1
|
Pandya V, Deng D, Gupta S. Hypersynchronous EEG Patterns in a Patient with Holoprosencephaly. Clin EEG Neurosci 2025:15500594251346337. [PMID: 40448330 DOI: 10.1177/15500594251346337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/02/2025]
Abstract
Holoprosencephaly is a congenital malformation of the central nervous system resulting from failure of the rostral neural tube to bifurcate into the two cerebral hemispheres. Deep brain structures including the thalamus, hypothalamus, and basal ganglia can also be affected to varying degrees. Here we present a patient with a rare de novo pathogenic variant in the PPP1R12A gene and the middle interhemispheric (MIH) variant of holoprosencephaly with hypersynchronous patterns on electroencephalography (EEG). The most prevalent abnormal pattern was abundant hypersynchronous rhythmic theta activity most prominent over the bilateral centro-parietal regions. There was also frequent hypersynchronous rhythmic beta activity and rhythmic alpha range activity, which occurred both synchronously and asynchronously. Finally, there were occasional periods of voltage attenuation interrupting hypersynchronous theta activity. While hypersynchronous theta activity and episodic attenuation have been previously described in alobar and semilobar variants of holoprosencephaly, our report is the first to describe these findings in a patient with the MIH variant as well as the first to describe EEG patterns in a patient with a pathogenic variant in the PPP1R12A gene mutations in which are associated with urogenital and/or brain malformation syndrome. Additionally, the hypersynchronous alpha activity is the first report of such an EEG pattern in holoprosencephaly. In order to develop a more complete understanding of EEG patterns in holoprosencephaly further study is needed but this is challenged by the relative rarity of the disease.
Collapse
Affiliation(s)
- Vishal Pandya
- Comprehensive Epilepsy Center, Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Doris Deng
- Comprehensive Epilepsy Center, Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Siddharth Gupta
- Comprehensive Epilepsy Center, Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD
- Kennedy Krieger Institute, Baltimore, MD
| |
Collapse
|
2
|
Caballero-Mancebo S, Shinde R, Bolger-Munro M, Peruzzo M, Szep G, Steccari I, Labrousse-Arias D, Zheden V, Merrin J, Callan-Jones A, Voituriez R, Heisenberg CP. Friction forces determine cytoplasmic reorganization and shape changes of ascidian oocytes upon fertilization. NATURE PHYSICS 2024; 20:310-321. [PMID: 38370025 PMCID: PMC10866705 DOI: 10.1038/s41567-023-02302-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 10/23/2023] [Indexed: 02/20/2024]
Abstract
Contraction and flow of the actin cell cortex have emerged as a common principle by which cells reorganize their cytoplasm and take shape. However, how these cortical flows interact with adjacent cytoplasmic components, changing their form and localization, and how this affects cytoplasmic organization and cell shape remains unclear. Here we show that in ascidian oocytes, the cooperative activities of cortical actomyosin flows and deformation of the adjacent mitochondria-rich myoplasm drive oocyte cytoplasmic reorganization and shape changes following fertilization. We show that vegetal-directed cortical actomyosin flows, established upon oocyte fertilization, lead to both the accumulation of cortical actin at the vegetal pole of the zygote and compression and local buckling of the adjacent elastic solid-like myoplasm layer due to friction forces generated at their interface. Once cortical flows have ceased, the multiple myoplasm buckles resolve into one larger buckle, which again drives the formation of the contraction pole-a protuberance of the zygote's vegetal pole where maternal mRNAs accumulate. Thus, our findings reveal a mechanism where cortical actomyosin network flows determine cytoplasmic reorganization and cell shape by deforming adjacent cytoplasmic components through friction forces.
Collapse
Affiliation(s)
| | - Rushikesh Shinde
- Laboratoire de Matière et Systèmes Complexes, Université de Paris Cité and CNRS, Paris, France
| | | | - Matilda Peruzzo
- Institute of Science and Technology Austria, Klosterneuburg, Austria
| | | | - Irene Steccari
- Institute of Science and Technology Austria, Klosterneuburg, Austria
| | | | - Vanessa Zheden
- Institute of Science and Technology Austria, Klosterneuburg, Austria
| | - Jack Merrin
- Institute of Science and Technology Austria, Klosterneuburg, Austria
| | - Andrew Callan-Jones
- Laboratoire de Matière et Systèmes Complexes, Université de Paris Cité and CNRS, Paris, France
| | - Raphaël Voituriez
- Laboratoire Jean Perrin, Sorbonne Université and CNRS, Paris, France
- Laboratoire de Physique Théorique de la Matière Condensée, Sorbonne Université and CNRS, Paris, France
| | | |
Collapse
|
3
|
Functional interaction between Vangl2 and N-cadherin regulates planar cell polarization of the developing neural tube and cochlear sensory epithelium. Sci Rep 2023; 13:3905. [PMID: 36890135 PMCID: PMC9995352 DOI: 10.1038/s41598-023-30213-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 02/17/2023] [Indexed: 03/10/2023] Open
Abstract
Although the core constituents of the Wnt/planar cell polarity (PCP) signaling have been extensively studied, their downstream molecules and protein-protein interactions have not yet been fully elucidated. Here, we show genetic and molecular evidence that the PCP factor, Vangl2, functionally interacts with the cell-cell adhesion molecule, N-cadherin (also known as Cdh2), for typical PCP-dependent neural development. Vangl2 and N-cadherin physically interact in the neural plates undergoing convergent extension. Unlike monogenic heterozygotes, digenic heterozygous mice with Vangl2 and Cdh2 mutants exhibited defects in neural tube closure and cochlear hair cell orientation. Despite this genetic interaction, neuroepithelial cells derived from the digenic heterozygotes did not show additive changes from the monogenic heterozygotes of Vangl2 in the RhoA-ROCK-Mypt1 and c-Jun N-terminal kinase (JNK)-Jun pathways of Wnt/PCP signaling. Thus, cooperation between Vangl2 and N-cadherin is at least partly via direct molecular interaction; it is essential for the planar polarized development of neural tissues but not significantly associated with RhoA or JNK pathways.
Collapse
|
4
|
Nguyen NM, Merle T, Broders-Bondon F, Brunet AC, Battistella A, Land EBL, Sarron F, Jha A, Gennisson JL, Röttinger E, Fernández-Sánchez ME, Farge E. Mechano-biochemical marine stimulation of inversion, gastrulation, and endomesoderm specification in multicellular Eukaryota. Front Cell Dev Biol 2022; 10:992371. [PMID: 36531949 PMCID: PMC9754125 DOI: 10.3389/fcell.2022.992371] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 11/01/2022] [Indexed: 07/29/2023] Open
Abstract
The evolutionary emergence of the primitive gut in Metazoa is one of the decisive events that conditioned the major evolutionary transition, leading to the origin of animal development. It is thought to have been induced by the specification of the endomesoderm (EM) into the multicellular tissue and its invagination (i.e., gastrulation). However, the biochemical signals underlying the evolutionary emergence of EM specification and gastrulation remain unknown. Herein, we find that hydrodynamic mechanical strains, reminiscent of soft marine flow, trigger active tissue invagination/gastrulation or curvature reversal via a Myo-II-dependent mechanotransductive process in both the metazoan Nematostella vectensis (cnidaria) and the multicellular choanoflagellate Choanoeca flexa. In the latter, our data suggest that the curvature reversal is associated with a sensory-behavioral feeding response. Additionally, like in bilaterian animals, gastrulation in the cnidarian Nematostella vectensis is shown to participate in the biochemical specification of the EM through mechanical activation of the β-catenin pathway via the phosphorylation of Y654-βcatenin. Choanoflagellates are considered the closest living relative to metazoans, and the common ancestor of choanoflagellates and metazoans dates back at least 700 million years. Therefore, the present findings using these evolutionarily distant species suggest that the primitive emergence of the gut in Metazoa may have been initiated in response to marine mechanical stress already in multicellular pre-Metazoa. Then, the evolutionary transition may have been achieved by specifying the EM via a mechanosensitive Y654-βcatenin dependent mechanism, which appeared during early Metazoa evolution and is specifically conserved in all animals.
Collapse
Affiliation(s)
- Ngoc Minh Nguyen
- Mechanics and Genetics of Embryonic Development Group, Institut Curie, Centre OCAV PSL Research University, CNRS, UMR168, Inserm, Sorbonne University, Paris, France
| | - Tatiana Merle
- Mechanics and Genetics of Embryonic Development Group, Institut Curie, Centre OCAV PSL Research University, CNRS, UMR168, Inserm, Sorbonne University, Paris, France
| | - Florence Broders-Bondon
- Mechanics and Genetics of Embryonic Development Group, Institut Curie, Centre OCAV PSL Research University, CNRS, UMR168, Inserm, Sorbonne University, Paris, France
| | - Anne-Christine Brunet
- Mechanics and Genetics of Embryonic Development Group, Institut Curie, Centre OCAV PSL Research University, CNRS, UMR168, Inserm, Sorbonne University, Paris, France
| | - Aude Battistella
- Biochemistry, Molecular Biology, and Cells Platform, Institut Curie, CNRS, UMR 168, Inserm, Sorbonne University, Paris, France
| | - Emelie Britt Linnea Land
- Mechanics and Genetics of Embryonic Development Group, Institut Curie, Centre OCAV PSL Research University, CNRS, UMR168, Inserm, Sorbonne University, Paris, France
| | - Florian Sarron
- Sorbonne Université, CNRS, UMR 7095, Institut d'Astrophysique de Paris, Paris, France
| | - Aditya Jha
- Laboratoire Physique et Mécanique des Milieux Hétérogènes (PMMH), CNRS, ESPCI ParisTech, Université Pierre et Marie Curie, Université Paris Diderot, Paris, France
| | - Jean-Luc Gennisson
- Université Paris-Saclay, CEA, CNRS, Inserm, BioMaps, Service Hospitalier Frédéric Joliot, Orsay, France
| | - Eric Röttinger
- Université Côte d’Azur, CNRS, INSERM, Institute for Research on Cancer and Aging (IRCAN), Nice, France
- Université Côte d’Azur, Institut Fédératif de Recherche Ressources Marines (IFR MARRES), Nice, France
| | - María Elena Fernández-Sánchez
- Mechanics and Genetics of Embryonic Development Group, Institut Curie, Centre OCAV PSL Research University, CNRS, UMR168, Inserm, Sorbonne University, Paris, France
| | - Emmanuel Farge
- Mechanics and Genetics of Embryonic Development Group, Institut Curie, Centre OCAV PSL Research University, CNRS, UMR168, Inserm, Sorbonne University, Paris, France
| |
Collapse
|
5
|
Marchant CL, Malmi-Kakkada AN, Espina JA, Barriga EH. Cell clusters softening triggers collective cell migration in vivo. NATURE MATERIALS 2022; 21:1314-1323. [PMID: 35970965 PMCID: PMC9622418 DOI: 10.1038/s41563-022-01323-0] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 06/28/2022] [Indexed: 05/02/2023]
Abstract
Embryogenesis, tissue repair and cancer metastasis rely on collective cell migration. In vitro studies propose that cells are stiffer while migrating in stiff substrates, but softer when plated in compliant surfaces which are typically considered as non-permissive for migration. Here we show that cells within clusters from embryonic tissue dynamically decrease their stiffness in response to the temporal stiffening of their native substrate to initiate collective cell migration. Molecular and mechanical perturbations of embryonic tissues reveal that this unexpected mechanical response involves a mechanosensitive pathway relying on Piezo1-mediated microtubule deacetylation. We further show that decreasing microtubule acetylation and consequently cluster stiffness is sufficient to trigger collective cell migration in soft non-permissive substrates. This suggests that reaching an optimal cluster-to-substrate stiffness ratio is essential to trigger the onset of this collective process. Overall, these in vivo findings challenge the current understanding of collective cell migration and its physiological and pathological roles.
Collapse
Affiliation(s)
- Cristian L Marchant
- Mechanisms of Morphogenesis Laboratory, Gulbenkian Institute of Science (IGC), Oeiras, Portugal
| | - Abdul N Malmi-Kakkada
- Computational Biological Physics Laboratory, Department of Chemistry and Physics, Augusta University, Augusta, GA, USA
| | - Jaime A Espina
- Mechanisms of Morphogenesis Laboratory, Gulbenkian Institute of Science (IGC), Oeiras, Portugal
| | - Elias H Barriga
- Mechanisms of Morphogenesis Laboratory, Gulbenkian Institute of Science (IGC), Oeiras, Portugal.
| |
Collapse
|
6
|
Xie Q, Liu R, Zou Z, Feng Y, Huang Y, Xu G, Sun W, Liang Y, Zhong W. MYPT1 inhibits the metastasis of renal clear cell carcinoma via the MAPK8/N-cadherin pathway. FEBS Open Bio 2022; 12:2083-2095. [PMID: 36106411 PMCID: PMC9623519 DOI: 10.1002/2211-5463.13487] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Revised: 08/23/2022] [Accepted: 09/14/2022] [Indexed: 01/25/2023] Open
Abstract
Myosin phosphatase target subunit 1 (MYPT1) is a subunit of myosin phosphatase that is capable of regulating smooth muscle contraction. MYPT1 has been reported to be involved in a wide variety of tumours, but its expression and biological functions in renal clear cell carcinoma (ccRCC) remain obscure. Herein, we analysed the relationship between patient clinicopathological characteristics and MYPT1 expression levels in ccRCC patients using a tissue microarray (TMA) and data retrieved from the TCGA-KIRC dataset. MYPT1 was overexpressed or depleted using siRNA in ccRCC cells to assess the effects on migration and invasion in vitro and in vivo. Additionally, RNA-sequencing and bioinformatics analysis were performed to investigate the precise mechanism. MYPT1 expression in ccRCC tissues was observed to be lower than that in nonmalignant tissues (P < 0.05). In addition, MYPT1 downregulation was closely linked to advanced pathological stage (P < 0.05), and poor OS (overall survival; P < 0.05). Functionally, increased expression of MYPT1 suppressed ccRCC migration and invasion in vitro, and inhibited tumour metastasis in vivo. In addition, MYPT1 overexpression exerted its suppressive effects via the MAPK8/N-cadherin pathway in ccRCC.
Collapse
Affiliation(s)
- Qingling Xie
- Guangdong Provincial Institute of Nephrology, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
| | - Ren Liu
- Guangdong Provincial Institute of Nephrology, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
| | - Zhihao Zou
- Department of Urology, Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, School of MedicineSouth China University of TechnologyGuangzhouChina
| | - Yuanfa Feng
- Department of Urology, Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, School of MedicineSouth China University of TechnologyGuangzhouChina
| | - Yiqiao Huang
- Department of UrologyThe Fifth Affiliated Hospital of Guangzhou Medical UniversityChina
| | - Guibin Xu
- Department of UrologyThe Fifth Affiliated Hospital of Guangzhou Medical UniversityChina
| | - Wei Sun
- Department of Urology, Huadu District People's HospitalSouthern Medical UniversityGuangzhouChina
| | - Yuxiang Liang
- Department of Urology, Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, School of MedicineSouth China University of TechnologyGuangzhouChina
| | - Weide Zhong
- Guangdong Provincial Institute of Nephrology, Nanfang HospitalSouthern Medical UniversityGuangzhouChina,Department of Urology, Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, School of MedicineSouth China University of TechnologyGuangzhouChina
| |
Collapse
|
7
|
Matsuda M, Chu CW, Sokol SY. Lmo7 recruits myosin II heavy chain to regulate actomyosin contractility and apical domain size in Xenopus ectoderm. Development 2022; 149:275389. [PMID: 35451459 PMCID: PMC9188752 DOI: 10.1242/dev.200236] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 03/30/2022] [Indexed: 11/20/2022]
Abstract
ABSTRACT
Apical constriction, or a reduction in size of the apical domain, underlies many morphogenetic events during development. Actomyosin complexes play an essential role in apical constriction; however, the detailed analysis of molecular mechanisms is still pending. Here, we show that Lim domain only protein 7 (Lmo7), a multidomain adaptor at apical junctions, promotes apical constriction in the Xenopus superficial ectoderm, whereas apical domain size increases in Lmo7-depleted cells. Lmo7 is primarily localized at apical junctions and promotes the formation of the dense circumferential actomyosin belt. Strikingly, Lmo7 binds non-muscle myosin II (NMII) and recruits it to apical junctions and the apical cortex. This NMII recruitment is essential for Lmo7-mediated apical constriction. Lmo7 knockdown decreases NMIIA localization at apical junctions and delays neural tube closure in Xenopus embryos. Our findings suggest that Lmo7 serves as a scaffold that regulates actomyosin contractility and apical domain size.
Collapse
Affiliation(s)
- Miho Matsuda
- Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Chih-Wen Chu
- Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Sergei Y. Sokol
- Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
8
|
Zou Z, Liu R, Liang Y, Zhou R, Dai Q, Han Z, Jiang M, Zhuo Y, Zhang Y, Feng Y, Zhu X, Cai S, Lin J, Tang Z, Zhong W, Liang Y. Identification and Validation of a PPP1R12A-Related Five-Gene Signature Associated With Metabolism to Predict the Prognosis of Patients With Prostate Cancer. Front Genet 2021; 12:703210. [PMID: 34484299 PMCID: PMC8414655 DOI: 10.3389/fgene.2021.703210] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 06/28/2021] [Indexed: 01/23/2023] Open
Abstract
Background Prostate cancer (PCa) is the most common malignant male neoplasm in the American male population. Our prior studies have demonstrated that protein phosphatase 1 regulatory subunit 12A (PPP1R12A) could be an efficient prognostic factor in patients with PCa, promoting further investigation. The present study attempted to construct a gene signature based on PPP1R12A and metabolism-related genes to predict the prognosis of PCa patients. Methods The mRNA expression profiles of 499 tumor and 52 normal tissues were extracted from The Cancer Genome Atlas (TCGA) database. We selected differentially expressed PPP1R12A-related genes among these mRNAs. Tandem affinity purification-mass spectrometry was used to identify the proteins that directly interact with PPP1R12A. Gene set enrichment analysis (GSEA) was used to extract metabolism-related genes. Univariate Cox regression analysis and a random survival forest algorithm were used to confirm optimal genes to build a prognostic risk model. Results We identified a five-gene signature (PPP1R12A, PTGS2, GGCT, AOX1, and NT5E) that was associated with PPP1R12A and metabolism in PCa, which effectively predicted disease-free survival (DFS) and biochemical relapse-free survival (BRFS). Moreover, the signature was validated by two internal datasets from TCGA and one external dataset from the Gene Expression Omnibus (GEO). Conclusion The five-gene signature is an effective potential factor to predict the prognosis of PCa, classifying PCa patients into high- and low-risk groups, which might provide potential novel treatment strategies for these patients.
Collapse
Affiliation(s)
- Zhihao Zou
- Department of Geriatrics, The Second Affiliated Hospital of South China University of Technology, Guangzhou, China.,Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Department of Urology, School of Medicine, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, China
| | - Ren Liu
- Guangdong Provincial Institute of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yingke Liang
- Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Department of Urology, School of Medicine, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, China
| | - Rui Zhou
- Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Department of Urology, School of Medicine, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, China
| | - Qishan Dai
- Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Department of Urology, School of Medicine, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, China
| | - Zhaodong Han
- Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Department of Urology, School of Medicine, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, China
| | - Minyao Jiang
- Department of Urology, Huadu District People's Hospital, Southern Medical University, Guangzhou, China
| | - Yangjia Zhuo
- Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Department of Urology, School of Medicine, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, China
| | - Yixun Zhang
- Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Department of Urology, School of Medicine, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, China
| | - Yuanfa Feng
- Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Department of Urology, School of Medicine, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, China
| | - Xuejin Zhu
- Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Department of Urology, School of Medicine, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, China
| | - Shanghua Cai
- Urology Key Laboratory of Guangdong Province, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
| | - Jundong Lin
- Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Department of Urology, School of Medicine, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, China
| | - Zhenfeng Tang
- Urology Key Laboratory of Guangdong Province, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
| | - Weide Zhong
- Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Department of Urology, School of Medicine, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, China.,Guangdong Provincial Institute of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China.,Urology Key Laboratory of Guangdong Province, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China.,State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau, China
| | - Yuxiang Liang
- Department of Geriatrics, The Second Affiliated Hospital of South China University of Technology, Guangzhou, China.,Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Department of Urology, School of Medicine, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, China.,Department of Urology, Huizhou Municipal Central Hospital, Huizhou, China
| |
Collapse
|
9
|
Zhu Z, Wang J, Tan J, Yao Y, He Z, Xie X, Yan Z, Fu W, Liu Q, Wang Y, Luo T, Bian X. Calcyphosine promotes the proliferation of glioma cells and serves as a potential therapeutic target. J Pathol 2021; 255:374-386. [PMID: 34370292 PMCID: PMC9291001 DOI: 10.1002/path.5776] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Revised: 07/26/2021] [Accepted: 08/05/2021] [Indexed: 12/01/2022]
Abstract
Calcyphosine (CAPS) was initially identified from the canine thyroid. It also exists in many types of tumor, but its expression and function in glioma remain unknown. Here we explored the clinical significance and the functional mechanisms of CAPS in glioma. We found that CAPS was highly expressed in glioma and high expression of CAPS was correlated with poor survival, in glioma patients and public databases. Cox regression analysis showed that CAPS was an independent prognostic factor for glioma patients. Knockdown of CAPS suppressed the proliferation, whereas overexpression of CAPS promoted the proliferation of glioma both in vitro and in vivo. CAPS regulated the G2/M phase transition of the cell cycle, but had no obvious effect on apoptosis. CAPS affected PLK1 phosphorylation through interaction with MYPT1. CAPS knockdown decreased p‐MYPT1 at S507 and p‐PLK1 at S210. Expression of MYPT1 S507 phosphomimic rescued PLK1 phosphorylation and the phenotype caused by CAPS knockdown. The PLK1 inhibitor volasertib enhanced the therapeutic effect of temozolomide in glioma. Our data suggest that CAPS promotes the proliferation of glioma by regulating the cell cycle and the PLK1 inhibitor volasertib might be a chemosensitizer of glioma. © 2021 The Authors. The Journal of Pathology published by John Wiley & Sons, Ltd. on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Zheng Zhu
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University) and Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, China.,Research Department, PLA Rocket Force Characteristic Medical Center, Beijing, China
| | - Jiao Wang
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University) and Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, China
| | - Juan Tan
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University) and Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, China
| | - Yueliang Yao
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University) and Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, China
| | - Zhicheng He
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University) and Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, China
| | - Xiaoqing Xie
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University) and Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, China
| | - Zexuan Yan
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University) and Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, China
| | - Wenjuan Fu
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University) and Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, China
| | - Qing Liu
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University) and Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, China
| | - Yanxia Wang
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University) and Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, China
| | - Tao Luo
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University) and Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, China
| | - Xiuwu Bian
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University) and Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, China
| |
Collapse
|
10
|
Mancini P, Ossipova O, Sokol SY. The dorsal blastopore lip is a source of signals inducing planar cell polarity in the Xenopus neural plate. Biol Open 2021; 10:bio058761. [PMID: 34259326 PMCID: PMC8325942 DOI: 10.1242/bio.058761] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 06/04/2021] [Indexed: 12/03/2022] Open
Abstract
Coordinated polarization of cells in the tissue plane, known as planar cell polarity (PCP), is associated with a signaling pathway critical for the control of morphogenetic processes. Although the segregation of PCP components to opposite cell borders is believed to play a critical role in this pathway, whether PCP derives from egg polarity or preexistent long-range gradient, or forms in response to a localized cue, remains a challenging question. Here we investigate the Xenopus neural plate, a tissue that has been previously shown to exhibit PCP. By imaging Vangl2 and Prickle3, we show that PCP is progressively acquired in the neural plate and requires a signal from the posterior region of the embryo. Tissue transplantations indicated that PCP is triggered in the neural plate by a planar cue from the dorsal blastopore lip. The PCP cue did not depend on the orientation of the graft and was distinct from neural inducers. These observations suggest that neuroectodermal PCP is not instructed by a preexisting molecular gradient but induced by a signal from the dorsal blastopore lip.
Collapse
Affiliation(s)
| | | | - Sergei Y. Sokol
- Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
11
|
Torban E, Sokol SY. Planar cell polarity pathway in kidney development, function and disease. Nat Rev Nephrol 2021; 17:369-385. [PMID: 33547419 PMCID: PMC8967065 DOI: 10.1038/s41581-021-00395-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/07/2021] [Indexed: 02/08/2023]
Abstract
Planar cell polarity (PCP) refers to the coordinated orientation of cells in the tissue plane. Originally discovered and studied in Drosophila melanogaster, PCP is now widely recognized in vertebrates, where it is implicated in organogenesis. Specific sets of PCP genes have been identified. The proteins encoded by these genes become asymmetrically distributed to opposite sides of cells within a tissue plane and guide many processes that include changes in cell shape and polarity, collective cell movements or the uniform distribution of cell appendages. A unifying characteristic of these processes is that they often involve rearrangement of actomyosin. Mutations in PCP genes can cause malformations in organs of many animals, including humans. In the past decade, strong evidence has accumulated for a role of the PCP pathway in kidney development including outgrowth and branching morphogenesis of ureteric bud and podocyte development. Defective PCP signalling has been implicated in the pathogenesis of developmental kidney disorders of the congenital anomalies of the kidney and urinary tract spectrum. Understanding the origins, molecular constituents and cellular targets of PCP provides insights into the involvement of PCP molecules in normal kidney development and how dysfunction of PCP components may lead to kidney disease.
Collapse
Affiliation(s)
- Elena Torban
- McGill University and McGill University Health Center Research Institute, 1001 Boulevard Decarie, Block E, Montreal, Quebec, Canada, H4A3J1.,Corresponding authors: Elena Torban (); Sergei Sokol ()
| | - Sergei Y. Sokol
- Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, One Gustave Levy Place, New York, 10029, USA,Corresponding authors: Elena Torban (); Sergei Sokol ()
| |
Collapse
|
12
|
Chao F, Song Z, Wang S, Ma Z, Zhuo Z, Meng T, Xu G, Chen G. Novel circular RNA circSOBP governs amoeboid migration through the regulation of the miR-141-3p/MYPT1/p-MLC2 axis in prostate cancer. Clin Transl Med 2021; 11:e360. [PMID: 33784000 PMCID: PMC8002909 DOI: 10.1002/ctm2.360] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 03/02/2021] [Accepted: 03/02/2021] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Metastatic prostate cancer is a fatal disease despite multiple new approvals in recent years. Recent studies revealed that circular RNAs (circRNAs) can be involved in cancer metastasis. Defining the role of circRNAs in prostate cancer metastasis and discovering therapeutic targets that block cancer metastasis is of great significance for the treatment of prostate cancer. METHODS The circSOBP levels in prostate cancer (PCa) were determined by qRT-PCR. We evaluated the function of circSOBP using a transwell assay and nude mice lung metastasis models. Immunofluorescence assay and electron microscopic assay were applied to determine the phenotypes of prostate cancer cells' migration. We used fluorescence in situ hybridization assay to determine the localization of RNAs. Dual luciferase and rescue assays were applied to verify the interactions between circSOBP, miR-141-3p, MYPT1, and phosphomyosin light chain (p-MLC2). RESULTS We observed that circSOBP level was significantly lower in PCa specimens compared with adjacent noncancerous prostate specimens, and was correlated with the grade group of PCa. Overexpression of circSOBP suppressed PCa migration and invasion in vitro and metastasis in vivo. CircSOBP depletion increased migration and invasion and induced amoeboid migration of PCa cells. Mechanistically, circSOBP bound miR-141-3p and regulated the MYPT1/p-MLC2 axis. Moreover, the depletion of MYPT1 reversed the inhibitory effect of circSOBP on the migration and invasion of PCa cells. Complementary intronic Alu elements induced but were not necessary for the formation of circSOBP. The nuclear export of circSOBP was mediated by URH49. CONCLUSION Our results suggest that circSOBP suppresses amoeboid migration of PCa cells and inhibits migration and invasion through sponging miR-141-3p and regulating the MYPT1/p-MLC2 axis.
Collapse
Affiliation(s)
- Fan Chao
- Department of UrologyJinshan HospitalFudan UniversityShanghaiP. R. China
- Department of SurgeryShanghai Medical CollegeFudan UniversityShanghaiP. R. China
| | - Zhenyu Song
- Department of UrologyJinshan HospitalFudan UniversityShanghaiP. R. China
| | - Shiyu Wang
- Department of UrologyJinshan HospitalFudan UniversityShanghaiP. R. China
- Department of SurgeryShanghai Medical CollegeFudan UniversityShanghaiP. R. China
| | - Zhe Ma
- Department of UrologyJinshan HospitalFudan UniversityShanghaiP. R. China
| | - Zhiyuan Zhuo
- Department of UrologyJinshan HospitalFudan UniversityShanghaiP. R. China
| | - Ting Meng
- Research Center for Clinical MedicineJinshan HospitalFudan UniversityShanghaiP. R. China
| | - Guoxiong Xu
- Research Center for Clinical MedicineJinshan HospitalFudan UniversityShanghaiP. R. China
| | - Gang Chen
- Department of UrologyJinshan HospitalFudan UniversityShanghaiP. R. China
- Department of SurgeryShanghai Medical CollegeFudan UniversityShanghaiP. R. China
| |
Collapse
|
13
|
Godard BG, Dumollard R, Munro E, Chenevert J, Hebras C, McDougall A, Heisenberg CP. Apical Relaxation during Mitotic Rounding Promotes Tension-Oriented Cell Division. Dev Cell 2020; 55:695-706.e4. [PMID: 33207225 DOI: 10.1016/j.devcel.2020.10.016] [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: 04/21/2020] [Revised: 09/09/2020] [Accepted: 10/23/2020] [Indexed: 12/16/2022]
Abstract
Global tissue tension anisotropy has been shown to trigger stereotypical cell division orientation by elongating mitotic cells along the main tension axis. Yet, how tissue tension elongates mitotic cells despite those cells undergoing mitotic rounding (MR) by globally upregulating cortical actomyosin tension remains unclear. We addressed this question by taking advantage of ascidian embryos, consisting of a small number of interphasic and mitotic blastomeres and displaying an invariant division pattern. We found that blastomeres undergo MR by locally relaxing cortical tension at their apex, thereby allowing extrinsic pulling forces from neighboring interphasic blastomeres to polarize their shape and thus division orientation. Consistently, interfering with extrinsic forces by reducing the contractility of interphasic blastomeres or disrupting the establishment of asynchronous mitotic domains leads to aberrant mitotic cell division orientations. Thus, apical relaxation during MR constitutes a key mechanism by which tissue tension anisotropy controls stereotypical cell division orientation.
Collapse
Affiliation(s)
- Benoit G Godard
- Laboratoire de Biologie du Développement de Villefranche-sur-mer, Institut de la Mer de Villefranche-sur-mer, Sorbonne Université, CNRS, 06230 Villefranche-sur-mer, France; Institute of Science and Technology Austria, Klosterneuburg, Austria
| | - Rémi Dumollard
- Laboratoire de Biologie du Développement de Villefranche-sur-mer, Institut de la Mer de Villefranche-sur-mer, Sorbonne Université, CNRS, 06230 Villefranche-sur-mer, France
| | - Edwin Munro
- Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, IL 60637, USA
| | - Janet Chenevert
- Laboratoire de Biologie du Développement de Villefranche-sur-mer, Institut de la Mer de Villefranche-sur-mer, Sorbonne Université, CNRS, 06230 Villefranche-sur-mer, France
| | - Céline Hebras
- Laboratoire de Biologie du Développement de Villefranche-sur-mer, Institut de la Mer de Villefranche-sur-mer, Sorbonne Université, CNRS, 06230 Villefranche-sur-mer, France
| | - Alex McDougall
- Laboratoire de Biologie du Développement de Villefranche-sur-mer, Institut de la Mer de Villefranche-sur-mer, Sorbonne Université, CNRS, 06230 Villefranche-sur-mer, France
| | | |
Collapse
|
14
|
Sarmasti Emami S, Zhang D, Yang X. Interaction of the Hippo Pathway and Phosphatases in Tumorigenesis. Cancers (Basel) 2020; 12:E2438. [PMID: 32867200 PMCID: PMC7564220 DOI: 10.3390/cancers12092438] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 08/21/2020] [Accepted: 08/25/2020] [Indexed: 01/05/2023] Open
Abstract
The Hippo pathway is an emerging tumor suppressor signaling pathway involved in a wide range of cellular processes. Dysregulation of different components of the Hippo signaling pathway is associated with a number of diseases including cancer. Therefore, identification of the Hippo pathway regulators and the underlying mechanism of its regulation may be useful to uncover new therapeutics for cancer therapy. The Hippo signaling pathway includes a set of kinases that phosphorylate different proteins in order to phosphorylate and inactivate its main downstream effectors, YAP and TAZ. Thus, modulating phosphorylation and dephosphorylation of the Hippo components by kinases and phosphatases play critical roles in the regulation of the signaling pathway. While information regarding kinase regulation of the Hippo pathway is abundant, the role of phosphatases in regulating this pathway is just beginning to be understood. In this review, we summarize the most recent reports on the interaction of phosphatases and the Hippo pathway in tumorigenesis. We have also introduced challenges in clarifying the role of phosphatases in the Hippo pathway and future direction of crosstalk between phosphatases and the Hippo pathway.
Collapse
Affiliation(s)
| | | | - Xiaolong Yang
- Department of Pathology and Molecular Medicine, Queen’s University, Kingston, ON K7L 3N6, Canada; (S.S.E.); (D.Z.)
| |
Collapse
|
15
|
Lang I, Virk G, Zheng DC, Young J, Nguyen MJ, Amiri R, Fong M, Arata A, Chadaideh KS, Walsh S, Weiser DC. The Evolution of Duplicated Genes of the Cpi-17/Phi-1 ( ppp1r14) Family of Protein Phosphatase 1 Inhibitors in Teleosts. Int J Mol Sci 2020; 21:ijms21165709. [PMID: 32784920 PMCID: PMC7460850 DOI: 10.3390/ijms21165709] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Accepted: 08/07/2020] [Indexed: 11/29/2022] Open
Abstract
The Cpi-17 (ppp1r14) gene family is an evolutionarily conserved, vertebrate specific group of protein phosphatase 1 (PP1) inhibitors. When phosphorylated, Cpi-17 is a potent inhibitor of myosin phosphatase (MP), a holoenzyme complex of the regulatory subunit Mypt1 and the catalytic subunit PP1. Myosin phosphatase dephosphorylates the regulatory myosin light chain (Mlc2) and promotes actomyosin relaxation, which in turn, regulates numerous cellular processes including smooth muscle contraction, cytokinesis, cell motility, and tumor cell invasion. We analyzed zebrafish homologs of the Cpi-17 family, to better understand the mechanisms of myosin phosphatase regulation. We found single homologs of both Kepi (ppp1r14c) and Gbpi (ppp1r14d) in silico, but we detected no expression of these genes during early embryonic development. Cpi-17 (ppp1r14a) and Phi-1 (ppp1r14b) each had two duplicate paralogs, (ppp1r14aa and ppp1r14ab) and (ppp1r14ba and ppp1r14bb), which were each expressed during early development. The spatial expression pattern of these genes has diverged, with ppp1r14aa and ppp1r14bb expressed primarily in smooth muscle and skeletal muscle, respectively, while ppp1r14ab and ppp1r14ba are primarily expressed in neural tissue. We observed that, in in vitro and heterologous cellular systems, the Cpi-17 paralogs both acted as potent myosin phosphatase inhibitors, and were indistinguishable from one another. In contrast, the two Phi-1 paralogs displayed weak myosin phosphatase inhibitory activity in vitro, and did not alter myosin phosphorylation in cells. Through deletion and chimeric analysis, we identified that the difference in specificity for myosin phosphatase between Cpi-17 and Phi-1 was encoded by the highly conserved PHIN (phosphatase holoenzyme inhibitory) domain, and not the more divergent N- and C- termini. We also showed that either Cpi-17 paralog can rescue the knockdown phenotype, but neither Phi-1 paralog could do so. Thus, we provide new evidence about the biochemical and developmental distinctions of the zebrafish Cpi-17 protein family.
Collapse
Affiliation(s)
- Irene Lang
- Department of Biological Sciences, University of the Pacific, Stockton, CA 98211, USA; (I.L.); (G.V.); (D.C.Z.); (J.Y.); (M.J.N.); (R.A.); (M.F.); (A.A.); (K.S.C.)
| | - Guneet Virk
- Department of Biological Sciences, University of the Pacific, Stockton, CA 98211, USA; (I.L.); (G.V.); (D.C.Z.); (J.Y.); (M.J.N.); (R.A.); (M.F.); (A.A.); (K.S.C.)
| | - Dale C. Zheng
- Department of Biological Sciences, University of the Pacific, Stockton, CA 98211, USA; (I.L.); (G.V.); (D.C.Z.); (J.Y.); (M.J.N.); (R.A.); (M.F.); (A.A.); (K.S.C.)
| | - Jason Young
- Department of Biological Sciences, University of the Pacific, Stockton, CA 98211, USA; (I.L.); (G.V.); (D.C.Z.); (J.Y.); (M.J.N.); (R.A.); (M.F.); (A.A.); (K.S.C.)
| | - Michael J. Nguyen
- Department of Biological Sciences, University of the Pacific, Stockton, CA 98211, USA; (I.L.); (G.V.); (D.C.Z.); (J.Y.); (M.J.N.); (R.A.); (M.F.); (A.A.); (K.S.C.)
| | - Rojin Amiri
- Department of Biological Sciences, University of the Pacific, Stockton, CA 98211, USA; (I.L.); (G.V.); (D.C.Z.); (J.Y.); (M.J.N.); (R.A.); (M.F.); (A.A.); (K.S.C.)
| | - Michelle Fong
- Department of Biological Sciences, University of the Pacific, Stockton, CA 98211, USA; (I.L.); (G.V.); (D.C.Z.); (J.Y.); (M.J.N.); (R.A.); (M.F.); (A.A.); (K.S.C.)
| | - Alisa Arata
- Department of Biological Sciences, University of the Pacific, Stockton, CA 98211, USA; (I.L.); (G.V.); (D.C.Z.); (J.Y.); (M.J.N.); (R.A.); (M.F.); (A.A.); (K.S.C.)
| | - Katia S. Chadaideh
- Department of Biological Sciences, University of the Pacific, Stockton, CA 98211, USA; (I.L.); (G.V.); (D.C.Z.); (J.Y.); (M.J.N.); (R.A.); (M.F.); (A.A.); (K.S.C.)
- Department of Human Evolutionary Biology, Harvard University, Cambridge, MA 02138, USA
| | - Susan Walsh
- Life Sciences, Soka University of America, Aliso Viejo, CA 92656, USA;
| | - Douglas C. Weiser
- Department of Biological Sciences, University of the Pacific, Stockton, CA 98211, USA; (I.L.); (G.V.); (D.C.Z.); (J.Y.); (M.J.N.); (R.A.); (M.F.); (A.A.); (K.S.C.)
- Correspondence: ; Tel.: +1-209-946-2955
| |
Collapse
|
16
|
Chen Y, Kotian N, Aranjuez G, Chen L, Messer CL, Burtscher A, Sawant K, Ramel D, Wang X, McDonald JA. Protein phosphatase 1 activity controls a balance between collective and single cell modes of migration. eLife 2020; 9:52979. [PMID: 32369438 PMCID: PMC7200163 DOI: 10.7554/elife.52979] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Accepted: 04/23/2020] [Indexed: 12/12/2022] Open
Abstract
Collective cell migration is central to many developmental and pathological processes. However, the mechanisms that keep cell collectives together and coordinate movement of multiple cells are poorly understood. Using the Drosophila border cell migration model, we find that Protein phosphatase 1 (Pp1) activity controls collective cell cohesion and migration. Inhibition of Pp1 causes border cells to round up, dissociate, and move as single cells with altered motility. We present evidence that Pp1 promotes proper levels of cadherin-catenin complex proteins at cell-cell junctions within the cluster to keep border cells together. Pp1 further restricts actomyosin contractility to the cluster periphery rather than at individual internal border cell contacts. We show that the myosin phosphatase Pp1 complex, which inhibits non-muscle myosin-II (Myo-II) activity, coordinates border cell shape and cluster cohesion. Given the high conservation of Pp1 complexes, this study identifies Pp1 as a major regulator of collective versus single cell migration.
Collapse
Affiliation(s)
- Yujun Chen
- Division of Biology, Kansas State University, Manhattan, United States
| | - Nirupama Kotian
- Division of Biology, Kansas State University, Manhattan, United States
| | - George Aranjuez
- Lerner Research Institute, Cleveland Clinic, Cleveland, United States
| | - Lin Chen
- LBCMCP, Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPS, Toulouse, France
| | - C Luke Messer
- Division of Biology, Kansas State University, Manhattan, United States
| | - Ashley Burtscher
- Lerner Research Institute, Cleveland Clinic, Cleveland, United States
| | - Ketki Sawant
- Division of Biology, Kansas State University, Manhattan, United States
| | - Damien Ramel
- LBCMCP, Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Xiaobo Wang
- LBCMCP, Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPS, Toulouse, France
| | | |
Collapse
|
17
|
Muñoz-Galván S, Felipe-Abrio B, Verdugo-Sivianes EM, Perez M, Jiménez-García MP, Suarez-Martinez E, Estevez-Garcia P, Carnero A. Downregulation of MYPT1 increases tumor resistance in ovarian cancer by targeting the Hippo pathway and increasing the stemness. Mol Cancer 2020; 19:7. [PMID: 31926547 PMCID: PMC6954568 DOI: 10.1186/s12943-020-1130-z] [Citation(s) in RCA: 88] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2019] [Accepted: 01/01/2020] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Ovarian cancer is one of the most common and malignant cancers, partly due to its late diagnosis and high recurrence. Chemotherapy resistance has been linked to poor prognosis and is believed to be linked to the cancer stem cell (CSC) pool. Therefore, elucidating the molecular mechanisms mediating therapy resistance is essential to finding new targets for therapy-resistant tumors. METHODS shRNA depletion of MYPT1 in ovarian cancer cell lines, miRNA overexpression, RT-qPCR analysis, patient tumor samples, cell line- and tumorsphere-derived xenografts, in vitro and in vivo treatments, analysis of data from ovarian tumors in public transcriptomic patient databases and in-house patient cohorts. RESULTS We show that MYPT1 (PPP1R12A), encoding myosin phosphatase target subunit 1, is downregulated in ovarian tumors, leading to reduced survival and increased tumorigenesis, as well as resistance to platinum-based therapy. Similarly, overexpression of miR-30b targeting MYPT1 results in enhanced CSC-like properties in ovarian tumor cells and is connected to the activation of the Hippo pathway. Inhibition of the Hippo pathway transcriptional co-activator YAP suppresses the resistance to platinum-based therapy induced by either low MYPT1 expression or miR-30b overexpression, both in vitro and in vivo. CONCLUSIONS Our work provides a functional link between the resistance to chemotherapy in ovarian tumors and the increase in the CSC pool that results from the activation of the Hippo pathway target genes upon MYPT1 downregulation. Combination therapy with cisplatin and YAP inhibitors suppresses MYPT1-induced resistance, demonstrating the possibility of using this treatment in patients with low MYPT1 expression, who are likely to be resistant to platinum-based therapy.
Collapse
Affiliation(s)
- Sandra Muñoz-Galván
- Instituto de Biomedicina de Sevilla, IBIS, Hospital Universitario Virgen del Rocío, Universidad de Sevilla, Consejo Superior de Investigaciones Científicas, Avda. Manuel Siurot s/n 41013, Seville, Spain.,CIBERONC, Instituto de Salud Carlos III, Madrid, Spain
| | - Blanca Felipe-Abrio
- Instituto de Biomedicina de Sevilla, IBIS, Hospital Universitario Virgen del Rocío, Universidad de Sevilla, Consejo Superior de Investigaciones Científicas, Avda. Manuel Siurot s/n 41013, Seville, Spain.,CIBERONC, Instituto de Salud Carlos III, Madrid, Spain
| | - Eva M Verdugo-Sivianes
- Instituto de Biomedicina de Sevilla, IBIS, Hospital Universitario Virgen del Rocío, Universidad de Sevilla, Consejo Superior de Investigaciones Científicas, Avda. Manuel Siurot s/n 41013, Seville, Spain.,CIBERONC, Instituto de Salud Carlos III, Madrid, Spain
| | - Marco Perez
- Instituto de Biomedicina de Sevilla, IBIS, Hospital Universitario Virgen del Rocío, Universidad de Sevilla, Consejo Superior de Investigaciones Científicas, Avda. Manuel Siurot s/n 41013, Seville, Spain.,CIBERONC, Instituto de Salud Carlos III, Madrid, Spain
| | - Manuel P Jiménez-García
- Instituto de Biomedicina de Sevilla, IBIS, Hospital Universitario Virgen del Rocío, Universidad de Sevilla, Consejo Superior de Investigaciones Científicas, Avda. Manuel Siurot s/n 41013, Seville, Spain.,CIBERONC, Instituto de Salud Carlos III, Madrid, Spain
| | - Elisa Suarez-Martinez
- Instituto de Biomedicina de Sevilla, IBIS, Hospital Universitario Virgen del Rocío, Universidad de Sevilla, Consejo Superior de Investigaciones Científicas, Avda. Manuel Siurot s/n 41013, Seville, Spain.,CIBERONC, Instituto de Salud Carlos III, Madrid, Spain
| | - Purificacion Estevez-Garcia
- Instituto de Biomedicina de Sevilla, IBIS, Hospital Universitario Virgen del Rocío, Universidad de Sevilla, Consejo Superior de Investigaciones Científicas, Avda. Manuel Siurot s/n 41013, Seville, Spain.,CIBERONC, Instituto de Salud Carlos III, Madrid, Spain
| | - Amancio Carnero
- Instituto de Biomedicina de Sevilla, IBIS, Hospital Universitario Virgen del Rocío, Universidad de Sevilla, Consejo Superior de Investigaciones Científicas, Avda. Manuel Siurot s/n 41013, Seville, Spain. .,CIBERONC, Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
18
|
Loss-of-Function Variants in PPP1R12A: From Isolated Sex Reversal to Holoprosencephaly Spectrum and Urogenital Malformations. Am J Hum Genet 2020; 106:121-128. [PMID: 31883643 DOI: 10.1016/j.ajhg.2019.12.004] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Accepted: 12/04/2019] [Indexed: 02/01/2023] Open
Abstract
In two independent ongoing next-generation sequencing projects for individuals with holoprosencephaly and individuals with disorders of sex development, and through international research collaboration, we identified twelve individuals with de novo loss-of-function (LoF) variants in protein phosphatase 1, regulatory subunit 12a (PPP1R12A), an important developmental gene involved in cell migration, adhesion, and morphogenesis. This gene has not been previously reported in association with human disease, and it has intolerance to LoF as illustrated by a very low observed-to-expected ratio of LoF variants in gnomAD. Of the twelve individuals, midline brain malformations were found in five, urogenital anomalies in nine, and a combination of both phenotypes in two. Other congenital anomalies identified included omphalocele, jejunal, and ileal atresia with aberrant mesenteric blood supply, and syndactyly. Six individuals had stop gain variants, five had a deletion or duplication resulting in a frameshift, and one had a canonical splice acceptor site loss. Murine and human in situ hybridization and immunostaining revealed PPP1R12A expression in the prosencephalic neural folds and protein localization in the lower urinary tract at critical periods for forebrain division and urogenital development. Based on these clinical and molecular findings, we propose the association of PPP1R12A pathogenic variants with a congenital malformations syndrome affecting the embryogenesis of the brain and genitourinary systems and including disorders of sex development.
Collapse
|
19
|
Williams ML, Solnica-Krezel L. Cellular and molecular mechanisms of convergence and extension in zebrafish. Curr Top Dev Biol 2020; 136:377-407. [DOI: 10.1016/bs.ctdb.2019.08.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
20
|
Chikina AS, Rubtsova SN, Lomakina ME, Potashnikova DM, Vorobjev IA, Alexandrova AY. Transition from mesenchymal to bleb-based motility is predominantly exhibited by CD133-positive subpopulation of fibrosarcoma cells. Biol Cell 2019; 111:245-261. [PMID: 31403697 DOI: 10.1111/boc.201800078] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 07/22/2019] [Accepted: 08/06/2019] [Indexed: 01/11/2023]
Abstract
BACKGROUND INFORMATION Metastatic disease is caused by the ability of cancer cells to reach distant organs and form secondary lesions at new locations. Dissemination of cancer cells depends on their migration plasticity - an ability to switch between motility modes driven by distinct molecular machineries. One of such switches is mesenchymal-to-amoeboid transition. Although mesenchymal migration of individual cells requires Arp2/3-dependent actin polymerisation, amoeboid migration is characterised by a high level of actomyosin contractility and often involves the formation of membrane blebs. The acquisition of amoeboid motility by mesenchymal cells is often associated with enhanced metastasis. RESULTS We studied the ability of mesenchymal HT1080 fibrosarcoma cells to switch to amoeboid motility. We induced the transition from lamellipodium-rich to blebbing phenotype either by down-regulating the Arp2/3 complex, pharmacologically or by RNAi, or by decreasing substrate adhesiveness. Each of these treatments induced blebbing in a subset of fibrosarcoma cells, but not in normal subcutaneous fibroblasts. A significant fraction of HT1080 cells that switched to blebbing behaviour exhibited stem cell-like features, such as expression of the stem cell marker CD133, an increased efflux of Hoechst-33342 and positive staining for Oct4, Sox2 and Nanog. Furthermore, the isolated CD133+ cells demonstrated an increased ability to switch to bleb-rich amoeboid phenotype both under inhibitor's treatment and in 3D collagen gels. CONCLUSIONS Together, our data show a significant correlation between the increased ability of cells to switch between migration modes and their stem-like features, suggesting that migration plasticity is an additional property of stem-like population of fibrosarcoma cells. This combination of features could facilitate both dissemination of these cells to distant locations, and their establishment self-renewal in a new microenvironment, as required for metastasis formation. SIGNIFICANCE These data suggest that migration plasticity is a new feature of cancer stem-like cells that can significantly facilitate their dissemination to a secondary location by allowing them to adapt quickly to challenging microenvironments. Moreover, it complements their resistance to apoptosis and self-renewal potential, thus enabling them not only to disseminate efficiently, but also to survive and colonise new niches.
Collapse
Affiliation(s)
- Aleksandra S Chikina
- N.N. Blokhin National Medical Research Center of Oncology, Moscow, 115478, Russia
| | - Svetlana N Rubtsova
- N.N. Blokhin National Medical Research Center of Oncology, Moscow, 115478, Russia
| | - Maria E Lomakina
- N.N. Blokhin National Medical Research Center of Oncology, Moscow, 115478, Russia
| | - Daria M Potashnikova
- Department of Biology and A.N. Belozersky Institute of Physico-Chemical Biology, M.V. Lomonosov Moscow State University, Moscow, 119992, Russia
| | - Ivan A Vorobjev
- Department of Biology, School of Science and Technology, Nazarbayev University, Astana, 010000, Kazakhstan
| | | |
Collapse
|
21
|
Organization of Embryonic Morphogenesis via Mechanical Information. Dev Cell 2019; 49:829-839.e5. [PMID: 31178400 DOI: 10.1016/j.devcel.2019.05.014] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 03/20/2019] [Accepted: 05/03/2019] [Indexed: 01/19/2023]
Abstract
Embryonic organizers establish gradients of diffusible signaling molecules to pattern the surrounding cells. Here, we elucidate an additional mechanism of embryonic organizers that is a secondary consequence of morphogen signaling. Using pharmacological and localized transgenic perturbations, 4D imaging of the zebrafish embryo, systematic analysis of cell motion, and computational modeling, we find that the vertebrate tail organizer orchestrates morphogenesis over distances beyond the range of morphogen signaling. The organizer regulates the rate and coherence of cell motion in the elongating embryo using mechanical information that is transmitted via relay between neighboring cells. This mechanism is similar to a pressure front in granular media and other jammed systems, but in the embryo the mechanical information emerges from self-propelled cell movement and not force transfer between cells. The propagation likely relies upon local biochemical signaling that affects cell contractility, cell adhesion, and/or cell polarity but is independent of transcription and translation.
Collapse
|
22
|
Samson SC, Elliott A, Mueller BD, Kim Y, Carney KR, Bergman JP, Blenis J, Mendoza MC. p90 ribosomal S6 kinase (RSK) phosphorylates myosin phosphatase and thereby controls edge dynamics during cell migration. J Biol Chem 2019; 294:10846-10862. [PMID: 31138649 PMCID: PMC6635457 DOI: 10.1074/jbc.ra119.007431] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 05/08/2019] [Indexed: 12/25/2022] Open
Abstract
Cell migration is essential to embryonic development, wound healing, and cancer cell dissemination. Cells move via leading-edge protrusion, substrate adhesion, and retraction of the cell's rear. The molecular mechanisms by which extracellular cues signal to the actomyosin cytoskeleton to control these motility mechanics are poorly understood. The growth factor-responsive and oncogenically activated protein extracellular signal-regulated kinase (ERK) promotes motility by signaling in actin polymerization-mediated edge protrusion. Using a combination of immunoblotting, co-immunoprecipitation, and myosin-binding experiments and cell migration assays, we show here that ERK also signals to the contractile machinery through its substrate, p90 ribosomal S6 kinase (RSK). We probed the signaling and migration dynamics of multiple mammalian cell lines and found that RSK phosphorylates myosin phosphatase–targeting subunit 1 (MYPT1) at Ser-507, which promotes an interaction of Rho kinase (ROCK) with MYPT1 and inhibits myosin targeting. We find that by inhibiting the myosin phosphatase, ERK and RSK promote myosin II–mediated tension for lamella expansion and optimal edge dynamics for cell migration. These findings suggest that ERK activity can coordinately amplify both protrusive and contractile forces for optimal cell motility.
Collapse
Affiliation(s)
- Shiela C Samson
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah 84112 and
| | - Andrew Elliott
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah 84112 and
| | - Brian D Mueller
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah 84112 and
| | - Yung Kim
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah 84112 and
| | - Keith R Carney
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah 84112 and
| | - Jared P Bergman
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah 84112 and
| | - John Blenis
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts 02115
| | - Michelle C Mendoza
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah 84112 and; Department of Cell Biology, Harvard Medical School, Boston, Massachusetts 02115.
| |
Collapse
|
23
|
Shamipour S, Kardos R, Xue SL, Hof B, Hannezo E, Heisenberg CP. Bulk Actin Dynamics Drive Phase Segregation in Zebrafish Oocytes. Cell 2019; 177:1463-1479.e18. [PMID: 31080065 DOI: 10.1016/j.cell.2019.04.030] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 02/07/2019] [Accepted: 04/12/2019] [Indexed: 10/26/2022]
Abstract
Segregation of maternal determinants within the oocyte constitutes the first step in embryo patterning. In zebrafish oocytes, extensive ooplasmic streaming leads to the segregation of ooplasm from yolk granules along the animal-vegetal axis of the oocyte. Here, we show that this process does not rely on cortical actin reorganization, as previously thought, but instead on a cell-cycle-dependent bulk actin polymerization wave traveling from the animal to the vegetal pole of the oocyte. This wave functions in segregation by both pulling ooplasm animally and pushing yolk granules vegetally. Using biophysical experimentation and theory, we show that ooplasm pulling is mediated by bulk actin network flows exerting friction forces on the ooplasm, while yolk granule pushing is achieved by a mechanism closely resembling actin comet formation on yolk granules. Our study defines a novel role of cell-cycle-controlled bulk actin polymerization waves in oocyte polarization via ooplasmic segregation.
Collapse
Affiliation(s)
- Shayan Shamipour
- Institute of Science and Technology Austria, Klosterneuburg, Austria
| | - Roland Kardos
- Institute of Science and Technology Austria, Klosterneuburg, Austria
| | - Shi-Lei Xue
- Institute of Science and Technology Austria, Klosterneuburg, Austria
| | - Björn Hof
- Institute of Science and Technology Austria, Klosterneuburg, Austria
| | - Edouard Hannezo
- Institute of Science and Technology Austria, Klosterneuburg, Austria.
| | | |
Collapse
|
24
|
The Cytoskeleton-A Complex Interacting Meshwork. Cells 2019; 8:cells8040362. [PMID: 31003495 PMCID: PMC6523135 DOI: 10.3390/cells8040362] [Citation(s) in RCA: 232] [Impact Index Per Article: 38.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 04/15/2019] [Accepted: 04/15/2019] [Indexed: 12/22/2022] Open
Abstract
The cytoskeleton of animal cells is one of the most complicated and functionally versatile structures, involved in processes such as endocytosis, cell division, intra-cellular transport, motility, force transmission, reaction to external forces, adhesion and preservation, and adaptation of cell shape. These functions are mediated by three classical cytoskeletal filament types, as follows: Actin, microtubules, and intermediate filaments. The named filaments form a network that is highly structured and dynamic, responding to external and internal cues with a quick reorganization that is orchestrated on the time scale of minutes and has to be tightly regulated. Especially in brain tumors, the cytoskeleton plays an important role in spreading and migration of tumor cells. As the cytoskeletal organization and regulation is complex and many-faceted, this review aims to summarize the findings about cytoskeletal filament types, including substructures formed by them, such as lamellipodia, stress fibers, and interactions between intermediate filaments, microtubules and actin. Additionally, crucial regulatory aspects of the cytoskeletal filaments and the formed substructures are discussed and integrated into the concepts of cell motility. Even though little is known about the impact of cytoskeletal alterations on the progress of glioma, a final point discussed will be the impact of established cytoskeletal alterations in the cellular behavior and invasion of glioma.
Collapse
|
25
|
LaFlamme A, Young KE, Lang I, Weiser DC. Alternative splicing of (ppp1r12a/mypt1) in zebrafish produces a novel myosin phosphatase targeting subunit. Gene 2018; 675:15-26. [PMID: 29960069 PMCID: PMC6123272 DOI: 10.1016/j.gene.2018.06.092] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2018] [Revised: 06/07/2018] [Accepted: 06/26/2018] [Indexed: 01/04/2023]
Abstract
Myosin phosphatase is an evolutionarily conserved regulator of actomyosin contractility, comprised of a regulatory subunit (Mypt1), and a catalytic subunit (PP1). Zebrafish has become an ideal model organism for the study of the genetic and cell physiological role of the myosin phosphatase in morphogenesis and embryonic development. We identified and characterized a novel splice variant of Mypt1 (ppp1r12a-tv202) from zebrafish, which is widely expressed during early embryonic development. Importantly, mutant alleles and antisense morpholinos that have been used to demonstrate the important role of Mypt1 in early development, not only disrupt the longer splice variants, but also tv202. The protein product of ppp1r12a-tv202 (Mypt1-202) contains the PP1-binding N-terminus, but lacks the regulatory C-terminus, which contains two highly conserved inhibitory phosphorylation sites. We observed that the protein product of tv202 assembled a constitutively active myosin phosphatase uninhibited by kinases such as Zipk. Thus, we propose that Mypt1-202 plays an important role in maintaining baseline Mlc2 dephosphorylation and actomyosin relaxation during early zebrafish development.
Collapse
Affiliation(s)
- Andrew LaFlamme
- Department of Biological Sciences, University of the Pacific, Stockton, CA 95211, USA
| | - Kyle E Young
- Department of Biological Sciences, University of the Pacific, Stockton, CA 95211, USA
| | - Irene Lang
- Department of Biological Sciences, University of the Pacific, Stockton, CA 95211, USA
| | - Douglas C Weiser
- Department of Biological Sciences, University of the Pacific, Stockton, CA 95211, USA.
| |
Collapse
|
26
|
Chu CW, Xiang B, Ossipova O, Ioannou A, Sokol SY. The Ajuba family protein Wtip regulates actomyosin contractility during vertebrate neural tube closure. J Cell Sci 2018; 131:jcs.213884. [PMID: 29661847 DOI: 10.1242/jcs.213884] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Accepted: 04/06/2018] [Indexed: 12/16/2022] Open
Abstract
Ajuba family proteins are implicated in the assembly of cell junctions and have been reported to antagonize Hippo signaling in response to cytoskeletal tension. To assess the role of these proteins in actomyosin contractility, we examined the localization and function of Wtip, a member of the Ajuba family, in Xenopus early embryos. Targeted in vivo depletion of Wtip inhibited apical constriction in neuroepithelial cells and elicited neural tube defects. Fluorescent protein-tagged Wtip showed predominant punctate localization along the cell junctions in the epidermis and a linear junctional pattern in the neuroectoderm. In cells undergoing Shroom3-induced apical constriction, the punctate distribution was reorganized into a linear pattern. Conversely, the linear junctional pattern of Wtip in neuroectoderm changed to a more punctate distribution in cells with reduced myosin II activity. The C-terminal fragment of Wtip physically associated with Shroom3 and interfered with Shroom3 activity and neural fold formation. We therefore propose that Wtip is a tension-sensitive cytoskeletal adaptor that regulates apical constriction during vertebrate neurulation.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Chih-Wen Chu
- Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Bo Xiang
- Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Olga Ossipova
- Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Andriani Ioannou
- Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Sergei Y Sokol
- Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
27
|
Tissue stiffening coordinates morphogenesis by triggering collective cell migration in vivo. Nature 2018; 554:523-527. [PMID: 29443958 PMCID: PMC6013044 DOI: 10.1038/nature25742] [Citation(s) in RCA: 350] [Impact Index Per Article: 50.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Accepted: 01/11/2018] [Indexed: 01/04/2023]
Abstract
Collective cell migration (CCM) is essential for morphogenesis, tissue remodelling, and cancer invasion1,2. In vivo, groups of cells move in an orchestrated way through tissues. This movement requires forces and involves mechanical as well as molecular interactions between cells and their environment. While the role of molecular signals in CCM is comparatively well understood1,2, how tissue mechanics influence CCM in vivo remains unknown. Here we investigated the importance of mechanical cues in the collective migration of the Xenopus laevis neural crest cells, an embryonic cell population whose migratory behaviour has been likened to cancer invasion3. We found that, during morphogenesis, the head mesoderm underlying the cephalic neural crest stiffens. This stiffening initiated an epithelial-to-mesenchymal transition (EMT) in neural crest cells and triggered their collective migration. To detect changes in their mechanical environment, neural crest use integrin/vinculin/talin-mediated mechanosensing. By performing mechanical and molecular manipulations, we showed that mesoderm stiffening is necessary and sufficient to trigger neural crest migration. Finally, we demonstrated that convergent extension of the mesoderm, which starts during gastrulation, leads to increased mesoderm stiffness by increasing the cell density underneath the neural crest. These results unveil a novel role for mesodermal convergent extension as a mechanical coordinator of morphogenesis, and thus reveal a new link between two apparently unconnected processes, gastrulation and neural crest migration, via changes in tissue mechanics. Overall, we provide the first demonstration that changes in substrate stiffness can trigger CCM by promoting EMT in vivo. More broadly, our results raise the exciting idea that tissue mechanics combines with molecular effectors to coordinate morphogenesis4.
Collapse
|
28
|
Pasiliao CC, Hopyan S. Cell ingression: Relevance to limb development and for adaptive evolution. Genesis 2017; 56. [PMID: 29280270 DOI: 10.1002/dvg.23086] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2017] [Revised: 11/16/2017] [Accepted: 12/05/2017] [Indexed: 12/11/2022]
Abstract
Cell ingression is an out-of-plane type of cell intercalation that is essential for the formation of multiple embryonic structures including the limbs. In particular, cell ingression underlies epithelial-to-mesenchymal transition of lateral plate cells to initiate limb bud growth, delamination of neural crest cells to generate peripheral nerve sheaths, and emigration of myoblasts from somites to assemble muscles. Individual cells that ingress undergo apical constriction to generate bottle shaped cells, diminish adhesion to their epithelial cell neighbors, and generate protrusive blebs that likely facilitate their ingression into a subepithelial tissue layer. How signaling pathways regulate the progression of delamination is important for understanding numerous developmental events. In this review, we focus on cellular and molecular mechanisms that drive cell ingression and draw comparisons between different morphogenetic contexts in various model organisms. We speculate that cell behaviors that facilitated tissue invagination among diploblasts subsequently drove individual cell ingression and epithelial-to-mesenchymal transition. Future insights that link signalling pathways to biophysical mechanisms will likely advance our comprehension of this phenomenon.
Collapse
Affiliation(s)
- Clarissa C Pasiliao
- Program in Developmental and Stem Cell Biology, Research Institute, The Hospital for Sick Children, Toronto, Ontario, M5G 0A4, Canada.,Department of Molecular Genetics, University of Toronto, M5S 1A8, Canada
| | - Sevan Hopyan
- Program in Developmental and Stem Cell Biology, Research Institute, The Hospital for Sick Children, Toronto, Ontario, M5G 0A4, Canada.,Department of Molecular Genetics, University of Toronto, M5S 1A8, Canada.,Division of Orthopaedic Surgery, Hospital for Sick Children and University of, Toronto, M5G 1X8, Canada
| |
Collapse
|
29
|
Reig G, Cerda M, Sepúlveda N, Flores D, Castañeda V, Tada M, Härtel S, Concha ML. Extra-embryonic tissue spreading directs early embryo morphogenesis in killifish. Nat Commun 2017; 8:15431. [PMID: 28580937 PMCID: PMC5465322 DOI: 10.1038/ncomms15431] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Accepted: 03/30/2017] [Indexed: 01/20/2023] Open
Abstract
The spreading of mesenchymal-like cell layers is critical for embryo morphogenesis and tissue repair, yet we know little of this process in vivo. Here we take advantage of unique developmental features of the non-conventional annual killifish embryo to study the principles underlying tissue spreading in a simple cellular environment, devoid of patterning signals and major morphogenetic cell movements. Using in vivo experimentation and physical modelling we reveal that the extra-embryonic epithelial enveloping cell layer, thought mainly to provide protection to the embryo, directs cell migration and the spreading of embryonic tissue during early development. This function relies on the ability of embryonic cells to couple their autonomous random motility to non-autonomous signals arising from the expansion of the extra-embryonic epithelium, mediated by cell membrane adhesion and tension. Thus, we present a mechanism of extra-embryonic control of embryo morphogenesis that couples the mechanical properties of adjacent tissues in the early killifish embryo.
Collapse
Affiliation(s)
- Germán Reig
- Anatomy and Developmental Biology Program, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, PO Box 70031, Santiago, Chile.,Biomedical Neuroscience Institute, Independencia 1027, Santiago, Chile
| | - Mauricio Cerda
- Anatomy and Developmental Biology Program, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, PO Box 70031, Santiago, Chile.,Biomedical Neuroscience Institute, Independencia 1027, Santiago, Chile
| | - Néstor Sepúlveda
- Department of Physics, Faculty of Physical and Mathematical Sciences, Universidad de Chile, PO Box 487-3, Santiago, Chile
| | - Daniela Flores
- Anatomy and Developmental Biology Program, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, PO Box 70031, Santiago, Chile.,Biomedical Neuroscience Institute, Independencia 1027, Santiago, Chile
| | - Victor Castañeda
- Anatomy and Developmental Biology Program, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, PO Box 70031, Santiago, Chile.,Biomedical Neuroscience Institute, Independencia 1027, Santiago, Chile
| | - Masazumi Tada
- Department of Cell and Developmental Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - Steffen Härtel
- Anatomy and Developmental Biology Program, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, PO Box 70031, Santiago, Chile.,Biomedical Neuroscience Institute, Independencia 1027, Santiago, Chile.,National Center for Health Information Systems CENS, Independencia 1027, Santiago, Chile
| | - Miguel L Concha
- Anatomy and Developmental Biology Program, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, PO Box 70031, Santiago, Chile.,Biomedical Neuroscience Institute, Independencia 1027, Santiago, Chile.,Center for Geroscience, Brain Health and Metabolism, Las Palmeras 3425, Ñuñoa, Santiago, Chile
| |
Collapse
|
30
|
Smutny M, Ákos Z, Grigolon S, Shamipour S, Ruprecht V, Čapek D, Behrndt M, Papusheva E, Tada M, Hof B, Vicsek T, Salbreux G, Heisenberg CP. Friction forces position the neural anlage. Nat Cell Biol 2017; 19:306-317. [PMID: 28346437 PMCID: PMC5635970 DOI: 10.1038/ncb3492] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Accepted: 02/17/2017] [Indexed: 12/23/2022]
Abstract
During embryonic development, mechanical forces are essential for cellular rearrangements driving tissue morphogenesis. Here, we show that in the early zebrafish embryo, friction forces are generated at the interface between anterior axial mesoderm (prechordal plate, ppl) progenitors migrating towards the animal pole and neurectoderm progenitors moving in the opposite direction towards the vegetal pole of the embryo. These friction forces lead to global rearrangement of cells within the neurectoderm and determine the position of the neural anlage. Using a combination of experiments and simulations, we show that this process depends on hydrodynamic coupling between neurectoderm and ppl as a result of E-cadherin-mediated adhesion between those tissues. Our data thus establish the emergence of friction forces at the interface between moving tissues as a critical force-generating process shaping the embryo.
Collapse
Affiliation(s)
- Michael Smutny
- Institute of Science and Technology Austria, Am Campus 1,
A-3400 Klosterneuburg, Austria
| | - Zsuzsa Ákos
- Department of Biological Physics, Eötvös
University, Pázmány Péter sétány 1A, Budapest
H-1117, Hungary
| | - Silvia Grigolon
- The Francis Crick Institute, 1 Midland Road, London NW1
1AT, UK
| | - Shayan Shamipour
- Institute of Science and Technology Austria, Am Campus 1,
A-3400 Klosterneuburg, Austria
| | - Verena Ruprecht
- Centre for Genomic Regulation (CRG), The Barcelona
Institute for Science and Technology, Dr. Aiguader 88, 08003 Barcelona, Spain
- Universitat Pompeu Fabra (UPF), 08003, Barcelona,
Spain
| | - Daniel Čapek
- Institute of Science and Technology Austria, Am Campus 1,
A-3400 Klosterneuburg, Austria
| | - Martin Behrndt
- Institute of Science and Technology Austria, Am Campus 1,
A-3400 Klosterneuburg, Austria
| | - Ekaterina Papusheva
- Institute of Science and Technology Austria, Am Campus 1,
A-3400 Klosterneuburg, Austria
| | - Masazumi Tada
- Department of Cell and Developmental Biology, University
College London, Gower Street, London, WC1E 6BT, UK
| | - Björn Hof
- Institute of Science and Technology Austria, Am Campus 1,
A-3400 Klosterneuburg, Austria
| | - Tamás Vicsek
- Department of Biological Physics, Eötvös
University, Pázmány Péter sétány 1A, Budapest
H-1117, Hungary
| | | | | |
Collapse
|
31
|
The guanine nucleotide exchange factor Net1 facilitates the specification of dorsal cell fates in zebrafish embryos by promoting maternal β-catenin activation. Cell Res 2016; 27:202-225. [PMID: 27910850 DOI: 10.1038/cr.2016.141] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2016] [Revised: 09/18/2016] [Accepted: 09/27/2016] [Indexed: 12/14/2022] Open
Abstract
Wnt/β-catenin signaling is essential for the initiation of dorsal-ventral patterning during vertebrate embryogenesis. Maternal β-catenin accumulates in dorsal marginal nuclei during cleavage stages, but its critical target genes essential for dorsalization are silent until mid-blastula transition (MBT). Here, we find that zebrafish net1, a guanine nucleotide exchange factor, is specifically expressed in dorsal marginal blastomeres after MBT, and acts as a zygotic factor to promote the specification of dorsal cell fates. Loss- and gain-of-function experiments show that the GEF activity of Net1 is required for the activation of Wnt/β-catenin signaling in zebrafish embryos and mammalian cells. Net1 dissociates and activates PAK1 dimers, and PAK1 kinase activation causes phosphorylation of S675 of β-catenin after MBT, which ultimately leads to the transcription of downstream target genes. In summary, our results reveal that Net1-regulated β-catenin activation plays a crucial role in the dorsal axis formation during zebrafish development.
Collapse
|
32
|
Myosin phosphatase Fine-tunes Zebrafish Motoneuron Position during Axonogenesis. PLoS Genet 2016; 12:e1006440. [PMID: 27855159 PMCID: PMC5147773 DOI: 10.1371/journal.pgen.1006440] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Accepted: 10/21/2016] [Indexed: 01/30/2023] Open
Abstract
During embryogenesis the spinal cord shifts position along the anterior-posterior axis relative to adjacent tissues. How motor neurons whose cell bodies are located in the spinal cord while their axons reside in adjacent tissues compensate for such tissue shift is not well understood. Using live cell imaging in zebrafish, we show that as motor axons exit from the spinal cord and extend through extracellular matrix produced by adjacent notochord cells, these cells shift several cell diameters caudally. Despite this pronounced shift, individual motoneuron cell bodies stay aligned with their extending axons. We find that this alignment requires myosin phosphatase activity within motoneurons, and that mutations in the myosin phosphatase subunit mypt1 increase myosin phosphorylation causing a displacement between motoneuron cell bodies and their axons. Thus, we demonstrate that spinal motoneurons fine-tune their position during axonogenesis and we identify the myosin II regulatory network as a key regulator.
Collapse
|
33
|
Abstract
Cellular motility is essential for many processes such as embryonic development, wound healing processes, tissue assembly and regeneration, immune cell trafficing and diseases such as cancer. The migration efficiency and the migratory potential depend on the type of migration mode. The previously established migration modes such as epithelial (non-migratory) and mesenchymal (migratory) as well as amoeboid (squeezing motility) relay mainly on phenomenological criteria such as cell morphology and molecular biological criteria such as gene expression. However, the physical view on the migration modes is still not well understood. As the process of malignant cancer progression such as metastasis depends on the migration of single cancer cells and their migration mode, this review focuses on the different migration strategies and discusses which mechanical prerequisites are necessary to perform a special migration mode through a 3-dimensional microenvironment. In particular, this review discusses how cells can distinguish and finally switch between the migration modes and what impact do the physical properties of cells and their microenvironment have on the transition between the novel migration modes such as blebbing and protrusive motility.
Collapse
Affiliation(s)
- Claudia Tanja Mierke
- a Faculty of Physics and Earth Science; Institute of Experimental Physics I; Biological Physics Division; University of Leipzig ; Leipzig , Germany
| |
Collapse
|
34
|
Wnt Signaling in Cell Motility and Invasion: Drawing Parallels between Development and Cancer. Cancers (Basel) 2016; 8:cancers8090080. [PMID: 27589803 PMCID: PMC5040982 DOI: 10.3390/cancers8090080] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Revised: 08/09/2016] [Accepted: 08/22/2016] [Indexed: 12/12/2022] Open
Abstract
The importance of canonical and non-canonical Wnt signal transduction cascades in embryonic development and tissue homeostasis is well recognized. The aberrant activation of these pathways in the adult leads to abnormal cellular behaviors, and tumor progression is frequently a consequence. Here we discuss recent findings and analogies between Wnt signaling in developmental processes and tumor progression, with a particular focus on cell motility and matrix invasion and highlight the roles of the ARF (ADP-Ribosylation Factor) and Rho-family small GTP-binding proteins. Wnt-regulated signal transduction from cell surface receptors, signaling endosomes and/or extracellular vesicles has the potential to profoundly influence cell movement, matrix degradation and paracrine signaling in both development and disease.
Collapse
|
35
|
Alexandrova AY. Plasticity of tumor cell migration: acquisition of new properties or return to the past? BIOCHEMISTRY (MOSCOW) 2015; 79:947-63. [PMID: 25385021 DOI: 10.1134/s0006297914090107] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
During tumor development cancer cells pass through several stages when cell morphology and migration abilities change remarkably. These stages are named epithelial-mesenchymal and mesenchymal-amoeboid transitions. The molecular mechanisms underlying cell motility are changing during these transitions. As result of transitions the cells acquire new characteristics and modes of motility. Cell migration becomes more independent from the environmental conditions, and thus cell dissemination becomes more aggressive, which leads to formation of distant metastases. In this review we discuss the characteristics of each of the transitions, cell morphology, and the specificity of cellular structures responsible for different modes of cell motility as well as molecular mechanisms regulating each transition.
Collapse
Affiliation(s)
- A Y Alexandrova
- Institute of Carcinogenesis, Blokhin Cancer Research Center, Russian Academy of Medical Sciences, Moscow, 115478, Russia.
| |
Collapse
|
36
|
Sugiyama T, Pramanik MK, Yumura S. Microtubule-Mediated Inositol Lipid Signaling Plays Critical Roles in Regulation of Blebbing. PLoS One 2015; 10:e0137032. [PMID: 26317626 PMCID: PMC4552846 DOI: 10.1371/journal.pone.0137032] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2015] [Accepted: 08/11/2015] [Indexed: 12/12/2022] Open
Abstract
Cells migrate by extending pseudopods such as lamellipodia and blebs. Although the signals leading to lamellipodia extension have been extensively investigated, those for bleb extension remain unclear. Here, we investigated signals for blebbing in Dictyostelium cells using a newly developed assay to induce blebbing. When cells were cut into two pieces with a microneedle, the anucleate fragments vigorously extended blebs. This assay enabled us to induce blebbing reproducibly, and analyses of knockout mutants and specific inhibitors identified candidate molecules that regulate blebbing. Blebs were also induced in anucleate fragments of leukocytes, indicating that this assay is generally applicable to animal cells. After cutting, microtubules in the anucleate fragments promptly depolymerized, followed by the extension of blebs. Furthermore, when intact cells were treated with a microtubule inhibitor, they frequently extended blebs. The depolymerization of microtubules induced the delocalization of inositol lipid phosphatidylinositol 3,4,5-trisphosphate from the cell membrane. PI3 kinase-null cells frequently extended blebs, whereas PTEN-null cells extended fewer blebs. From these observations, we propose a model in which microtubules play a critical role in bleb regulation via inositol lipid metabolism.
Collapse
Affiliation(s)
- Tatsuroh Sugiyama
- Department of Functional Molecular Biology, Graduate School of Medicine, Yamaguchi University, Yamaguchi, Japan
| | - Md. Kamruzzaman Pramanik
- Department of Functional Molecular Biology, Graduate School of Medicine, Yamaguchi University, Yamaguchi, Japan
- Microbiology & Industrial Irradiation Division, IFRB, AERE, Bangladesh Atomic Energy Commission, Dhaka, Bangladesh
| | - Shigehiko Yumura
- Department of Functional Molecular Biology, Graduate School of Medicine, Yamaguchi University, Yamaguchi, Japan
- * E-mail:
| |
Collapse
|
37
|
Ossipova O, Kim K, Sokol SY. Planar polarization of Vangl2 in the vertebrate neural plate is controlled by Wnt and Myosin II signaling. Biol Open 2015; 4:722-30. [PMID: 25910938 PMCID: PMC4467192 DOI: 10.1242/bio.201511676] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The vertebrate neural tube forms as a result of complex morphogenetic movements, which require the functions of several core planar cell polarity (PCP) proteins, including Vangl2 and Prickle. Despite the importance of these proteins for neurulation, their subcellular localization and the mode of action have remained largely unknown. Here we describe the anteroposterior planar cell polarity (AP-PCP) of the cells in the Xenopus neural plate. At the neural midline, the Vangl2 protein is enriched at anterior cell edges and that this localization is directed by Prickle, a Vangl2-interacting protein. Our further analysis is consistent with the model, in which Vangl2 AP-PCP is established in the neural plate as a consequence of Wnt-dependent phosphorylation. Additionally, we uncover feedback regulation of Vangl2 polarity by Myosin II, reiterating a role for mechanical forces in PCP. These observations indicate that both Wnt signaling and Myosin II activity regulate cell polarity and cell behaviors during vertebrate neurulation.
Collapse
Affiliation(s)
- Olga Ossipova
- Department of Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Kyeongmi Kim
- Department of Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Sergei Y Sokol
- Department of Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
38
|
Azevedo H, Fujita A, Bando SY, Iamashita P, Moreira-Filho CA. Transcriptional network analysis reveals that AT1 and AT2 angiotensin II receptors are both involved in the regulation of genes essential for glioma progression. PLoS One 2014; 9:e110934. [PMID: 25365520 PMCID: PMC4217762 DOI: 10.1371/journal.pone.0110934] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2014] [Accepted: 09/26/2014] [Indexed: 01/25/2023] Open
Abstract
Gliomas are aggressive primary brain tumors with high infiltrative potential. The expression of Angiotensin II (Ang II) receptors has been associated with poor prognosis in human astrocytomas, the most common type of glioma. In this study, we investigated the role of Angiotensin II in glioma malignancy through transcriptional profiling and network analysis of cultured C6 rat glioma cells exposed to Ang II and to inhibitors of its membrane receptor subtypes. C6 cells were treated with Ang II and specific antagonists of AT1 and AT2 receptors. Total RNA was isolated after three and six hours of Ang II treatment and analyzed by oligonucleotide microarray technology. Gene expression data was evaluated through transcriptional network modeling to identify how differentially expressed (DE) genes are connected to each other. Moreover, other genes co-expressing with the DE genes were considered in these analyses in order to support the identification of enriched functions and pathways. A hub-based network analysis showed that the most connected nodes in Ang II-related networks exert functions associated with cell proliferation, migration and invasion, key aspects for glioma progression. The subsequent functional enrichment analysis of these central genes highlighted their participation in signaling pathways that are frequently deregulated in gliomas such as ErbB, MAPK and p53. Noteworthy, either AT1 or AT2 inhibitions were able to down-regulate different sets of hub genes involved in protumoral functions, suggesting that both Ang II receptors could be therapeutic targets for intervention in glioma. Taken together, our results point out multiple actions of Ang II in glioma pathogenesis and reveal the participation of both Ang II receptors in the regulation of genes relevant for glioma progression. This study is the first one to provide systems-level molecular data for better understanding the protumoral effects of Ang II in the proliferative and infiltrative behavior of gliomas.
Collapse
Affiliation(s)
- Hátylas Azevedo
- Department of Pediatrics, Faculdade de Medicina da Universidade de São Paulo (FMUSP), São Paulo, SP, Brazil
| | - André Fujita
- Department of Computer Science, Instituto de Matemática e Estatística, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Silvia Yumi Bando
- Department of Pediatrics, Faculdade de Medicina da Universidade de São Paulo (FMUSP), São Paulo, SP, Brazil
| | - Priscila Iamashita
- Department of Pediatrics, Faculdade de Medicina da Universidade de São Paulo (FMUSP), São Paulo, SP, Brazil
| | - Carlos Alberto Moreira-Filho
- Department of Pediatrics, Faculdade de Medicina da Universidade de São Paulo (FMUSP), São Paulo, SP, Brazil
- * E-mail:
| |
Collapse
|
39
|
Carr BW, Basepayne TL, Chen L, Jayashankar V, Weiser DC. Characterization of the zebrafish homolog of zipper interacting protein kinase. Int J Mol Sci 2014; 15:11597-613. [PMID: 24983477 PMCID: PMC4139802 DOI: 10.3390/ijms150711597] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2014] [Revised: 06/12/2014] [Accepted: 06/23/2014] [Indexed: 11/16/2022] Open
Abstract
Zipper-interacting protein kinase (ZIPK) is a conserved vertebrate-specific regulator of actomyosin contractility in smooth muscle and non-muscle cells. Murine ZIPK has undergone an unusual divergence in sequence and regulation compared to other ZIPK orthologs. In humans, subcellular localization is controlled by phosphorylation of threonines 299 and 300. In contrast, ZIPK subcellular localization in mouse and rat is controlled by interaction with PAR-4. We carried out a comparative biochemical characterization of the regulation of the zebrafish ortholog of ZIPK. Like the human orthologs zebrafish ZIPK undergoes nucleocytoplasmic-shuttling and is abundant in the cytoplasm, unlike the primarily nuclear rat ZIPK. Rat ZIPK, but not human or zebrafish ZIPK, interacts with zebrafish PAR-4. Mutation of the conserved residues required for activation of the mammalian orthologs abrogated activity of the zebrafish ZIPK. In contrast to the human ortholog, mutation of threonine 299 and 300 in the zebrafish ZIPK has no effect on the activity or subcellular localization. Thus, we found that zebrafish ZIPK functions in a manner most similar to the human ZIPK and quite distinct from murine orthologs, yet the regulation of subcellular localization is not conserved.
Collapse
Affiliation(s)
- Brandon W Carr
- Department of Biological Sciences, University of the Pacific, Stockton, CA 98211, USA.
| | - Tamara L Basepayne
- Department of Biological Sciences, University of the Pacific, Stockton, CA 98211, USA.
| | - Lawrence Chen
- Department of Biological Sciences, University of the Pacific, Stockton, CA 98211, USA.
| | - Vaishali Jayashankar
- Department of Biological Sciences, University of the Pacific, Stockton, CA 98211, USA.
| | - Douglas C Weiser
- Department of Biological Sciences, University of the Pacific, Stockton, CA 98211, USA.
| |
Collapse
|
40
|
Abstract
Cadherins are transmembrane proteins that mediate cell-cell adhesion in animals. By regulating contact formation and stability, cadherins play a crucial role in tissue morphogenesis and homeostasis. Here, we review the three major functions of cadherins in cell-cell contact formation and stability. Two of those functions lead to a decrease in interfacial tension at the forming cell-cell contact, thereby promoting contact expansion--first, by providing adhesion tension that lowers interfacial tension at the cell-cell contact, and second, by signaling to the actomyosin cytoskeleton in order to reduce cortex tension and thus interfacial tension at the contact. The third function of cadherins in cell-cell contact formation is to stabilize the contact by resisting mechanical forces that pull on the contact.
Collapse
|
41
|
Jayashankar V, Nguyen MJ, Carr BW, Zheng DC, Rosales JB, Rosales JB, Weiser DC. Protein phosphatase 1 β paralogs encode the zebrafish myosin phosphatase catalytic subunit. PLoS One 2013; 8:e75766. [PMID: 24040418 PMCID: PMC3770619 DOI: 10.1371/journal.pone.0075766] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2013] [Accepted: 08/19/2013] [Indexed: 12/21/2022] Open
Abstract
Background The myosin phosphatase is a highly conserved regulator of actomyosin contractility. Zebrafish has emerged as an ideal model system to study the invivo role of myosin phosphatase in controlling cell contractility, cell movement and epithelial biology. Most work in zebrafish has focused on the regulatory subunit of the myosin phosphatase called Mypt1. In this work, we examined the critical role of Protein Phosphatase 1, PP1, the catalytic subunit of the myosin phosphatase. Methodology/Principal Findings We observed that in zebrafish two paralogous genes encoding PP1β, called ppp1cba and ppp1cbb, are both broadly expressed during early development. Furthermore, we found that both gene products interact with Mypt1 and assemble an active myosin phosphatase complex. In addition, expression of this complex results in dephosphorylation of the myosin regulatory light chain and large scale rearrangements of the actin cytoskeleton. Morpholino knock-down of ppp1cba and ppp1cbb results in severe defects in morphogenetic cell movements during gastrulation through loss of myosin phosphatase function. Conclusions/Significance Our work demonstrates that zebrafish have two genes encoding PP1β, both of which can interact with Mypt1 and assemble an active myosin phosphatase. In addition, both genes are required for convergence and extension during gastrulation and correct dosage of the protein products is required.
Collapse
Affiliation(s)
- Vaishali Jayashankar
- Department of Biological Sciences, University of the Pacific, Stockton, California, United States of America
| | - Michael J. Nguyen
- Department of Biological Sciences, University of the Pacific, Stockton, California, United States of America
| | - Brandon W. Carr
- Department of Biological Sciences, University of the Pacific, Stockton, California, United States of America
| | - Dale C. Zheng
- Department of Biological Sciences, University of the Pacific, Stockton, California, United States of America
| | - Joseph B. Rosales
- Department of Biological Sciences, University of the Pacific, Stockton, California, United States of America
| | - Joshua B. Rosales
- Department of Biological Sciences, University of the Pacific, Stockton, California, United States of America
| | - Douglas C. Weiser
- Department of Biological Sciences, University of the Pacific, Stockton, California, United States of America
- * E-mail:
| |
Collapse
|
42
|
The role and regulation of blebs in cell migration. Curr Opin Cell Biol 2013; 25:582-90. [PMID: 23786923 PMCID: PMC3989058 DOI: 10.1016/j.ceb.2013.05.005] [Citation(s) in RCA: 253] [Impact Index Per Article: 21.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2013] [Accepted: 05/25/2013] [Indexed: 12/22/2022]
Abstract
Blebs are cellular protrusions that have been shown to be instrumental for cell migration in development and disease. Bleb expansion is driven by hydrostatic pressure generated in the cytoplasm by the contractile actomyosin cortex. The mechanisms of bleb formation thus fundamentally differ from the actin polymerization-based mechanisms responsible for lamellipodia expansion. In this review, we summarize recent findings relevant for the mechanics of bleb formation and the underlying molecular pathways. We then review the processes involved in determining the type of protrusion formed by migrating cells, in particular in vivo, in the context of embryonic development. Finally, we discuss how cells utilize blebs for their forward movement in the presence or absence of strong substrate attachment.
Collapse
|
43
|
Tada M, Heisenberg CP. Convergent extension: using collective cell migration and cell intercalation to shape embryos. Development 2012; 139:3897-904. [PMID: 23048180 DOI: 10.1242/dev.073007] [Citation(s) in RCA: 180] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Body axis elongation represents a common and fundamental morphogenetic process in development. A key mechanism triggering body axis elongation without additional growth is convergent extension (CE), whereby a tissue undergoes simultaneous narrowing and extension. Both collective cell migration and cell intercalation are thought to drive CE and are used to different degrees in various species as they elongate their body axis. Here, we provide an overview of CE as a general strategy for body axis elongation and discuss conserved and divergent mechanisms underlying CE among different species.
Collapse
Affiliation(s)
- Masazumi Tada
- Department of Cell and Developmental Biology, University College London, Gower Street, London, WC1E 6BT, UK.
| | | |
Collapse
|
44
|
Regional cell shape changes control form and function of Kupffer's vesicle in the zebrafish embryo. Dev Biol 2012; 370:52-62. [PMID: 22841644 DOI: 10.1016/j.ydbio.2012.07.019] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2012] [Revised: 06/29/2012] [Accepted: 07/10/2012] [Indexed: 11/23/2022]
Abstract
Cilia-generated fluid flow in an 'organ of asymmetry' is critical for establishing the left-right body axis in several vertebrate embryos. However, the cell biology underlying how motile cilia produce coordinated flow and asymmetric signals is not well defined. In the zebrafish organ of asymmetry-called Kupffer's vesicle (KV)-ciliated cells are asymmetrically positioned along the anterior-posterior axis such that more cilia are placed in the anterior region. We previously demonstrated that Rho kinase 2b (Rock2b) is required for anteroposterior asymmetry and fluid flow in KV, but it remained unclear how the distribution of ciliated cells becomes asymmetric during KV development. Here, we identify a morphogenetic process we refer to as 'KV remodeling' that transforms initial symmetry in KV architecture into anteroposterior asymmetry. Live imaging of KV cells revealed region-specific cell shape changes that mediate tight packing of ciliated cells into the anterior pole. Mathematical modeling indicated that different interfacial tensions in anterior and posterior KV cells are involved in KV remodeling. Interfering with non-muscle myosin II (referred to as Myosin II) activity, which modulates cellular interfacial tensions and is regulated by Rock proteins, disrupted KV cell shape changes and the anteroposterior distribution of KV cilia. Similar defects were observed in Rock2b depleted embryos. Furthermore, inhibiting Myosin II at specific stages of KV development perturbed asymmetric flow and left-right asymmetry. These results indicate that regional cell shape changes control the development of anteroposterior asymmetry in KV, which is necessary to generate coordinated asymmetric fluid flow and left-right patterning of the embryo.
Collapse
|
45
|
Abstract
Gastrulation is a fundamental phase of animal embryogenesis during which germ layers are specified, rearranged, and shaped into a body plan with organ rudiments. Gastrulation involves four evolutionarily conserved morphogenetic movements, each of which results in a specific morphologic transformation. During emboly, mesodermal and endodermal cells become internalized beneath the ectoderm. Epibolic movements spread and thin germ layers. Convergence movements narrow germ layers dorsoventrally, while concurrent extension movements elongate them anteroposteriorly. Each gastrulation movement can be achieved by single or multiple motile cell behaviors, including cell shape changes, directed migration, planar and radial intercalations, and cell divisions. Recent studies delineate cyclical and ratchet-like behaviors of the actomyosin cytoskeleton as a common mechanism underlying various gastrulation cell behaviors. Gastrulation movements are guided by differential cell adhesion, chemotaxis, chemokinesis, and planar polarity. Coordination of gastrulation movements with embryonic polarity involves regulation by anteroposterior and dorsoventral patterning systems of planar polarity signaling, expression of chemokines, and cell adhesion molecules.
Collapse
Affiliation(s)
- Lila Solnica-Krezel
- Department of Developmental Biology, Washington University School of Medicine in St. Louis, St. Louis, Missouri 63110, USA.
| | | |
Collapse
|
46
|
Blebbing as a physical force in cancer EMT - parallels with mitosis. Semin Cancer Biol 2012; 22:369-73. [PMID: 22659342 DOI: 10.1016/j.semcancer.2012.05.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2012] [Revised: 04/19/2012] [Accepted: 05/24/2012] [Indexed: 12/15/2022]
Abstract
Epithelial to mesenchymal transition (EMT) during metastasis is initially a two-step process beginning with delamination of cells from the solid tumor followed by acquisition of a migratory phenotype. Several reports indicate that plasma membrane blebbing, associated with cell division, coincides with cell delamination during developmental EMT. This raises a speculative question if blebbing drives EMT in cancer cells in a similar way. Here, we review available data on factors and processes that may support such a connection.
Collapse
|
47
|
Schepis A, Nelson WJ. Adherens junction function and regulation during zebrafish gastrulation. Cell Adh Migr 2012; 6:173-8. [PMID: 22568981 PMCID: PMC3427231 DOI: 10.4161/cam.20583] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
The adherens junction (AJ) comprises multi-protein complexes required for cell-cell adhesion in embryonic development and adult tissue homeostasis. Mutations in key proteins and mis-regulation of AJ adhesive properties can lead to pathologies such as cancer. In recent years, the zebrafish has become an excellent model organism to integrate cell biology in the context of a multicellular organization. The combination of classical genetic approaches with new tools for live imaging and biophysical approaches has revealed new aspects of AJ biology, particularly during zebrafish gastrulation. These studies have resulted in progress in understanding the relationship between cell-cell adhesion, cell migration and plasma membrane blebbing.
Collapse
|
48
|
Du S, Draper BW, Mione M, Moens CB, Bruce AEE. Differential regulation of epiboly initiation and progression by zebrafish Eomesodermin A. Dev Biol 2012; 362:11-23. [PMID: 22142964 PMCID: PMC3259739 DOI: 10.1016/j.ydbio.2011.10.036] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2011] [Revised: 09/24/2011] [Accepted: 10/19/2011] [Indexed: 01/07/2023]
Abstract
The T-box transcription factor Eomesodermin (Eomes) has been implicated in patterning and morphogenesis in frog, fish and mouse. In zebrafish, one of the two Eomes homologs, Eomesa, has been implicated in dorsal-ventral patterning, epiboly and endoderm specification in experiments employing over-expression, dominant-negative constructs and antisense morpholino oligonucleotides. Here we report for the first time the identification and characterization of an Eomesa mutant generated by TILLING. We find that Eomesa has a strictly maternal role in the initiation of epiboly, which involves doming of the yolk cell up into the overlying blastoderm. By contrast, epiboly progression is normal, demonstrating for the first time that epiboly initiation is genetically separable from progression. The yolk cell microtubules, which are required for epiboly, are defective in maternal-zygotic eomesa mutant embryos. In addition, the deep cells of the blastoderm are more tightly packed and exhibit more bleb-like protrusions than cells in control embryos. We postulate that the doming delay may be the consequence both of overly stabilized yolk cell microtubules and defects in the adhesive properties or motility of deep cells. We also show that Eomesa is required for normal expression of the endoderm markers sox32, bon and og9x; however it is not essential for endoderm formation.
Collapse
Affiliation(s)
- Susan Du
- Department of Cell and Systems Biology University of Toronto 25 Harbord Street Toronto, ON M5S 3G5 Canada
| | - Bruce W. Draper
- Molecular and Cellular Biology University of California, Davis One Shields Avenue Davis, CA 95616 USA
| | - Marina Mione
- IFOM, Istituto FIRC di Oncologia Molecolare Via Adamello 16 Milan, I-20139 Italy
| | - Cecilia B. Moens
- Howard Hughes Medical Institute Division of Basic Science Fred Hutchinson Cancer Research Center P.O. Box 19024 1100 Fairview Avenue North Seattle, WA 98109-1024 USA
| | - Ashley E. E. Bruce
- Department of Cell and Systems Biology University of Toronto 25 Harbord Street Toronto, ON M5S 3G5 Canada
| |
Collapse
|
49
|
Weiser DC, Kimelman D. Analysis of cell shape and polarity during zebrafish gastrulation. Methods Mol Biol 2012; 839:53-68. [PMID: 22218892 DOI: 10.1007/978-1-61779-510-7_5] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
Gastrulation is a complex set of cellular rearrangements that establish the overall shape of the body plan during development. In addition to being an essential and fascinating aspect of development, the cells of the gastrulating zebrafish embryo also provide an ideal in vivo system to study the interplay of cell polarity and movement in a native 3D environment. During gastrulation, zebrafish mesodermal cells undergo a series of conversions from initial non-polarized amoeboid cell movements to more mesenchymal and finally highly polarized and intercalative cell behaviors. Many of the cellular behavior changes of these cells are under the control of the RhoA pathway, which in turn is regulated by many signals, including non-canonical Wnts. The goal of this chapter is to provide researchers with the necessary protocols to examine changes in cell polarity and movement in the developing zebrafish embryo.
Collapse
Affiliation(s)
- Douglas C Weiser
- Department of Biological Sciences, University of the Pacific, Stockton, CA, USA
| | | |
Collapse
|
50
|
Abstract
Planar cell polarity is a fundamental concept to understanding the coordination of cell movements in the plane of a tissue. Since the planar cell polarity pathway was discovered in mesenchymal tissues involving cell interaction during vertebrate gastrulation, there is an emerging evidence that a variety of mesenchymal and epithelial cells utilize this genetic pathway to mediate the coordination of cells in directed movements. In this review, we focus on how the planar cell polarity pathway is mediated by migrating cells to communicate with one another in different developmental processes.
Collapse
|