RCA: 142] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2009] [Accepted: 07/09/2009] [Indexed: 01/09/2023] Open
Abstract
During embryogenesis, the neural stem cells (NSC) of the developing cerebral cortex are located in the ventricular zone (VZ) lining the cerebral ventricles. They exhibit apical and basal processes that contact the ventricular surface and the pial basement membrane, respectively. This unique architecture is important for VZ physical integrity and fate determination of NSC daughter cells. In addition, the shorter apical process is critical for interkinetic nuclear migration (INM), which enables VZ cell mitoses at the ventricular surface. Despite their importance, the mechanisms required for NSC adhesion to the ventricle are poorly understood. We have shown previously that one class of candidate adhesion molecules, laminins, are present in the ventricular region and that their integrin receptors are expressed by NSC. However, prior studies only demonstrate a role for their interaction in the attachment of the basal process to the overlying pial basement membrane. Here we use antibody-blocking and genetic experiments to reveal an additional and novel requirement for laminin/integrin interactions in apical process adhesion and NSC regulation. Transient abrogation of integrin binding and signalling using blocking antibodies to specifically target the ventricular region in utero results in abnormal INM and alterations in the orientation of NSC divisions. We found that these defects were also observed in laminin alpha2 deficient mice. More detailed analyses using a multidisciplinary approach to analyse stem cell behaviour by expression of fluorescent transgenes and multiphoton time-lapse imaging revealed that the transient embryonic disruption of laminin/integrin signalling at the VZ surface resulted in apical process detachment from the ventricular surface, dystrophic radial glia fibers, and substantial layering defects in the postnatal neocortex. Collectively, these data reveal novel roles for the laminin/integrin interaction in anchoring embryonic NSCs to the ventricular surface and maintaining the physical integrity of the neocortical niche, with even transient perturbations resulting in long-lasting cortical defects.
Collapse
Affiliation(s)
- Karine Loulier
- Center for Neuroscience, Children's National Medical Center, Washington, D.C., United States of America
| | - Justin D. Lathia
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom
- Department of Medical Genetics, University of Cambridge, Cambridge, United Kingdom
- Centre for Brain Repair, University of Cambridge, Cambridge, United Kingdom
- Laboratory of Neuroscience, National Institute on Aging Intramural Research Program, Baltimore, Maryland, United States of America
| | - Veronique Marthiens
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom
- Department of Medical Genetics, University of Cambridge, Cambridge, United Kingdom
| | - Jenne Relucio
- Department of Pharmacology, State University of New York at Stony Brook, Stony Brook, New York, United States of America
| | - Mohamed R. Mughal
- Laboratory of Neuroscience, National Institute on Aging Intramural Research Program, Baltimore, Maryland, United States of America
| | - Sung-Chun Tang
- Laboratory of Neuroscience, National Institute on Aging Intramural Research Program, Baltimore, Maryland, United States of America
| | - Turhan Coksaygan
- School of Medicine, University of Maryland, Baltimore, Maryland, United States of America
| | - Peter E. Hall
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom
- Department of Medical Genetics, University of Cambridge, Cambridge, United Kingdom
- Centre for Brain Repair, University of Cambridge, Cambridge, United Kingdom
| | - Srinivasulu Chigurupati
- Laboratory of Neuroscience, National Institute on Aging Intramural Research Program, Baltimore, Maryland, United States of America
| | - Bruce Patton
- Center for Research on Occupational and Environmental Toxicology, Oregon Health and Science University, Portland, Oregon, United States of America
| | - Holly Colognato
- Department of Pharmacology, State University of New York at Stony Brook, Stony Brook, New York, United States of America
| | - Mahendra S. Rao
- Corporate Research Laboratories, Invitrogen Corporation, Carlsbad, California, United States of America
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Mark P. Mattson
- Laboratory of Neuroscience, National Institute on Aging Intramural Research Program, Baltimore, Maryland, United States of America
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Tarik F. Haydar
- Center for Neuroscience, Children's National Medical Center, Washington, D.C., United States of America
| | | |
Collapse
|