1
|
Ravichandran N, Iyer M, Uvarajan D, Kirola L, Kumra SM, Babu HWS, HariKrishnaReddy D, Vellingiri B, Narayanasamy A. New insights on the regulators and inhibitors of RhoA-ROCK signalling in Parkinson's disease. Metab Brain Dis 2025; 40:90. [PMID: 39775342 DOI: 10.1007/s11011-024-01500-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Accepted: 12/09/2024] [Indexed: 01/11/2025]
Abstract
A multifaceted and widely prevalent neurodegenerative disease, Parkinson's disease (PD) is typified by the loss of dopaminergic neurons in the midbrain. The discovery of novel treatment(s) that can reverse or halt the course of the disease progression along with identifying the most reliable biomarker(s) in PD remains the crucial concern. RhoA in its active state has been demonstrated to interact with three distinct domains located in the central coiled-coil region of ROCK. RhoA appears to activate effectors most frequently by breaking the intramolecular autoinhibitory connections, which releases functional domains from the effector protein. Additionally, RhoA is highly expressed in the nervous system and it acts as a central molecule for its several downstream effector proteins in multiple signalling pathways both in neurons and glial cells. Mitochondrial dysfunction, vesicle transport malfunction and aggregation of α-Synuclein, a presynaptic neuronal protein genetically and neuropathologically associated with PD. While the RhoA-ROCK signalling pathway appears to have a significant role in PD symptoms, suggesting it could be a promising target for therapeutic interventions. Thus, this review article addresses the potential involvement of the RhoA-ROCK signalling system in the pathophysiology of neurodegenerative illnesses, with an emphasis on its biology and function. We also provide an overview of the state of research on RhoA regulation and its downstream biological activities, focusing on the role of RhoA signalling in neurodegenerative illnesses and the potential benefits of RhoA inhibition as a treatment for neurodegeneration.
Collapse
Affiliation(s)
- Nandita Ravichandran
- Disease Proteomics Laboratory, Department of Zoology, Bharathiar University, Coimbatore, 641046, Tamil Nadu, India
| | - Mahalaxmi Iyer
- Department of Microbiology, Central University of Punjab, Bathinda, 151401, Punjab, India
| | - Deenathayalan Uvarajan
- Department of Biochemistry, PSG College of Arts & Science, Coimbatore, Tamil Nadu, India
| | - Laxmi Kirola
- Department of Biotechnology, School of Health Sciences & Technology (SoHST), UPES Dehradun, Dehradun, India
| | - Sindduja Muthu Kumra
- Human Cytogenetics and Stem Cell Laboratory, Department of Zoology, Central University of Punjab, Bathinda, 151401, Punjab, India
| | - Harysh Winster Suresh Babu
- Human Cytogenetics and Stem Cell Laboratory, Department of Zoology, Central University of Punjab, Bathinda, 151401, Punjab, India
| | - Dibbanti HariKrishnaReddy
- Advanced Pharmacology and Neuroscience Laboratory, Department of Pharmacology, School of Health Sciences, Central University of Punjab, Bathinda, 151401, Punjab, India
| | - Balachandar Vellingiri
- Human Cytogenetics and Stem Cell Laboratory, Department of Zoology, Central University of Punjab, Bathinda, 151401, Punjab, India.
| | - Arul Narayanasamy
- Disease Proteomics Laboratory, Department of Zoology, Bharathiar University, Coimbatore, 641046, Tamil Nadu, India.
| |
Collapse
|
2
|
Durmusalioglu EA, Dogan YC, Tan TT, Cogulu D, Isik E, Cogulu O, Atik T. From Clinical Observation to Genetic Confirmation: Somatic Mosaic Mutations in RHOA on Ectodermal Dysplasia With Multi-System Involvement. Am J Med Genet A 2024:e63934. [PMID: 39564940 DOI: 10.1002/ajmg.a.63934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 09/25/2024] [Accepted: 10/25/2024] [Indexed: 11/21/2024]
Abstract
Ectodermal dysplasia with facial dysmorphism and acral, ocular, and brain anomalies (EDFAOB) is a rare neuroectodermal syndrome caused by somatic mosaic mutations in the RHOA gene. It presents with linear skin hypopigmentation, facial and limb asymmetry, dental and acral anomalies, and leukoencephalopathy, generally preserving intellectual and neurological functions. We report two cases of EDFAOB. Both cases initially presented with notable facial-body asymmetry, thin hair, dental issues, digital anomalies, and Blaschko's lines-aligned hypopigmentation. A 6-year-old girl exhibited esotropia, visual center atrophy, and bilateral white matter hyperintensities on MRI. A 10-year-old girl had unilateral hyperintense lesions in the left cerebral hemisphere on MRI. Both had normal neuromotor development without intellectual impairment. RHOA gene sequencing from hypopigmented skin biopsies revealed the c.139G > A (p.Glu47Lys) mutation, with allele fractions of 20% and 10%, respectively, absent in blood leukocytes and parental DNA. These cases highlight the clinical and genetic features of EDFAOB and underscore the importance of thorough clinical evaluation to guide precise genetic testing. The identification of mutations exclusively in affected tissues supports a postzygotic mosaic distribution, refining the diagnostic approach for this syndrome.
Collapse
Affiliation(s)
| | | | | | | | - Esra Isik
- Pediatric Genetics, Ege University, Izmir, Turkey
| | - Ozgur Cogulu
- Pediatric Genetics, Ege University, Izmir, Turkey
| | - Tahir Atik
- Pediatric Genetics, Ege University, Izmir, Turkey
| |
Collapse
|
3
|
Yu W, Kastriti ME, Ishan M, Choudhary SK, Rashid MM, Kramer N, Do HGT, Wang Z, Xu T, Schwabe RF, Ye K, Adameyko I, Liu HX. The duct of von Ebner's glands is a source of Sox10 + taste bud progenitors and susceptible to pathogen infections. Front Cell Dev Biol 2024; 12:1460669. [PMID: 39247625 PMCID: PMC11377339 DOI: 10.3389/fcell.2024.1460669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Accepted: 08/01/2024] [Indexed: 09/10/2024] Open
Abstract
Introduction We have recently demonstrated that Sox10-expressing (Sox10 +) cells give rise to mainly type-III neuronal taste bud cells that are responsible for sour and salt taste. The two tissue compartments containing Sox10 + cells in the surrounding of taste buds include the connective tissue core of taste papillae and von Ebner's glands (vEGs) that are connected to the trench of circumvallate and foliate papillae. Methods In this study, we performed single cell RNA-sequencing of the epithelium of Sox10-Cre/tdT mouse circumvallate/vEG complex and used inducible Cre mouse models to map the cell lineages of vEGs and/or connective tissue (including stromal and Schwann cells). Results Transcriptomic analysis indicated that Sox10 expression was enriched in the cell clusters of vEG ducts that contained abundant proliferating cells, while Sox10-Cre/tdT expression was enriched in type-III taste bud cells and vEG ductal cells. In vivo lineage mapping showed that the traced cells were distributed in circumvallate taste buds concurrently with those in the vEGs, but not in the connective tissue. Moreover, multiple genes encoding pathogen receptors were enriched in the vEG ducts hosting Sox10 + cells. Discussion Our data supports that it is the vEGs, not connective tissue core, that serve as the niche of Sox10 + taste bud progenitors. If this is also true in humans, our data indicates that vEG duct is a source of Sox10 + taste bud progenitors and susceptible to pathogen infections.
Collapse
Affiliation(s)
- Wenxin Yu
- Department of Animal and Dairy Science, Regenerative Bioscience Center, College of Agricultural and Environmental Sciences, University of Georgia, Athens, GA, United States
| | | | - Mohamed Ishan
- Department of Animal and Dairy Science, Regenerative Bioscience Center, College of Agricultural and Environmental Sciences, University of Georgia, Athens, GA, United States
| | | | - Md Mamunur Rashid
- Department of Animal and Dairy Science, Regenerative Bioscience Center, College of Agricultural and Environmental Sciences, University of Georgia, Athens, GA, United States
| | - Naomi Kramer
- Department of Animal and Dairy Science, Regenerative Bioscience Center, College of Agricultural and Environmental Sciences, University of Georgia, Athens, GA, United States
| | - Hy Gia Truong Do
- Department of Genetics, University of Georgia, Athens, GA, United States
| | - Zhonghou Wang
- Department of Animal and Dairy Science, Regenerative Bioscience Center, College of Agricultural and Environmental Sciences, University of Georgia, Athens, GA, United States
| | - Ting Xu
- Department of Animal and Dairy Science, Regenerative Bioscience Center, College of Agricultural and Environmental Sciences, University of Georgia, Athens, GA, United States
| | - Robert F Schwabe
- Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, NY, United States
| | - Kaixiong Ye
- Institute of Bioinformatics, University of Georgia, Athens, GA, United States
- Department of Genetics, University of Georgia, Athens, GA, United States
| | - Igor Adameyko
- Department of Neuroimmunology, Medical University of Vienna, Vienna, Austria
- Department of Physiology and Pharmacology, Karolinska Institutet, Solna, Sweden
| | - Hong-Xiang Liu
- Department of Animal and Dairy Science, Regenerative Bioscience Center, College of Agricultural and Environmental Sciences, University of Georgia, Athens, GA, United States
| |
Collapse
|
4
|
Takatani H, Fujita N, Imai F, Yoshida Y. Forelimb motor recovery by modulating extrinsic and intrinsic signaling as well as neuronal activity after the cervical spinal cord injury. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.22.600167. [PMID: 38979293 PMCID: PMC11230274 DOI: 10.1101/2024.06.22.600167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Singular strategies for promoting axon regeneration and motor recovery after spinal cord injury (SCI) have been attempted with limited success. Here, we propose the combinatorial approach of deleting extrinsic and intrinsic factors paired with neural stimulation, will enhance adaptive axonal growth and motor recovery after SCI. We previously showed the deletion of RhoA and Pten in corticospinal neurons inhibits axon dieback and promotes axon sprouting after lumbar SCI. Here, we examined the effects of RhoA;Pten deletion coupled with neural stimulation after cervical SCI. This combinatorial approach promoted more boutons on injured corticospinal neurons in the spinal cord compared to sole RhoA;Pten deletion. Although RhoA;Pten deletion does not promote motor recovery in the forelimb after SCI, stimulating corticospinal neurons in those mice results in partial motor recovery. These results demonstrate that a combinatorial approach that pairs genetic modifications with neuronal stimulation can promote axon sprouting and motor recovery following SCI.
Collapse
Affiliation(s)
- Hirohide Takatani
- Burke Neurological Institute, White Plains, New York, United States
- Brain and Mind Research Institute, Weill Cornell Medicine, New York, United States
- Laboratory of Veterinary Surgery, Graduate School of Agriculture and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Naoki Fujita
- Laboratory of Veterinary Surgery, Graduate School of Agriculture and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Fumiyasu Imai
- Burke Neurological Institute, White Plains, New York, United States
- Brain and Mind Research Institute, Weill Cornell Medicine, New York, United States
| | - Yutaka Yoshida
- Burke Neurological Institute, White Plains, New York, United States
- Brain and Mind Research Institute, Weill Cornell Medicine, New York, United States
- Neural Circuit Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
- Lead contact
| |
Collapse
|
5
|
Yu W, Kastriti ME, Ishan M, Choudhary SK, Kramer N, Rashid MM, Truong Do HG, Wang Z, Xu T, Schwabe RF, Ye K, Adameyko I, Liu HX. The main duct of von Ebner's glands is a source of Sox10 + taste bud progenitors and susceptible to pathogen infections. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.14.594215. [PMID: 38798668 PMCID: PMC11118543 DOI: 10.1101/2024.05.14.594215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
We have recently demonstrated that Sox10 -expressing ( Sox10 + ) cells give rise to mainly type-III neuronal taste bud cells that are responsible for sour and salt taste. The two tissue compartments containing Sox10 + cells in the surrounding of taste buds include the connective tissue core of taste papillae and von Ebner's glands (vEGs) that are connected to the trench of circumvallate and foliate papillae. In this study, we used inducible Cre mouse models to map the cell lineages of connective tissue (including stromal and Schwann cells) and vEGs and performed single cell RNA-sequencing of the epithelium of Sox10-Cre/tdT mouse circumvallate/vEG complex. In vivo lineage mapping showed that the distribution of traced cells in circumvallate taste buds was closely linked with that in the vEGs, but not in the connective tissue. Sox10 , but not the known stem cells marker Lgr5 , expression was enriched in the cell clusters of main ducts of vEGs that contained abundant proliferating cells, while Sox10-Cre/tdT expression was enriched in type-III taste bud cells and excretory ductal cells. Moreover, multiple genes encoding pathogen receptors are enriched in the vEG main ducts. Our data indicate that the main duct of vEGs is a source of Sox10 + taste bud progenitors and susceptible to pathogen infections.
Collapse
|
6
|
Soh JEC, Shimizu A, Sato A, Ogita H. Novel cardiovascular protective effects of RhoA signaling and its therapeutic implications. Biochem Pharmacol 2023; 218:115899. [PMID: 37907138 DOI: 10.1016/j.bcp.2023.115899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 10/24/2023] [Accepted: 10/26/2023] [Indexed: 11/02/2023]
Abstract
Ras homolog gene family member A (RhoA) belongs to the Rho GTPase superfamily, which was first studied in cancers as one of the essential regulators controlling cellular function. RhoA has long attracted attention as a key molecule involved in cell signaling and gene transcription, through which it affects cellular processes. A series of studies have demonstrated that RhoA plays crucial roles under both physiological states and pathological conditions in cardiovascular diseases. RhoA has been identified as an important regulator in cardiac remodeling by regulating actin stress fiber dynamics and cytoskeleton formation. However, its underlying mechanisms remain poorly understood, preventing definitive conclusions being drawn about its protective role in the cardiovascular system. In this review, we outline the characteristics of RhoA and its related signaling molecules, and present an overview of RhoA classical function and the corresponding cellular responses of RhoA under physiological and pathological conditions. Overall, we provide an update on the novel signaling under RhoA in the cardiovascular system and its potential clinical and therapeutic targets in cardiovascular medicine.
Collapse
Affiliation(s)
- Joanne Ern Chi Soh
- Division of Molecular Medical Biochemistry, Department of Biochemistry and Molecular Biology, Shiga University of Medical Science, Otsu, Japan
| | - Akio Shimizu
- Division of Molecular Medical Biochemistry, Department of Biochemistry and Molecular Biology, Shiga University of Medical Science, Otsu, Japan
| | - Akira Sato
- Division of Molecular Medical Biochemistry, Department of Biochemistry and Molecular Biology, Shiga University of Medical Science, Otsu, Japan
| | - Hisakazu Ogita
- Division of Molecular Medical Biochemistry, Department of Biochemistry and Molecular Biology, Shiga University of Medical Science, Otsu, Japan.
| |
Collapse
|
7
|
Cossard A, Stam K, Smets A, Jossin Y. MKL/SRF and Bcl6 mutual transcriptional repression safeguards the fate and positioning of neocortical progenitor cells mediated by RhoA. SCIENCE ADVANCES 2023; 9:eadd0676. [PMID: 37967194 PMCID: PMC10651131 DOI: 10.1126/sciadv.add0676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 10/16/2023] [Indexed: 11/17/2023]
Abstract
During embryogenesis, multiple intricate and intertwined cellular signaling pathways coordinate cell behavior. Their slightest alterations can have dramatic consequences for the cells and the organs they form. The transcriptional repressor Bcl6 was recently found as important for brain development. However, its regulation and integration with other signals is unknown. Using in vivo functional approaches combined with molecular mechanistic analysis, we identified a reciprocal regulatory loop between B cell lymphoma 6 (Bcl6) and the RhoA-regulated transcriptional complex megakaryoblastic leukemia/serum response factor (MKL/SRF). We show that Bcl6 physically interacts with MKL/SRF, resulting in a down-regulation of the transcriptional activity of both Bcl6 and MKL/SRF. This molecular cross-talk is essential for the control of proliferation, neurogenesis, and spatial positioning of neural progenitors. Overall, our data highlight a regulatory mechanism that controls neuronal production and neocortical development and reveal an MKL/SRF and Bcl6 interaction that may have broader implications in other physiological functions and in diseases.
Collapse
Affiliation(s)
- Alexia Cossard
- Laboratory of Mammalian Development and Cell Biology, Institute of Neuroscience, Université Catholique de Louvain, Brussels 1200, Belgium
| | | | | | | |
Collapse
|
8
|
Yamamoto-Fukuda T, Akiyama N, Tatsumi N, Okabe M, Kojima H. Keratinocyte Growth Factor Stimulates Growth of p75 + Neural Crest Lineage Cells During Middle Ear Cholesteatoma Formation in Mice. THE AMERICAN JOURNAL OF PATHOLOGY 2022; 192:1573-1591. [PMID: 36210210 DOI: 10.1016/j.ajpath.2022.07.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Revised: 06/20/2022] [Accepted: 07/21/2022] [Indexed: 11/06/2022]
Abstract
During development, cranial neural crest (NC) cells display a striking transition from collective to single-cell migration and undergo a mesenchymal-to-epithelial transformation to form a part of the middle ear epithelial cells (MEECs). While MEECs derived from NC are known to control homeostasis of the epithelium and repair from otitis media, paracrine action of keratinocyte growth factor (KGF) promotes the growth of MEECs and induces middle ear cholesteatoma (cholesteatoma). The animal model of cholesteatoma was previously established by transfecting a human KGF-expression vector. Herein, KGF-inducing cholesteatoma was studied in Wnt1-Cre/Floxed-enhanced green fluorescent protein (EGFP) mice that conditionally express EGFP in the NC lineages. The cytokeratin 14-positive NC lineage expanded into the middle ear and formed cholesteatoma. Moreover, the green fluorescent protein-positive NC lineages comprising the cholesteatoma tissue expressed p75, an NC marker, with high proliferative activity. Similarly, a large number of p75-positive cells were observed in human cholesteatoma tissues. Injections of the immunotoxin murine p75-saporin induced depletion of the p75-positive NC lineages, resulting in the reduction of cholesteatoma in vivo. The p75 knockout in the MEECs had low proliferative activity with or without KGF protein in vitro. Controlling p75 signaling may reduce the proliferation of NC lineages and may represent a new therapeutic target for cholesteatoma.
Collapse
Affiliation(s)
- Tomomi Yamamoto-Fukuda
- Department of Otorhinolaryngology, Jikei University School of Medicine, Tokyo, Japan; Department of Anatomy, Jikei University School of Medicine, Tokyo, Japan.
| | - Naotaro Akiyama
- Department of Anatomy, Jikei University School of Medicine, Tokyo, Japan; Department of Otorhinolaryngology, Toho University School of Medicine, Tokyo, Japan
| | - Norifumi Tatsumi
- Department of Anatomy, Jikei University School of Medicine, Tokyo, Japan
| | - Masataka Okabe
- Department of Anatomy, Jikei University School of Medicine, Tokyo, Japan
| | - Hiromi Kojima
- Department of Otorhinolaryngology, Jikei University School of Medicine, Tokyo, Japan
| |
Collapse
|
9
|
Rac-deficient cerebellar granule neurons die before they migrate to the internal granule layer. Sci Rep 2022; 12:14848. [PMID: 36050459 PMCID: PMC9436960 DOI: 10.1038/s41598-022-19252-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 08/26/2022] [Indexed: 11/23/2022] Open
Abstract
Granule neurons are the most common cell type in the cerebellum. They are generated in the external granule layer and migrate inwardly, forming the internal granule layer. Small Rho GTPases play various roles during development of the nervous system and may be involved in generation, differentiation and migration of granule neurons. We deleted Rac1, a member of small Rho GTPases, by GFAP-Cre driver in cerebellar granule neurons and Bergmann glial cells. Rac1flox/flox; Cre mice showed impaired migration and slight reduction in the number of granule neurons in the internal granule layer. Deletion of both Rac1 and Rac3 resulted in almost complete absence of granule neurons. Rac-deficient granule neurons differentiated into p27 and NeuN-expressing post mitotic neurons, but died before migration to the internal granule layer. Loss of Rac3 has little effect on granule neuron development. Rac1flox/flox; Rac3+/−; Cre mice showed intermediate phenotype between Rac1flox/flox; Cre and Rac1flox/flox; Rac3−/−; Cre mice in both survival and migration of granule neurons. Rac3 itself seems to be unimportant in the development of the cerebellum, but has some roles in Rac1-deleted granule neurons. Conversely, overall morphology of Rac1+/flox; Rac3−/−; Cre cerebella was normal. One allele of Rac1 is therefore thought to be sufficient to promote development of cerebellar granule neurons.
Collapse
|
10
|
Avansini SH, Puppo F, Adams JW, Vieira AS, Coan AC, Rogerio F, Torres FR, Araújo PAOR, Martin M, Montenegro MA, Yasuda CL, Tedeschi H, Ghizoni E, França AFEC, Alvim MKM, Athié MC, Rocha CS, Almeida VS, Dias EV, Delay L, Molina E, Yaksh TL, Cendes F, Lopes Cendes I, Muotri AR. Junctional instability in neuroepithelium and network hyperexcitability in a focal cortical dysplasia human model. Brain 2022; 145:1962-1977. [PMID: 34957478 PMCID: PMC9336577 DOI: 10.1093/brain/awab479] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 10/15/2021] [Accepted: 11/19/2021] [Indexed: 11/14/2022] Open
Abstract
Focal cortical dysplasia is a highly epileptogenic cortical malformation with few treatment options. Here, we generated human cortical organoids from patients with focal cortical dysplasia type II. Using this human model, we mimicked some focal cortical dysplasia hallmarks, such as impaired cell proliferation, the presence of dysmorphic neurons and balloon cells, and neuronal network hyperexcitability. Furthermore, we observed alterations in the adherens junctions zonula occludens-1 and partitioning defective 3, reduced polarization of the actin cytoskeleton, and fewer synaptic puncta. Focal cortical dysplasia cortical organoids showed downregulation of the small GTPase RHOA, a finding that was confirmed in brain tissue resected from these patients. Functionally, both spontaneous and optogenetically-evoked electrical activity revealed hyperexcitability and enhanced network connectivity in focal cortical dysplasia organoids. Taken together, our findings suggest a ventricular zone instability in tissue cohesion of neuroepithelial cells, leading to a maturational arrest of progenitors or newborn neurons, which may predispose to cellular and functional immaturity and compromise the formation of neural networks in focal cortical dysplasia.
Collapse
Affiliation(s)
- Simoni H Avansini
- Department of Pediatrics/Rady Children’s Hospital-San Diego, Department of Cellular & Molecular Medicine, School of Medicine, University of California San Diego, La Jolla, CA 92037, USA
- Department of Translational Medicine, School of Medical Sciences, University of Campinas, Campinas, Sao Paulo 13083-887, Brazil
- Brazilian Institute of Neuroscience and Neurotechnology (BRAINN), University of Campinas, Campinas, Sao Paulo 13083-888, Brazil
| | - Francesca Puppo
- Department of Pediatrics/Rady Children’s Hospital-San Diego, Department of Cellular & Molecular Medicine, School of Medicine, University of California San Diego, La Jolla, CA 92037, USA
| | - Jason W Adams
- Department of Pediatrics/Rady Children’s Hospital-San Diego, Department of Cellular & Molecular Medicine, School of Medicine, University of California San Diego, La Jolla, CA 92037, USA
| | - Andre S Vieira
- Brazilian Institute of Neuroscience and Neurotechnology (BRAINN), University of Campinas, Campinas, Sao Paulo 13083-888, Brazil
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas, Campinas Sao Paulo 13083-887, Brazil
| | - Ana C Coan
- Brazilian Institute of Neuroscience and Neurotechnology (BRAINN), University of Campinas, Campinas, Sao Paulo 13083-888, Brazil
- Department of Neurology, School of Medical Sciences, University of Campinas, Campinas Sao Paulo 13083-887, Brazil
| | - Fabio Rogerio
- Brazilian Institute of Neuroscience and Neurotechnology (BRAINN), University of Campinas, Campinas, Sao Paulo 13083-888, Brazil
- Department of Pathology, School of Medical Sciences, University of Campinas, Campinas, Sao Paulo 13083-887, Brazil
| | - Fabio R Torres
- Department of Translational Medicine, School of Medical Sciences, University of Campinas, Campinas, Sao Paulo 13083-887, Brazil
- Brazilian Institute of Neuroscience and Neurotechnology (BRAINN), University of Campinas, Campinas, Sao Paulo 13083-888, Brazil
| | - Patricia A O R Araújo
- Department of Translational Medicine, School of Medical Sciences, University of Campinas, Campinas, Sao Paulo 13083-887, Brazil
- Brazilian Institute of Neuroscience and Neurotechnology (BRAINN), University of Campinas, Campinas, Sao Paulo 13083-888, Brazil
| | - Mariana Martin
- Department of Translational Medicine, School of Medical Sciences, University of Campinas, Campinas, Sao Paulo 13083-887, Brazil
- Brazilian Institute of Neuroscience and Neurotechnology (BRAINN), University of Campinas, Campinas, Sao Paulo 13083-888, Brazil
| | - Maria A Montenegro
- Brazilian Institute of Neuroscience and Neurotechnology (BRAINN), University of Campinas, Campinas, Sao Paulo 13083-888, Brazil
- Department of Neurology, School of Medical Sciences, University of Campinas, Campinas Sao Paulo 13083-887, Brazil
| | - Clarissa L Yasuda
- Brazilian Institute of Neuroscience and Neurotechnology (BRAINN), University of Campinas, Campinas, Sao Paulo 13083-888, Brazil
- Department of Neurology, School of Medical Sciences, University of Campinas, Campinas Sao Paulo 13083-887, Brazil
| | - Helder Tedeschi
- Brazilian Institute of Neuroscience and Neurotechnology (BRAINN), University of Campinas, Campinas, Sao Paulo 13083-888, Brazil
- Department of Neurology, School of Medical Sciences, University of Campinas, Campinas Sao Paulo 13083-887, Brazil
| | - Enrico Ghizoni
- Brazilian Institute of Neuroscience and Neurotechnology (BRAINN), University of Campinas, Campinas, Sao Paulo 13083-888, Brazil
- Department of Neurology, School of Medical Sciences, University of Campinas, Campinas Sao Paulo 13083-887, Brazil
| | - Andréa F E C França
- Department of Clinical Medicine, School of Medical Sciences, University of Campinas, Campinas, Sao Paulo 13083-887, Brazil
| | - Marina K M Alvim
- Brazilian Institute of Neuroscience and Neurotechnology (BRAINN), University of Campinas, Campinas, Sao Paulo 13083-888, Brazil
- Department of Neurology, School of Medical Sciences, University of Campinas, Campinas Sao Paulo 13083-887, Brazil
| | - Maria C Athié
- Department of Translational Medicine, School of Medical Sciences, University of Campinas, Campinas, Sao Paulo 13083-887, Brazil
- Brazilian Institute of Neuroscience and Neurotechnology (BRAINN), University of Campinas, Campinas, Sao Paulo 13083-888, Brazil
| | - Cristiane S Rocha
- Department of Translational Medicine, School of Medical Sciences, University of Campinas, Campinas, Sao Paulo 13083-887, Brazil
- Brazilian Institute of Neuroscience and Neurotechnology (BRAINN), University of Campinas, Campinas, Sao Paulo 13083-888, Brazil
| | - Vanessa S Almeida
- Department of Translational Medicine, School of Medical Sciences, University of Campinas, Campinas, Sao Paulo 13083-887, Brazil
- Brazilian Institute of Neuroscience and Neurotechnology (BRAINN), University of Campinas, Campinas, Sao Paulo 13083-888, Brazil
| | - Elayne V Dias
- Department of Anesthesiology/Medical Center Hillcrest, School of Medicine, University of California San Diego, Hillcrest, CA 92103, USA
| | - Lauriane Delay
- Department of Anesthesiology/Medical Center Hillcrest, School of Medicine, University of California San Diego, Hillcrest, CA 92103, USA
| | - Elsa Molina
- Stem Cell Genomics and Microscopy Core, Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, CA 92037, USA
| | - Tony L Yaksh
- Department of Anesthesiology/Medical Center Hillcrest, School of Medicine, University of California San Diego, Hillcrest, CA 92103, USA
| | - Fernando Cendes
- Brazilian Institute of Neuroscience and Neurotechnology (BRAINN), University of Campinas, Campinas, Sao Paulo 13083-888, Brazil
- Department of Neurology, School of Medical Sciences, University of Campinas, Campinas Sao Paulo 13083-887, Brazil
| | - Iscia Lopes Cendes
- Department of Translational Medicine, School of Medical Sciences, University of Campinas, Campinas, Sao Paulo 13083-887, Brazil
- Brazilian Institute of Neuroscience and Neurotechnology (BRAINN), University of Campinas, Campinas, Sao Paulo 13083-888, Brazil
| | - Alysson R Muotri
- Department of Pediatrics/Rady Children’s Hospital-San Diego, Department of Cellular & Molecular Medicine, School of Medicine, University of California San Diego, La Jolla, CA 92037, USA
- Kavli Institute for Brain and Mind, Archealization Center (ArchC), Center for Academic Research and Training in Anthropogeny (CARTA), University of California San Diego, La Jolla, CA 92093, USA
| |
Collapse
|
11
|
Camargo Ortega G, Götz M. Centrosome heterogeneity in stem cells regulates cell diversity. Trends Cell Biol 2022; 32:707-719. [PMID: 35750615 DOI: 10.1016/j.tcb.2022.03.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 03/14/2022] [Accepted: 03/21/2022] [Indexed: 11/27/2022]
Abstract
Stem cells are at the source of creating cellular diversity. Multiple mechanisms, including basic cell biological processes, regulate their fate. The centrosome is at the core of many stem cell functions and recent work highlights the association of distinct proteins at the centrosome in stem cell differentiation. As showcased by a novel centrosome protein regulating neural stem cell differentiation, it is timely to review the heterogeneity of the centrosome at protein and RNA levels and how this impacts their function in stem and progenitor cells. Together with evidence for heterogeneity of other organelles so far considered as similar between cells, we call for exploring the cell type-specific composition of organelles as a way to expand protein function in development with relevance to regenerative medicine.
Collapse
Affiliation(s)
- Germán Camargo Ortega
- Department of Biosystems Science and Engineering, ETH, Zurich, 4058 Basel, Switzerland.
| | - Magdalena Götz
- Institute of Stem Cell Research, Helmholtz Center Munich, 82152 Planegg-Martinsried, Germany; Physiological Genomics, Biomedical Center, Ludwig-Maximilians University, 82152 Planegg-Martinsried, Germany; 4 SYNERGY, Excellence Cluster of Systems Neurology, Biomedical Center, Ludwig-Maximilians-University, 82152 Planegg-Martinsried, Germany.
| |
Collapse
|
12
|
Vaid S, Huttner WB. Progenitor-Based Cell Biological Aspects of Neocortex Development and Evolution. Front Cell Dev Biol 2022; 10:892922. [PMID: 35602606 PMCID: PMC9119302 DOI: 10.3389/fcell.2022.892922] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 04/14/2022] [Indexed: 11/13/2022] Open
Abstract
During development, the decision of stem and progenitor cells to switch from proliferation to differentiation is of critical importance for the overall size of an organ. Too early a switch will deplete the stem/progenitor cell pool, and too late a switch will not generate the required differentiated cell types. With a focus on the developing neocortex, a six-layered structure constituting the major part of the cerebral cortex in mammals, we discuss here the cell biological features that are crucial to ensure the appropriate proliferation vs. differentiation decision in the neural progenitor cells. In the last two decades, the neural progenitor cells giving rise to the diverse types of neurons that function in the neocortex have been intensely investigated for their role in cortical expansion and gyrification. In this review, we will first describe these different progenitor types and their diversity. We will then review the various cell biological features associated with the cell fate decisions of these progenitor cells, with emphasis on the role of the radial processes emanating from these progenitor cells. We will also discuss the species-specific differences in these cell biological features that have allowed for the evolutionary expansion of the neocortex in humans. Finally, we will discuss the emerging role of cell cycle parameters in neocortical expansion.
Collapse
Affiliation(s)
- Samir Vaid
- Department of Basic Neurosciences, University of Geneva, Geneva, Switzerland
- *Correspondence: Samir Vaid, ; Wieland B. Huttner,
| | - Wieland B. Huttner
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
- *Correspondence: Samir Vaid, ; Wieland B. Huttner,
| |
Collapse
|
13
|
Wang Z, Ren D, Zheng P. The role of Rho/ROCK in epileptic seizure-related neuronal damage. Metab Brain Dis 2022; 37:881-887. [PMID: 35119588 PMCID: PMC9042975 DOI: 10.1007/s11011-022-00909-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Accepted: 01/06/2022] [Indexed: 01/09/2023]
Abstract
Epilepsy is one of the most severe neurological disorders characterized by spontaneous recurrent seizures. Although more than two-thirds of patients can be cured with anti-epileptic drugs (AEDs), the rest one-third of epilepsy patients are resistant to AEDs. A series of studies have demonstrated Rho/Rho-associated kinase (ROCK) pathway might be involved in the pathogenesis of epilepsy in the recent twenty years. Several related pathway inhibitors of Rho/ROCK have been used in the treatment of epilepsy. We searched PubMed from Jan 1, 2000 to Dec 31, 2020, using the terms "epilepsy AND Rho AND ROCK" and "seizure AND Rho AND ROCK". We selected articles that characterized Rho/ROCK in animal models of epilepsy and patients. We then chose the most relevant research studies including in-vitro, in-vivo and clinical trials. The expression of Rho/ROCK could be a potential non-invasive biomarker to apply in treatment for patients with epilepsy. RhoA and ROCK show significant upregulation in the acute and chronic stage of epilepsy. ROCK inhibitors can reduce the epilepsy, epileptic seizure-related neuronal death and comorbidities. These findings demonstrate the novel development for diagnosis and treatment for patients with epilepsy. Rho/ROCK signaling pathway inhibitors may show more promising effects in epilepsy and related neurological diseases.
Collapse
Affiliation(s)
- Zhihan Wang
- Department of Neurosurgery, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, 201399, China
| | - Dabin Ren
- Department of Neurusurgery & Key Laboratory, Shanghai Pudong New area People's Hospital, Shanghai, 201299, China
| | - Ping Zheng
- Department of Neurusurgery & Key Laboratory, Shanghai Pudong New area People's Hospital, Shanghai, 201299, China.
| |
Collapse
|
14
|
Sokpor G, Brand-Saberi B, Nguyen HP, Tuoc T. Regulation of Cell Delamination During Cortical Neurodevelopment and Implication for Brain Disorders. Front Neurosci 2022; 16:824802. [PMID: 35281509 PMCID: PMC8904418 DOI: 10.3389/fnins.2022.824802] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 01/31/2022] [Indexed: 11/13/2022] Open
Abstract
Cortical development is dependent on key processes that can influence apical progenitor cell division and progeny. Pivotal among such critical cellular processes is the intricate mechanism of cell delamination. This indispensable cell detachment process mainly entails the loss of apical anchorage, and subsequent migration of the mitotic derivatives of the highly polarized apical cortical progenitors. Such apical progenitor derivatives are responsible for the majority of cortical neurogenesis. Many factors, including transcriptional and epigenetic/chromatin regulators, are known to tightly control cell attachment and delamination tendency in the cortical neurepithelium. Activity of these molecular regulators principally coordinate morphogenetic cues to engender remodeling or disassembly of tethering cellular components and external cell adhesion molecules leading to exit of differentiating cells in the ventricular zone. Improper cell delamination is known to frequently impair progenitor cell fate commitment and neuronal migration, which can cause aberrant cortical cell number and organization known to be detrimental to the structure and function of the cerebral cortex. Indeed, some neurodevelopmental abnormalities, including Heterotopia, Schizophrenia, Hydrocephalus, Microcephaly, and Chudley-McCullough syndrome have been associated with cell attachment dysregulation in the developing mammalian cortex. This review sheds light on the concept of cell delamination, mechanistic (transcriptional and epigenetic regulation) nuances involved, and its importance for corticogenesis. Various neurodevelopmental disorders with defective (too much or too little) cell delamination as a notable etiological underpinning are also discussed.
Collapse
Affiliation(s)
- Godwin Sokpor
- Department of Human Genetics, Ruhr University Bochum, Bochum, Germany
- Department of Anatomy and Molecular Embryology, Ruhr University Bochum, Bochum, Germany
- *Correspondence: Godwin Sokpor,
| | - Beate Brand-Saberi
- Department of Anatomy and Molecular Embryology, Ruhr University Bochum, Bochum, Germany
| | - Huu Phuc Nguyen
- Department of Human Genetics, Ruhr University Bochum, Bochum, Germany
| | - Tran Tuoc
- Department of Human Genetics, Ruhr University Bochum, Bochum, Germany
- Tran Tuoc,
| |
Collapse
|
15
|
Vav Proteins in Development of the Brain: A Potential Relationship to the Pathogenesis of Congenital Zika Syndrome? Viruses 2022; 14:v14020386. [PMID: 35215978 PMCID: PMC8874935 DOI: 10.3390/v14020386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 02/07/2022] [Accepted: 02/10/2022] [Indexed: 12/07/2022] Open
Abstract
Zika virus (ZIKV) infection during pregnancy can result in a significant impact on the brain and eye of the developing fetus, termed congenital zika syndrome (CZS). At a morphological level, the main serious presentations of CZS are microcephaly and retinal scarring. At a cellular level, many cell types of the brain may be involved, but primarily neuronal progenitor cells (NPC) and developing neurons. Vav proteins have guanine exchange activity in converting GDP to GTP on proteins such as Rac1, Cdc42 and RhoA to stimulate intracellular signaling pathways. These signaling pathways are known to play important roles in maintaining the polarity and self-renewal of NPC pools by coordinating the formation of adherens junctions with cytoskeletal rearrangements. In developing neurons, these same pathways are adopted to control the formation and growth of neurites and mediate axonal guidance and targeting in the brain and retina. This review describes the role of Vavs in these processes and highlights the points of potential ZIKV interaction, such as (i) the binding and entry of ZIKV in cells via TAM receptors, which may activate Vav/Rac/RhoA signaling; (ii) the functional convergence of ZIKV NS2A with Vav in modulating adherens junctions; (iii) ZIKV NS4A/4B protein effects on PI3K/AKT in a regulatory loop via PPI3 to influence Vav/Rac1 signaling in neurite outgrowth; and (iv) the induction of SOCS1 and USP9X following ZIKV infection to regulate Vav protein degradation or activation, respectively, and impact Vav/Rac/RhoA signaling in NPC and neurons. Experiments to define these interactions will further our understanding of the molecular basis of CZS and potentially other developmental disorders stemming from in utero infections. Additionally, Vav/Rac/RhoA signaling pathways may present tractable targets for therapeutic intervention or molecular rationale for disease severity in CZS.
Collapse
|
16
|
Duman JG, Blanco FA, Cronkite CA, Ru Q, Erikson KC, Mulherkar S, Saifullah AB, Firozi K, Tolias KF. Rac-maninoff and Rho-vel: The symphony of Rho-GTPase signaling at excitatory synapses. Small GTPases 2022; 13:14-47. [PMID: 33955328 PMCID: PMC9707551 DOI: 10.1080/21541248.2021.1885264] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 01/22/2021] [Accepted: 01/28/2021] [Indexed: 01/15/2023] Open
Abstract
Synaptic connections between neurons are essential for every facet of human cognition and are thus regulated with extreme precision. Rho-family GTPases, molecular switches that cycle between an active GTP-bound state and an inactive GDP-bound state, comprise a critical feature of synaptic regulation. Rho-GTPases are exquisitely controlled by an extensive suite of activators (GEFs) and inhibitors (GAPs and GDIs) and interact with many different signalling pathways to fulfill their roles in orchestrating the development, maintenance, and plasticity of excitatory synapses of the central nervous system. Among the mechanisms that control Rho-GTPase activity and signalling are cell surface receptors, GEF/GAP complexes that tightly regulate single Rho-GTPase dynamics, GEF/GAP and GEF/GEF functional complexes that coordinate multiple Rho-family GTPase activities, effector positive feedback loops, and mutual antagonism of opposing Rho-GTPase pathways. These complex regulatory mechanisms are employed by the cells of the nervous system in almost every step of development, and prominently figure into the processes of synaptic plasticity that underlie learning and memory. Finally, misregulation of Rho-GTPases plays critical roles in responses to neuronal injury, such as traumatic brain injury and neuropathic pain, and in neurodevelopmental and neurodegenerative disorders, including intellectual disability, autism spectrum disorder, schizophrenia, and Alzheimer's Disease. Thus, decoding the mechanisms of Rho-GTPase regulation and function at excitatory synapses has great potential for combatting many of the biggest current challenges in mental health.
Collapse
Affiliation(s)
- Joseph G. Duman
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - Francisco A. Blanco
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
- Integrative Molecular and Biomedical Science Graduate Program, Baylor College of Medicine, Houston, TX, USA
| | - Christopher A. Cronkite
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX, USA
- Medical Scientist Training Program, Baylor College of Medicine, Houston, TX, USA
| | - Qin Ru
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - Kelly C. Erikson
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - Shalaka Mulherkar
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - Ali Bin Saifullah
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - Karen Firozi
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - Kimberley F. Tolias
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
17
|
Zhang Z, Liu M, Zheng Y. Role of Rho GTPases in stem cell regulation. Biochem Soc Trans 2021; 49:2941-2955. [PMID: 34854916 PMCID: PMC9008577 DOI: 10.1042/bst20211071] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 11/17/2021] [Accepted: 11/18/2021] [Indexed: 02/05/2023]
Abstract
The future of regenerative medicine relies on our understanding of stem cells which are essential for tissue/organ generation and regeneration to maintain and/or restore tissue homeostasis. Rho family GTPases are known regulators of a wide variety of cellular processes related to cytoskeletal dynamics, polarity and gene transcription. In the last decade, major new advances have been made in understanding the regulatory role and mechanism of Rho GTPases in self-renewal, differentiation, migration, and lineage specification in tissue-specific signaling mechanisms in various stem cell types to regulate embryonic development, adult tissue homeostasis, and tissue regeneration upon stress or damage. Importantly, implication of Rho GTPases and their upstream regulators or downstream effectors in the transformation, migration, invasion and tumorigenesis of diverse cancer stem cells highlights the potential of Rho GTPase targeting in cancer therapy. In this review, we discuss recent evidence of Rho GTPase signaling in the regulation of embryonic stem cells, multiple somatic stem cells, and cancer stem cells. We propose promising areas where Rho GTPase pathways may serve as useful targets for stem cell manipulation and related future therapies.
Collapse
Affiliation(s)
- Zheng Zhang
- Division of Experimental Hematology and Cancer Biology, Children’s Hospital Medical Center, University of Cincinnati, 3333 Burnet Avenue, Cincinnati, OH 45229, U.S.A
| | - Ming Liu
- Department of Abdominal Oncology, West China Hospital, Sichuan University, Chengdu, China
| | - Yi Zheng
- Division of Experimental Hematology and Cancer Biology, Children’s Hospital Medical Center, University of Cincinnati, 3333 Burnet Avenue, Cincinnati, OH 45229, U.S.A
| |
Collapse
|
18
|
Kim SE, Robles-Lopez K, Cao X, Liu K, Chothani PJ, Bhavani N, Rahman L, Mukhopadhyay S, Wlodarczyk BJ, Finnell RH. Wnt1 Lineage Specific Deletion of Gpr161 Results in Embryonic Midbrain Malformation and Failure of Craniofacial Skeletal Development. Front Genet 2021; 12:761418. [PMID: 34887903 PMCID: PMC8650154 DOI: 10.3389/fgene.2021.761418] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 11/04/2021] [Indexed: 11/13/2022] Open
Abstract
Sonic hedgehog (Shh) signaling regulates multiple morphogenetic processes during embryonic neurogenesis and craniofacial skeletal development. Gpr161 is a known negative regulator of Shh signaling. Nullizygous Gpr161 mice are embryonic lethal, presenting with structural defects involving the neural tube and the craniofacies. However, the lineage specific role of Gpr161 in later embryonic development has not been thoroughly investigated. We studied the Wnt1-Cre lineage specific role of Gpr161 during mouse embryonic development. We observed three major gross morphological phenotypes in Gpr161 cKO (Gpr161 f/f; Wnt1-Cre) fetuses; protrusive tectum defect, encephalocele, and craniofacial skeletal defect. The overall midbrain tissues were expanded and cell proliferation in ventricular zones of midbrain was increased in Gpr161 cKO fetuses, suggesting that protrusive tectal defects in Gpr161 cKO are secondary to the increased proliferation of midbrain neural progenitor cells. Shh signaling activity as well as upstream Wnt signaling activity were increased in midbrain tissues of Gpr161 cKO fetuses. RNA sequencing further suggested that genes in the Shh, Wnt, Fgf and Notch signaling pathways were differentially regulated in the midbrain of Gpr161 cKO fetuses. Finally, we determined that cranial neural crest derived craniofacial bone formation was significantly inhibited in Gpr161 cKO fetuses, which partly explains the development of encephalocele. Our results suggest that Gpr161 plays a distinct role in midbrain development and in the formation of the craniofacial skeleton during mouse embryogenesis.
Collapse
Affiliation(s)
- Sung-Eun Kim
- Department of Pediatrics, Dell Pediatric Research Institute, University of Texas at Austin Dell Medical School, Austin, TX, United States
| | - Karla Robles-Lopez
- Department of Pediatrics, Dell Pediatric Research Institute, University of Texas at Austin Dell Medical School, Austin, TX, United States.,Center for Precision Environmental Health, Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, United States
| | - Xuanye Cao
- Center for Precision Environmental Health, Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, United States
| | - Kristyn Liu
- Department of Pediatrics, Dell Pediatric Research Institute, University of Texas at Austin Dell Medical School, Austin, TX, United States
| | - Pooja J Chothani
- Department of Pediatrics, Dell Pediatric Research Institute, University of Texas at Austin Dell Medical School, Austin, TX, United States
| | - Nikitha Bhavani
- Department of Pediatrics, Dell Pediatric Research Institute, University of Texas at Austin Dell Medical School, Austin, TX, United States
| | - Lauren Rahman
- Department of Pediatrics, Dell Pediatric Research Institute, University of Texas at Austin Dell Medical School, Austin, TX, United States
| | - Saikat Mukhopadhyay
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Bogdan J Wlodarczyk
- Center for Precision Environmental Health, Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, United States
| | - Richard H Finnell
- Department of Pediatrics, Dell Pediatric Research Institute, University of Texas at Austin Dell Medical School, Austin, TX, United States.,Center for Precision Environmental Health, Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, United States.,Departments of Molecular and Human Genetics and Medicine, Baylor College of Medicine, Houston, TX, United States
| |
Collapse
|
19
|
Abstract
Neural crest stem/progenitor cells arise early during vertebrate embryogenesis at the border of the forming central nervous system. They subsequently migrate throughout the body, eventually differentiating into diverse cell types ranging from neurons and glia of the peripheral nervous system to bones of the face, portions of the heart, and pigmentation of the skin. Along the body axis, the neural crest is heterogeneous, with different subpopulations arising in the head, neck, trunk, and tail regions, each characterized by distinct migratory patterns and developmental potential. Modern genomic approaches like single-cell RNA- and ATAC-sequencing (seq) have greatly enhanced our understanding of cell lineage trajectories and gene regulatory circuitry underlying the developmental progression of neural crest cells. Here, we discuss how genomic approaches have provided new insights into old questions in neural crest biology by elucidating transcriptional and posttranscriptional mechanisms that govern neural crest formation and the establishment of axial level identity. Expected final online publication date for the Annual Review of Genetics, Volume 55 is November 2021. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Shashank Gandhi
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California 91125, USA; ,
| | - Marianne E Bronner
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California 91125, USA; ,
| |
Collapse
|
20
|
Zhou P, Qi Y, Fang X, Yang M, Zheng S, Liao C, Qin F, Liu L, Li H, Li Y, Ravindran E, Sun C, Wei X, Wang W, Fang L, Han D, Peng C, Chen W, Li N, Kaindl AM, Hu H. Arhgef2 regulates neural differentiation in the cerebral cortex through mRNA m 6A-methylation of Npdc1 and Cend1. iScience 2021; 24:102645. [PMID: 34142067 PMCID: PMC8185223 DOI: 10.1016/j.isci.2021.102645] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 04/16/2021] [Accepted: 05/20/2021] [Indexed: 12/23/2022] Open
Abstract
N6-methyladenosine (m6A) is emerging as a vital factor regulating neural differentiation. Here, we report that deficiency of Arhgef2, a novel cause of a neurodevelopmental disorder we identified recently, impairs neurogenesis, neurite outgrowth, and synaptic formation by regulating m6A methylation. Arhgef2 knockout decreases expression of Mettl14 and total m6A level significantly in the cerebral cortex. m6A sequencing reveals that loss of Arhgef2 reduces m6A methylation of 1,622 mRNAs, including Npdc1 and Cend1, which are both strongly associated with cell cycle exit and terminal neural differentiation. Arhgef2 deficiency decreases m6A methylations of the Npdc1 and Cend1 mRNAs via down-regulation of Mettl14, and thereby inhibits the translation of Npdc1 and nuclear export of Cend1 mRNAs. Overexpression of Mettl14, Npdc1, and Cend1 rescue the abnormal phenotypes in Arhgef2 knockout mice, respectively. Our study provides a critical insight into a mechanism by which defective Arhgef2 mediates m6A-tagged target mRNAs to impair neural differentiation. Arhgef2 mediates total m6A level via Mettl14 Arhgef2 affects m6A methylations of the Npdc1 and Cend1 mRNAs Decreased m6A methylations inhibits translation of Npdc1 and nuclear export of Cend1 Reduced protein expression of Npdc1 and Cend1 hinders neural differentiation
Collapse
Affiliation(s)
- Pei Zhou
- Laboratory of Medical Systems Biology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 510623 Guangzhou, China
| | - Yifei Qi
- Division of Uterine Vascular Biology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 510623 Guangzhou, China
| | - Xiang Fang
- Laboratory of Medical Systems Biology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 510623 Guangzhou, China
| | - Miaomiao Yang
- Laboratory of Medical Systems Biology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 510623 Guangzhou, China
| | - Shuxin Zheng
- Laboratory of Medical Systems Biology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 510623 Guangzhou, China
| | - Caihua Liao
- Laboratory of Medical Systems Biology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 510623 Guangzhou, China
| | - Fengying Qin
- Laboratory of Medical Systems Biology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 510623 Guangzhou, China
| | - Lili Liu
- Laboratory of Medical Systems Biology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 510623 Guangzhou, China
| | - Hong Li
- Laboratory of Medical Systems Biology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 510623 Guangzhou, China
| | - Yan Li
- Laboratory of Medical Systems Biology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 510623 Guangzhou, China
| | - Ethiraj Ravindran
- Charité - Universitätsmedizin Berlin, Institute of Cell Biology and Neurobiology, Berlin, Germany.,Charité - Universitätsmedizin Berlin, Department of Pediatric Neurology, Berlin, Germany.,Charité - Universitätsmedizin Berlin, Center for Chronically Sick Children, Berlin, Germany
| | - Chuanbo Sun
- Laboratory of Medical Systems Biology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 510623 Guangzhou, China
| | - Xinshu Wei
- Laboratory of Medical Systems Biology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 510623 Guangzhou, China.,School of Medicine, South China University of Technology, 510006 Guangzhou, China
| | - Wen Wang
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, School of Life Sciences and Academy for Advanced Interdisciplinary Studies, Southern University of Science and Technology, Shenzhen 518005, China
| | - Liang Fang
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, School of Life Sciences and Academy for Advanced Interdisciplinary Studies, Southern University of Science and Technology, Shenzhen 518005, China
| | - Dingding Han
- Laboratory of Medical Systems Biology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 510623 Guangzhou, China
| | - Changgeng Peng
- The First Rehabilitation Hospital of Shanghai, Tongji University School of Medicine, 200029 Shanghai, China
| | - Wei Chen
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, School of Life Sciences and Academy for Advanced Interdisciplinary Studies, Southern University of Science and Technology, Shenzhen 518005, China
| | - Na Li
- Laboratory of Medical Systems Biology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 510623 Guangzhou, China
| | - Angela M Kaindl
- Charité - Universitätsmedizin Berlin, Institute of Cell Biology and Neurobiology, Berlin, Germany.,Charité - Universitätsmedizin Berlin, Department of Pediatric Neurology, Berlin, Germany.,Charité - Universitätsmedizin Berlin, Center for Chronically Sick Children, Berlin, Germany
| | - Hao Hu
- Laboratory of Medical Systems Biology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 510623 Guangzhou, China.,School of Medicine, South China University of Technology, 510006 Guangzhou, China.,Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 510623 Guangzhou, China.,Third Affiliated Hospital of Zhengzhou University, 450052 Zhengzhou, China
| |
Collapse
|
21
|
Marfull-Oromí P, Fleitas C, Zammou B, Rocandio D, Ballester-Lurbe B, Terrado J, Perez-Roger I, Espinet C, Egea J. Genetic ablation of the Rho GTPase Rnd3 triggers developmental defects in internal capsule and the globus pallidus formation. J Neurochem 2021; 158:197-216. [PMID: 33576044 DOI: 10.1111/jnc.15322] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 01/20/2021] [Accepted: 02/08/2021] [Indexed: 12/24/2022]
Abstract
The forebrain includes the cerebral cortex, the thalamus, and the striatum and globus pallidus (GP) in the subpallium. The formation of these structures and their interconnections by specific axonal tracts take place in a precise and orchestrated time and spatial-dependent manner during development. However, the knowledge of the molecular and cellular mechanisms that are involved is rather limited. Moreover, while many extracellular cues and specific receptors have been shown to play a role in different aspects of nervous system development, including neuron migration and axon guidance, examples of intracellular signaling effectors involved in these processes are sparse. In the present work, we have shown that the atypical RhoGTPase, Rnd3, is expressed very early during brain development and keeps a dynamic expression in several brain regions including the cortex, the thalamus, and the subpallium. By using a gene-trap allele (Rnd3gt ) and immunological techniques, we have shown that Rnd3gt/gt embryos display severe defects in striatal and thalamocortical axonal projections (SAs and TCAs, respectively) and defects in GP formation already at early stages. Surprisingly, the corridor, an important intermediate target for TCAs is still present in these mutants. Mechanistically, a conditional genetic deletion approach revealed that Rnd3 is primarily required for the normal development of Medial Ganglionic Eminence-derived structures, such as the GP, and therefore acts non-cell autonomously in SAs and TCAs. In conclusion, we have demonstrated the important role of Rnd3 as an early regulator of subpallium development in vivo and revealed new insights about SAs and TCAs development.
Collapse
Affiliation(s)
| | | | | | | | - Begoña Ballester-Lurbe
- Departamento de Medicina y Cirugía Animal, Facultad de Veterinaria, Universidad CEU Cardenal Herrera, Valencia, Spain
| | - Jose Terrado
- Departamento de Medicina y Cirugía Animal, Facultad de Veterinaria, Universidad CEU Cardenal Herrera, Valencia, Spain
| | - Ignacio Perez-Roger
- Departamento de Medicina y Cirugía Animal, Facultad de Veterinaria, Universidad CEU Cardenal Herrera, Valencia, Spain
| | | | - Joaquim Egea
- IRBLLEIDA/Universitat de Lleida, Serra Húnter associate professor, Lleida, Spain
| |
Collapse
|
22
|
Meyerink BL, Tiwari NK, Pilaz LJ. Ariadne's Thread in the Developing Cerebral Cortex: Mechanisms Enabling the Guiding Role of the Radial Glia Basal Process during Neuron Migration. Cells 2020; 10:E3. [PMID: 33375033 PMCID: PMC7822038 DOI: 10.3390/cells10010003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 12/17/2020] [Accepted: 12/17/2020] [Indexed: 11/16/2022] Open
Abstract
Radial neuron migration in the developing cerebral cortex is a complex journey, starting in the germinal zones and ending in the cortical plate. In mice, migratory distances can reach several hundreds of microns, or millimeters in humans. Along the migratory path, radially migrating neurons slither through cellularly dense and complex territories before they reach their final destination in the cortical plate. This task is facilitated by radial glia, the neural stem cells of the developing cortex. Indeed, radial glia have a unique bipolar morphology, enabling them to serve as guides for neuronal migration. The key guiding structure of radial glia is the basal process, which traverses the entire thickness of the developing cortex. Neurons recognize the basal process as their guide and maintain physical interactions with this structure until the end of migration. Thus, the radial glia basal process plays a key role during radial migration. In this review, we highlight the pathways enabling neuron-basal process interactions during migration, as well as the known mechanisms regulating the morphology of the radial glia basal process. Throughout, we describe how dysregulation of these interactions and of basal process morphology can have profound effects on cortical development, and therefore lead to neurodevelopmental diseases.
Collapse
Affiliation(s)
- Brandon L. Meyerink
- Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, SD 57104, USA; (B.L.M.); (N.K.T.)
- Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, SD 57069, USA
| | - Neeraj K. Tiwari
- Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, SD 57104, USA; (B.L.M.); (N.K.T.)
| | - Louis-Jan Pilaz
- Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, SD 57104, USA; (B.L.M.); (N.K.T.)
- Department of Pediatrics, Sanford School of Medicine, University of South Dakota, Vermillion, SD 57069, USA
| |
Collapse
|
23
|
Wang X, Li S, Ma Y, Xu Y, Ogbuehi AC, Hu X, Acharya A, Haak R, Ziebolz D, Schmalz G, Li H, Gaus S, Lethaus B, Savkovic V, Su Z. Identification of miRNAs as the Crosstalk in the Interaction between Neural Stem/Progenitor Cells and Endothelial Cells. DISEASE MARKERS 2020; 2020:6630659. [PMID: 33381243 PMCID: PMC7758130 DOI: 10.1155/2020/6630659] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 11/22/2020] [Accepted: 11/24/2020] [Indexed: 12/12/2022]
Abstract
AIM This study is aimed at identifying genetic and epigenetic crosstalk molecules and their target drugs involved in the interaction between neural stem/progenitor cells (NSPCs) and endothelial cells (ECs). MATERIALS AND METHODS Datasets pertaining to reciprocal mRNA and noncoding RNA changes induced by the interaction between NSPCs and ECs were obtained from the GEO database. Differential expression analysis (DEA) was applied to identify NSPC-induced EC alterations by comparing the expression profiles between monoculture of ECs and ECs grown in EC/NSPC cocultures. DEA was also utilized to identify EC-induced NSPC alterations by comparing the expression profiles between monoculture of NSPCs and NSPCs grown in EC/NSPC cocultures. The DEGs and DEmiRNAs shared by NSPC-induced EC alterations and EC-induced NSPC alterations were then identified. Furthermore, miRNA crosstalk analysis and functional enrichment analysis were performed, and the relationship between DEmiRNAs and small molecular drug targets/environment chemical compounds was investigated. RESULTS One dataset (GSE29759) was included and analyzed in this study. Six genes (i.e., MMP14, TIMP3, LOXL1, CCK, SMAD6, and HSPA2), three miRNAs (i.e., miR-210, miR-230a, and miR-23b), and three pathways (i.e., Akt, ERK1/2, and BMPs) were identified as crosstalk molecules. Six small molecular drugs (i.e., deptropine, fluphenazine, lycorine, quinostatin, resveratrol, and thiamazole) and seven environmental chemical compounds (i.e., folic acid, dexamethasone, choline, doxorubicin, thalidomide, bisphenol A, and titanium dioxide) were identified to be potential target drugs of the identified DEmiRNAs. CONCLUSION To conclude, three miRNAs (i.e., miR-210, miR-230a, and miR-23b) were identified to be crosstalks linking the interaction between ECs and NSPCs by implicating in both angiogenesis and neurogenesis. These crosstalk molecules might provide a basis for devising novel strategies for fabricating neurovascular models in stem cell tissue engineering.
Collapse
Affiliation(s)
- Xin Wang
- Department of Neurology, First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Simin Li
- Department of Cariology, Endodontology and Periodontology, University Leipzig, Liebigstr. 12, 04103 Leipzig, Germany
| | - Yihong Ma
- Department of Neurology, Graduate School of Medical Sciences, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Yuzhen Xu
- Department of Neurology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Middle Yanchang Road, Shanghai, China
| | | | - Xianda Hu
- Laboratory of Molecular Cell Biology, Beijing Tibetan Hospital, China Tibetology Research Center, 218 Anwaixiaoguanbeili Street, Chaoyang, Beijing 100029, China
| | - Aneesha Acharya
- Faculty of Dentistry, University of Hong Kong, Hong Kong, China
| | - Rainer Haak
- Department of Cariology, Endodontology and Periodontology, University Leipzig, Liebigstr. 12, 04103 Leipzig, Germany
| | - Dirk Ziebolz
- Department of Cariology, Endodontology and Periodontology, University Leipzig, Liebigstr. 12, 04103 Leipzig, Germany
| | - Gerhard Schmalz
- Department of Cariology, Endodontology and Periodontology, University Leipzig, Liebigstr. 12, 04103 Leipzig, Germany
| | - Hanluo Li
- Department of Cranio Maxillofacial Surgery, University Clinic Leipzig, Liebigstr. 12, 04103 Leipzig, Germany
| | - Sebastian Gaus
- Department of Cranio Maxillofacial Surgery, University Clinic Leipzig, Liebigstr. 12, 04103 Leipzig, Germany
| | - Bernd Lethaus
- Department of Cranio Maxillofacial Surgery, University Clinic Leipzig, Liebigstr. 12, 04103 Leipzig, Germany
| | - Vuk Savkovic
- Department of Cranio Maxillofacial Surgery, University Clinic Leipzig, Liebigstr. 12, 04103 Leipzig, Germany
| | - Zhiqiang Su
- Department of Neurology, First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| |
Collapse
|
24
|
Jossin Y. Molecular mechanisms of cell polarity in a range of model systems and in migrating neurons. Mol Cell Neurosci 2020; 106:103503. [PMID: 32485296 DOI: 10.1016/j.mcn.2020.103503] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 04/20/2020] [Accepted: 05/23/2020] [Indexed: 01/09/2023] Open
Abstract
Cell polarity is defined as the asymmetric distribution of cellular components along an axis. Most cells, from the simplest single-cell organisms to highly specialized mammalian cells, are polarized and use similar mechanisms to generate and maintain polarity. Cell polarity is important for cells to migrate, form tissues, and coordinate activities. During development of the mammalian cerebral cortex, cell polarity is essential for neurogenesis and for the migration of newborn but as-yet undifferentiated neurons. These oriented migrations include both the radial migration of excitatory projection neurons and the tangential migration of inhibitory interneurons. In this review, I will first describe the development of the cerebral cortex, as revealed at the cellular level. I will then define the core molecular mechanisms - the Par/Crb/Scrib polarity complexes, small GTPases, the actin and microtubule cytoskeletons, and phosphoinositides/PI3K signaling - that are required for asymmetric cell division, apico-basal and front-rear polarity in model systems, including C elegans zygote, Drosophila embryos and cultured mammalian cells. As I go through each core mechanism I will explain what is known about its importance in radial and tangential migration in the developing mammalian cerebral cortex.
Collapse
Affiliation(s)
- Yves Jossin
- Laboratory of Mammalian Development & Cell Biology, Institute of Neuroscience, Université Catholique de Louvain, Brussels, Belgium.
| |
Collapse
|
25
|
Lian G, Wong T, Lu J, Hu J, Zhang J, Sheen V. Cytoskeletal Associated Filamin A and RhoA Affect Neural Progenitor Specification During Mitosis. Cereb Cortex 2020; 29:1280-1290. [PMID: 29462287 DOI: 10.1093/cercor/bhy033] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Indexed: 12/23/2022] Open
Abstract
Neural progenitor proliferation and cell fate decision from self-renewal to differentiation are crucial factors in determining brain size and morphology. The cytoskeletal dependent regulation of these processes is not entirely known. The actin-binding filamin A (FlnA) was shown to regulate proliferation of progenitors by directing changes in cell cycles proteins such as Cdk1 during G2/M phase. Here we report that functional loss of FlnA not only affects the rate of proliferation by altering cell cycle length but also causes a defect in early differentiation through changes in cell fate specification. FlnA interacts with Rho GTPase RhoA, and FlnA loss impairs RhoA activation. Disruption of either of these cytoskeletal associated proteins delays neurogenesis and promotes neural progenitors to remain in proliferative states. Aurora kinase B (Aurkb) has been implicated in cytokinesis, and peaks in expression during the G2/M phase. Inhibition of FlnA or RhoA impairs Aurkb degradation and alters its localization during mitosis. Overexpression of Aurkb replicates the same delay in neurogenesis seen with loss of FlnA or RhoA. Our findings suggest that shared cytoskeletal processes can direct neural progenitor proliferation by regulating the expression and localization of proteins that are implicated in the cell cycle progression and cell fate specification.
Collapse
Affiliation(s)
- Gewei Lian
- Department of Neurology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Timothy Wong
- Department of Neurology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Jie Lu
- Department of Neurology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Jianjun Hu
- Department of Neurology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Jingping Zhang
- Department of Neurology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Volney Sheen
- Department of Neurology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
26
|
Buchsbaum IY, Kielkowski P, Giorgio G, O'Neill AC, Di Giaimo R, Kyrousi C, Khattak S, Sieber SA, Robertson SP, Cappello S. ECE2 regulates neurogenesis and neuronal migration during human cortical development. EMBO Rep 2020; 21:e48204. [PMID: 32207244 PMCID: PMC7202216 DOI: 10.15252/embr.201948204] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 02/18/2020] [Accepted: 02/21/2020] [Indexed: 11/24/2022] Open
Abstract
During embryonic development, excitatory projection neurons migrate in the cerebral cortex giving rise to organised layers. Periventricular heterotopia (PH) is a group of aetiologically heterogeneous disorders in which a subpopulation of newborn projection neurons fails to initiate their radial migration to the cortex, ultimately resulting in bands or nodules of grey matter lining the lateral ventricles. Although a number of genes have been implicated in its cause, currently they only satisfactorily explain the pathogenesis of the condition for 50% of patients. Novel gene discovery is complicated by the extreme genetic heterogeneity recently described to underlie its cause. Here, we study the neurodevelopmental role of endothelin‐converting enzyme‐2 (ECE2) for which two biallelic variants have been identified in two separate patients with PH. Our results show that manipulation of ECE2 levels in human cerebral organoids and in the developing mouse cortex leads to ectopic localisation of neural progenitors and neurons. We uncover the role of ECE2 in neurogenesis, and mechanistically, we identify its involvement in the generation and secretion of extracellular matrix proteins in addition to cytoskeleton and adhesion.
Collapse
Affiliation(s)
- Isabel Y Buchsbaum
- Max Planck Institute of Psychiatry, Munich, Germany.,Graduate School of Systemic Neurosciences, Ludwig-Maximilians-University Munich, Planegg, Germany
| | - Pavel Kielkowski
- Department of Chemistry, Chair of Organic Chemistry II, Center for Integrated Protein Science (CIPSM), Technische Universität München, Garching bei München, Germany
| | - Grazia Giorgio
- Max Planck Institute of Psychiatry, Munich, Germany.,Graduate School of Systemic Neurosciences, Ludwig-Maximilians-University Munich, Planegg, Germany
| | - Adam C O'Neill
- Department of Women's and Children's Health, University of Otago, Dunedin, New Zealand
| | - Rossella Di Giaimo
- Max Planck Institute of Psychiatry, Munich, Germany.,Department of Biology, University of Naples Federico II, Naples, Italy
| | | | - Shahryar Khattak
- DFG Center for Regenerative Therapies, Dresden (CRTD), Technische Universität Dresden, Dresden, Germany
| | - Stephan A Sieber
- Department of Chemistry, Chair of Organic Chemistry II, Center for Integrated Protein Science (CIPSM), Technische Universität München, Garching bei München, Germany
| | - Stephen P Robertson
- Department of Women's and Children's Health, University of Otago, Dunedin, New Zealand
| | | |
Collapse
|
27
|
Moon HM, Hippenmeyer S, Luo L, Wynshaw-Boris A. LIS1 determines cleavage plane positioning by regulating actomyosin-mediated cell membrane contractility. eLife 2020; 9:51512. [PMID: 32159512 PMCID: PMC7112955 DOI: 10.7554/elife.51512] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Accepted: 03/10/2020] [Indexed: 11/16/2022] Open
Abstract
Heterozygous loss of human PAFAH1B1 (coding for LIS1) results in the disruption of neurogenesis and neuronal migration via dysregulation of microtubule (MT) stability and dynein motor function/localization that alters mitotic spindle orientation, chromosomal segregation, and nuclear migration. Recently, human- induced pluripotent stem cell (iPSC) models revealed an important role for LIS1 in controlling the length of terminal cell divisions of outer radial glial (oRG) progenitors, suggesting cellular functions of LIS1 in regulating neural progenitor cell (NPC) daughter cell separation. Here, we examined the late mitotic stages NPCs in vivo and mouse embryonic fibroblasts (MEFs) in vitro from Pafah1b1-deficient mutants. Pafah1b1-deficient neocortical NPCs and MEFs similarly exhibited cleavage plane displacement with mislocalization of furrow-associated markers, associated with actomyosin dysfunction and cell membrane hyper-contractility. Thus, it suggests LIS1 acts as a key molecular link connecting MTs/dynein and actomyosin, ensuring that cell membrane contractility is tightly controlled to execute proper daughter cell separation.
Collapse
Affiliation(s)
- Hyang Mi Moon
- Department of Pediatrics, Institute for Human Genetics, Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, United States
| | - Simon Hippenmeyer
- Howard Hughes Medical Institute and Department of Biology, Stanford University, Stanford, United States
| | - Liqun Luo
- Howard Hughes Medical Institute and Department of Biology, Stanford University, Stanford, United States
| | - Anthony Wynshaw-Boris
- Department of Pediatrics, Institute for Human Genetics, Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, United States.,Department of Genetics and Genome Sciences, Case Western Reserve University, School of Medicine, Cleveland, United States
| |
Collapse
|
28
|
Veeraval L, O'Leary CJ, Cooper HM. Adherens Junctions: Guardians of Cortical Development. Front Cell Dev Biol 2020; 8:6. [PMID: 32117958 PMCID: PMC7025593 DOI: 10.3389/fcell.2020.00006] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 01/10/2020] [Indexed: 12/01/2022] Open
Abstract
Apical radial glia comprise the pseudostratified neuroepithelium lining the embryonic lateral ventricles and give rise to the extensive repertoire of pyramidal neuronal subtypes of the neocortex. The establishment of a highly apicobasally polarized radial glial morphology is a mandatory prerequisite for cortical development as it governs neurogenesis, neural migration and the integrity of the ventricular wall. As in all epithelia, cadherin-based adherens junctions (AJs) play an obligate role in the maintenance of radial glial apicobasal polarity and neuroepithelial cohesion. In addition, the assembly of resilient AJs is critical to the integrity of the neuroepithelium which must resist the tensile forces arising from increasing CSF volume and other mechanical stresses associated with the expansion of the ventricles in the embryo and neonate. Junctional instability leads to the collapse of radial glial morphology, disruption of the ventricular surface and cortical lamination defects due to failed neuronal migration. The fidelity of cortical development is therefore dependent on AJ assembly and stability. Mutations in genes known to control radial glial junction formation are causative for a subset of inherited cortical malformations (neuronal heterotopias) as well as perinatal hydrocephalus, reinforcing the concept that radial glial junctions are pivotal determinants of successful corticogenesis. In this review we explore the key animal studies that have revealed important insights into the role of AJs in maintaining apical radial glial morphology and function, and as such, have provided a deeper understanding of the aberrant molecular and cellular processes contributing to debilitating cortical malformations. We highlight the reciprocal interactions between AJs and the epithelial polarity complexes that impose radial glial apicobasal polarity. We also discuss the critical molecular networks promoting AJ assembly in apical radial glia and emphasize the role of the actin cytoskeleton in the stabilization of cadherin adhesion – a crucial factor in buffering the mechanical forces exerted as a consequence of cortical expansion.
Collapse
Affiliation(s)
- Lenin Veeraval
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Conor J O'Leary
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Helen M Cooper
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
29
|
Yigit G, Saida K, DeMarzo D, Miyake N, Fujita A, Yang Tan T, White SM, Wadley A, Toliat MR, Motameny S, Franitza M, Stutterd CA, Chong PF, Kira R, Sengoku T, Ogata K, Guillen Sacoto MJ, Fresen C, Beck BB, Nürnberg P, Dieterich C, Wollnik B, Matsumoto N, Altmüller J. The recurrent postzygotic pathogenic variant p.Glu47Lys in RHOA causes a novel recognizable neuroectodermal phenotype. Hum Mutat 2019; 41:591-599. [PMID: 31821646 DOI: 10.1002/humu.23964] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 11/29/2019] [Accepted: 12/02/2019] [Indexed: 01/24/2023]
Abstract
RHOA is a member of the Rho family of GTPases that are involved in fundamental cellular processes including cell adhesion, migration, and proliferation. RHOA can stimulate the formation of stress fibers and focal adhesions and is a key regulator of actomyosin dynamics in various tissues. In a Genematcher-facilitated collaboration, we were able to identify four unrelated individuals with a specific phenotype characterized by hypopigmented areas of the skin, dental anomalies, body asymmetry, and limb length discrepancy due to hemihypotrophy of one half of the body, as well as brain magnetic resonance imaging (MRI) anomalies. Using whole-exome and ultra-deep amplicon sequencing and comparing genomic data of affected and unaffected areas of the skin, we discovered that all four individuals carried the identical RHOA missense variant, c.139G>A; p.Glu47Lys, in a postzygotic state. Molecular modeling and in silico analysis of the affected p.Glu47Lys residue in RHOA indicated that this exchange is predicted to specifically alter the interaction of RHOA with its downstream effectors containing a PKN-type binding domain and thereby disrupts its ability to activate signaling. Our findings indicate that the recurrent postzygotic RHOA missense variant p.Glu47Lys causes a specific mosaic disorder in humans.
Collapse
Affiliation(s)
- Gökhan Yigit
- Institute of Human Genetics, University Medical Center Göttingen, Göttingen, Germany
| | - Ken Saida
- Department of Human Genetics, Graduate School of Medicine, Yokohama City University, Yokohama, Japan
| | - Danielle DeMarzo
- University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Noriko Miyake
- Department of Human Genetics, Graduate School of Medicine, Yokohama City University, Yokohama, Japan
| | - Atsushi Fujita
- Department of Human Genetics, Graduate School of Medicine, Yokohama City University, Yokohama, Japan
| | - Tiong Yang Tan
- Victorian Clinical Genetics Services, Murdoch Children's Research Institute, Melbourne, Australia.,Department of Paediatrics, University of Melbourne, Melbourne, Australia
| | - Susan M White
- Victorian Clinical Genetics Services, Murdoch Children's Research Institute, Melbourne, Australia.,Department of Paediatrics, University of Melbourne, Melbourne, Australia
| | - Alexandrea Wadley
- University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Mohammad R Toliat
- Cologne Center for Genomics, University of Cologne, Cologne, Germany
| | - Susanne Motameny
- Cologne Center for Genomics, University of Cologne, Cologne, Germany
| | - Marek Franitza
- Cologne Center for Genomics, University of Cologne, Cologne, Germany
| | - Chloe A Stutterd
- Victorian Clinical Genetics Services, Murdoch Children's Research Institute, Melbourne, Australia.,Department of Paediatrics, University of Melbourne, Melbourne, Australia
| | - Pin F Chong
- Department of Pediatric Neurology, Fukuoka Children's Hospital, Fukuoka, Japan
| | - Ryutaro Kira
- Department of Pediatric Neurology, Fukuoka Children's Hospital, Fukuoka, Japan
| | - Toru Sengoku
- Department of Biochemistry, Graduate School of Medicine, Yokohama City University, Yokohama, Japan
| | - Kazuhiro Ogata
- Department of Biochemistry, Graduate School of Medicine, Yokohama City University, Yokohama, Japan
| | | | - Christine Fresen
- Department of Psychosomatics and Psychotherapy, University Hospital Cologne, Cologne, Germany
| | - Bodo B Beck
- Institute of Human Genetics, University Hospital Cologne, Cologne, Germany.,Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany.,Center for Rare Diseases Cologne (ZSEK), University of Cologne, Cologne, Germany
| | - Peter Nürnberg
- Cologne Center for Genomics, University of Cologne, Cologne, Germany.,Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Christoph Dieterich
- Department of Internal Medicine III, Partner Site Heidelberg/Mannheim, DZHK (German Centre for Cardiovascular Research), University Hospital Heidelberg, Heidelberg, Germany
| | - Bernd Wollnik
- Institute of Human Genetics, University Medical Center Göttingen, Göttingen, Germany.,Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Göttingen, Germany
| | - Naomichi Matsumoto
- Department of Human Genetics, Graduate School of Medicine, Yokohama City University, Yokohama, Japan
| | - Janine Altmüller
- Cologne Center for Genomics, University of Cologne, Cologne, Germany.,Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| |
Collapse
|
30
|
O'Neill AC, Kyrousi C, Klaus J, Leventer RJ, Kirk EP, Fry A, Pilz DT, Morgan T, Jenkins ZA, Drukker M, Berkovic SF, Scheffer IE, Guerrini R, Markie DM, Götz M, Cappello S, Robertson SP. A Primate-Specific Isoform of PLEKHG6 Regulates Neurogenesis and Neuronal Migration. Cell Rep 2019; 25:2729-2741.e6. [PMID: 30517861 DOI: 10.1016/j.celrep.2018.11.029] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Revised: 09/06/2018] [Accepted: 11/05/2018] [Indexed: 12/24/2022] Open
Abstract
The mammalian neocortex has undergone remarkable changes through evolution. A consequence of such rapid evolutionary events could be a trade-off that has rendered the brain susceptible to certain neurodevelopmental and neuropsychiatric conditions. We analyzed the exomes of 65 patients with the structural brain malformation periventricular nodular heterotopia (PH). De novo coding variants were observed in excess in genes defining a transcriptomic signature of basal radial glia, a cell type linked to brain evolution. In addition, we located two variants in human isoforms of two genes that have no ortholog in mice. Modulating the levels of one of these isoforms for the gene PLEKHG6 demonstrated its role in regulating neuroprogenitor differentiation and neuronal migration via RhoA, with phenotypic recapitulation of PH in human cerebral organoids. This suggests that this PLEKHG6 isoform is an example of a primate-specific genomic element supporting brain development.
Collapse
Affiliation(s)
- Adam C O'Neill
- Department of Women's and Children's Health, University of Otago, Dunedin, New Zealand; Institute of Stem Cell Research, Helmholtz Center, Munich, Germany; Physiological Genomics, Biomedical Center Ludwig-Maximilians-Universitaet, Munich, Germany
| | | | | | - Richard J Leventer
- Department of Neurology, Murdoch Children's Research Institute, Parkville, VIC, Australia; Department of Paediatrics, University of Melbourne, Parkville, VIC, Australia
| | - Edwin P Kirk
- Sydney Children's Hospital, University of New South Wales, Randwick, NSW, Australia; New South Wales Health Pathology, Randwick, NSW, Australia
| | - Andrew Fry
- Institute of Medical Genetics, University Hospital of Wales, Heath Park, Cardiff CF14 4XW, UK
| | - Daniela T Pilz
- West of Scotland Genetics Service, Laboratory Medicine Building, Queen Elizabeth University Hospital, Glasgow G51 4TF, UK
| | - Tim Morgan
- Department of Women's and Children's Health, University of Otago, Dunedin, New Zealand
| | - Zandra A Jenkins
- Department of Women's and Children's Health, University of Otago, Dunedin, New Zealand
| | - Micha Drukker
- Institute of Stem Cell Research, Helmholtz Center, Munich, Germany
| | - Samuel F Berkovic
- Epilepsy Research Centre, Department of Medicine, University of Melbourne, Austin Health, Heidelberg, VIC 3084, Australia
| | - Ingrid E Scheffer
- Epilepsy Research Centre, Department of Medicine, University of Melbourne, Austin Health, Heidelberg, VIC 3084, Australia; The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC 3052, Australia
| | - Renzo Guerrini
- Pediatric Neurology Unit and Laboratories, Children's Hospital A. Meyer-University of Florence, Florence, Italy
| | - David M Markie
- Department of Pathology, University of Otago, Dunedin, New Zealand
| | - Magdalena Götz
- Institute of Stem Cell Research, Helmholtz Center, Munich, Germany; Physiological Genomics, Biomedical Center Ludwig-Maximilians-Universitaet, Munich, Germany; Excellence Cluster of Systems Neurology (SYNERGY), 82152 Planegg/Martinsried, Germany
| | | | - Stephen P Robertson
- Department of Women's and Children's Health, University of Otago, Dunedin, New Zealand.
| |
Collapse
|
31
|
Yang JQ, Kalim KW, Li Y, Zheng Y, Guo F. Ablation of RhoA impairs Th17 cell differentiation and alleviates house dust mite-triggered allergic airway inflammation. J Leukoc Biol 2019; 106:1139-1151. [PMID: 31260596 DOI: 10.1002/jlb.3a0119-025rrr] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2019] [Revised: 06/17/2019] [Accepted: 06/18/2019] [Indexed: 01/10/2023] Open
Abstract
Asthma is a heterogeneous chronic airway inflammation in which Th2 and Th17 cells are key players in its pathogenesis. We have reported that RhoA of Rho GTPases orchestrated glycolysis for Th2 cell differentiation and allergic airway inflammation by the use of a conditional RhoA-deficient mouse line. However, the role of RhoA in Th17 cells remains to be elucidated. In this study, we investigated the effects of RhoA deficiency on Th17 cells in the context of ex vivo cell culture systems and an in vivo house dust mites (HDM)-induced allergic airway inflammation. We found that RhoA deficiency inhibited Th17 differentiation and effector cytokine secretion, which was associated with the downregulations of Stat3 and Rorγt, key Th17 transcription factors. Furthermore, loss of RhoA markedly suppressed Th17 and neutrophil-involved airway inflammation induced by HDM in mice. The infiltrating inflammatory cells in the lungs and bronchoalveolar lavage (BAL) fluids were dramatically reduced in conditional RhoA-deficient mice. Th17 as well as Th2 effector cytokines were suppressed in the airways at both protein and mRNA levels. Interestingly, Y16, a specific RhoA inhibitor, was able to recapitulate the most phenotypes of RhoA genetic deletion in Th17 differentiation and allergic airway inflammation. Our data demonstrate that RhoA is a key regulator of Th17 cell differentiation and function. RhoA might serve as a potential novel therapeutic target for asthma and other inflammatory disorders.
Collapse
Affiliation(s)
- Jun-Qi Yang
- Division of Experimental Hematology and Cancer Biology, Children's Hospital Medical Center, and Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA.,Key Laboratory of National Health Commission on Parasitic Disease Control and Prevention, Jiangsu Provincial Key Laboratory on Parasitic and Vector Control, Jiangsu Institute of Parasitic Diseases and Public Health Research Center, Jiangnan University, Wuxi, Jiangsu, China
| | - Khalid W Kalim
- Division of Experimental Hematology and Cancer Biology, Children's Hospital Medical Center, and Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Yuan Li
- Division of Experimental Hematology and Cancer Biology, Children's Hospital Medical Center, and Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Yi Zheng
- Division of Experimental Hematology and Cancer Biology, Children's Hospital Medical Center, and Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Fukun Guo
- Division of Experimental Hematology and Cancer Biology, Children's Hospital Medical Center, and Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| |
Collapse
|
32
|
Xu Z, Chen Y, Chen Y. Spatiotemporal Regulation of Rho GTPases in Neuronal Migration. Cells 2019; 8:cells8060568. [PMID: 31185627 PMCID: PMC6627650 DOI: 10.3390/cells8060568] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2019] [Revised: 06/01/2019] [Accepted: 06/04/2019] [Indexed: 12/17/2022] Open
Abstract
Neuronal migration is essential for the orchestration of brain development and involves several contiguous steps: interkinetic nuclear movement (INM), multipolar–bipolar transition, locomotion, and translocation. Growing evidence suggests that Rho GTPases, including RhoA, Rac, Cdc42, and the atypical Rnd members, play critical roles in neuronal migration by regulating both actin and microtubule cytoskeletal components. This review focuses on the spatiotemporal-specific regulation of Rho GTPases as well as their regulators and effectors in distinct steps during the neuronal migration process. Their roles in bridging extracellular signals and cytoskeletal dynamics to provide optimal structural support to the migrating neurons will also be discussed.
Collapse
Affiliation(s)
- Zhenyan Xu
- The Brain Cognition and Brain Disease Institute, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen 518055, Guangdong, China.
| | - Yuewen Chen
- The Brain Cognition and Brain Disease Institute, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen 518055, Guangdong, China.
- Guangdong Provincial Key Laboratory of Brain Science, Disease and Drug Development, HKUST Shenzhen Research Institute, Shenzhen 518057, Guangdong, China.
| | - Yu Chen
- The Brain Cognition and Brain Disease Institute, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen 518055, Guangdong, China.
- Guangdong Provincial Key Laboratory of Brain Science, Disease and Drug Development, HKUST Shenzhen Research Institute, Shenzhen 518057, Guangdong, China.
| |
Collapse
|
33
|
Fostok SF, El-Sibai M, El-Sabban M, Talhouk RS. Gap Junctions and Wnt Signaling in the Mammary Gland: a Cross-Talk? J Mammary Gland Biol Neoplasia 2019; 24:17-38. [PMID: 30194659 DOI: 10.1007/s10911-018-9411-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Accepted: 09/03/2018] [Indexed: 12/21/2022] Open
Abstract
Connexins (Cxs), the building blocks of gap junctions (GJs), exhibit spatiotemporal patterns of expression and regulate the development and differentiation of the mammary gland, acting via channel-dependent and channel-independent mechanisms. Impaired Cx expression and localization are reported in breast cancer, suggesting a tumor suppressive role for Cxs. The signaling events that mediate the role of GJs in the development and tumorigenesis of the mammary gland remain poorly identified. The Wnt pathways, encompassing the canonical or the Wnt/β-catenin pathway and the noncanonical β-catenin-independent pathway, also play important roles in those processes. Indeed, aberrant Wnt signaling is associated with breast cancer. Despite the coincident roles of Cxs and Wnt pathways, the cross-talk in the breast tissue is poorly defined, although this is reported in a number of other tissues. Our previous studies revealed a channel-independent role for Cx43 in inducing differentiation or suppressing tumorigenesis of mammary epithelial cells by acting as a negative regulator of the Wnt/β-catenin pathway. Here, we provide a brief overview of mammary gland development, with emphasis on the role of Cxs in development and tumorigenesis of this tissue. We also discuss the role of Wnt signaling in similar contexts, and review the literature illustrating interplay between Cxs and Wnt pathways.
Collapse
Affiliation(s)
- Sabreen F Fostok
- Department of Biology, Faculty of Arts and Sciences, American University of Beirut (AUB), P.O. Box: 11-0236, Beirut, Lebanon
| | - Mirvat El-Sibai
- Department of Natural Sciences, School of Arts and Sciences, Lebanese American University (LAU), Beirut, Lebanon
| | - Marwan El-Sabban
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut (AUB), Beirut, Lebanon
| | - Rabih S Talhouk
- Department of Biology, Faculty of Arts and Sciences, American University of Beirut (AUB), P.O. Box: 11-0236, Beirut, Lebanon.
| |
Collapse
|
34
|
Dominguez Gonzalez B, Billion K, Rous S, Pavie B, Lange C, Goodchild R. Excess LINC complexes impair brain morphogenesis in a mouse model of recessive TOR1A disease. Hum Mol Genet 2019; 27:2154-2170. [PMID: 29868845 DOI: 10.1093/hmg/ddy125] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Accepted: 04/06/2018] [Indexed: 01/08/2023] Open
Abstract
Heterozygosity for the TOR1A-Δgag mutation causes semi-penetrant childhood-onset dystonia (OMIM #128100). More recently, homozygous TOR1A mutations were shown to cause severe neurological dysfunction in infants. However, there is little known about the recessive cases, including whether existing reports define the full spectrum of recessive TOR1A disease. Here we describe abnormal brain morphogenesis in ∼30% of Tor1a-/- mouse embryos while, in contrast, this is not found in Tor1aΔgag/Δgag mice. The abnormal Tor1a-/- brains contain excess neural tissue, as well as proliferative zone cytoarchitectural defects related to radial glial cell polarity and cytoskeletal organization. In cultured cells torsinA effects the linker of nucleoskeleton and cytoskeleton (LINC) complex that couples the nucleus and cytoskeleton. Here we identify that torsinA loss elevates LINC complex levels in the proliferative zone, and that genetic reduction of LINC complexes prevents abnormal brain morphogenesis in Tor1a-/- embryos. These data show that Tor1a affects radial glial cells via a LINC complex mediated mechanism. They also predict human TOR1A disease will include incompletely penetrant defects in embryonic brain morphogenesis in cases where mutations ablate TOR1A function.
Collapse
Affiliation(s)
- Beatriz Dominguez Gonzalez
- VIB & KU Leuven Centre for Brain & Disease Research, Campus Gasthuisberg, 3000 Leuven, Belgium.,Department of Neurosciences, Campus Gasthuisberg, KU Leuven, 3000 Leuven, Belgium
| | - Karolien Billion
- VIB & KU Leuven Centre for Brain & Disease Research, Campus Gasthuisberg, 3000 Leuven, Belgium.,Department of Neurosciences, Campus Gasthuisberg, KU Leuven, 3000 Leuven, Belgium
| | - Stef Rous
- VIB & KU Leuven Centre for Brain & Disease Research, Campus Gasthuisberg, 3000 Leuven, Belgium.,Department of Neurosciences, Campus Gasthuisberg, KU Leuven, 3000 Leuven, Belgium
| | - Benjamin Pavie
- VIB & KU Leuven Centre for Brain & Disease Research, Campus Gasthuisberg, 3000 Leuven, Belgium.,VIB Bio Imaging Core, Campus Gasthuisberg, 3000 Leuven, Belgium
| | - Christian Lange
- DFG-Research Center and Cluster of Excellence for Regenerative Therapies (CRTD), Technische Universität Dresden, Fetscherstr. 105, D-01307, Dresden, Germany
| | - Rose Goodchild
- VIB & KU Leuven Centre for Brain & Disease Research, Campus Gasthuisberg, 3000 Leuven, Belgium.,Department of Neurosciences, Campus Gasthuisberg, KU Leuven, 3000 Leuven, Belgium
| |
Collapse
|
35
|
Arai Y, Taverna E. Neural Progenitor Cell Polarity and Cortical Development. Front Cell Neurosci 2017; 11:384. [PMID: 29259543 PMCID: PMC5723293 DOI: 10.3389/fncel.2017.00384] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Accepted: 11/17/2017] [Indexed: 12/15/2022] Open
Abstract
Neurons populating the cerebral cortex are generated during embryonic development from neural stem and progenitor cells in a process called neurogenesis. Neural stem and progenitor cells are classified into several classes based on the different location of mitosis (apical or basal) and polarity features (bipolar, monopolar and non-polar). The polarized architecture of stem cells is linked to the asymmetric localization of proteins, mRNAs and organelles, such as the centrosome and the Golgi apparatus (GA). Polarity affects stem cell function and allows stem cells to integrate environmental cues from distinct niches in the developing cerebral cortex. The crucial role of polarity in neural stem and progenitor cells is highlighted by the fact that impairment of cell polarity is linked to neurodevelopmental disorders such as Down syndrome, Fragile X syndrome, autism spectrum disorders (ASD) and schizophrenia.
Collapse
Affiliation(s)
- Yoko Arai
- Centre for Interdisciplinary Research in Biology (CIRB), Collège de France, CNRS UMR 7241/INSERM U1050, PSL Research University, Paris, France
| | - Elena Taverna
- Department of Evolutionary Genetics, Max Planck Institute for Evolutionary Anthropology (MPG), Leipzig, Germany
| |
Collapse
|
36
|
Yang L, Dai F, Tang L, Le Y, Yao W. Macrophage differentiation induced by PMA is mediated by activation of RhoA/ROCK signaling. J Toxicol Sci 2017; 42:763-771. [PMID: 29142175 DOI: 10.2131/jts.42.763] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
In order to investigate the effects of RhoA/ROCK signaling in macrophage differentiation, we used 100 ng/mL PMA to induce macrophage differentiation from U937 cells in vitro. The observation of cell morphology and the expression of CD68 and SR-A were performed to confirm the differentiation induced by PMA. Western blot analysis showed that the expression of ROCK1 and ROCK2 and the phosphorylation of MYPT1 were significantly increased after PMA treatment. Pulldown assay showed that the activation of RhoA was obviously enhanced when U937 cells were treated with PMA. In order to further demonstrate whether RhoA/ROCK signaling could mediate the macrophage differentiation induced by PMA, we successfully suppressed the expression of RhoA, ROCK1 and ROCK2 by performing siRNA technology in U937 cells, respectively. The macrophage differentiation and the expression of CD68 and SR-A were significantly inhibited by the suppression of RhoA, ROCK1 or ROCK2 in PMA-induced U937 cells, indicating that the macrophage differentiation induced by PMA is associated with RhoA/ROCK signaling pathway. In addition, we pretreated U937 cells with Y27632 (ROCK inhibitor, 20 μM) for 30 min and then observed the macrophage differentiation induced by PMA. The result illustrated that Y27632 pretreatment obviously inhibited PMA-induced differentiation and the expression of CD68 and SR-A. In conclusion, the activation of RhoA/ROCK signaling is responsible for the macrophage differentiation induced by PMA.
Collapse
Affiliation(s)
- Lifeng Yang
- School of Pharmacy, Nantong University, China
| | - Fan Dai
- School of Pharmacy, Nantong University, China
| | - Lian Tang
- School of Pharmacy, Nantong University, China
| | - Yulan Le
- School of Pharmacy, Nantong University, China
| | - Wenjuan Yao
- School of Pharmacy, Nantong University, China
| |
Collapse
|
37
|
Liu M, Zhang Z, Sampson L, Zhou X, Nalapareddy K, Feng Y, Akunuru S, Melendez J, Davis AK, Bi F, Geiger H, Xin M, Zheng Y. RHOA GTPase Controls YAP-Mediated EREG Signaling in Small Intestinal Stem Cell Maintenance. Stem Cell Reports 2017; 9:1961-1975. [PMID: 29129684 PMCID: PMC5785633 DOI: 10.1016/j.stemcr.2017.10.004] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2017] [Revised: 10/10/2017] [Accepted: 10/11/2017] [Indexed: 02/05/2023] Open
Abstract
RHOA, a founding member of the Rho GTPase family, is critical for actomyosin dynamics, polarity, and morphogenesis in response to developmental cues, mechanical stress, and inflammation. In murine small intestinal epithelium, inducible RHOA deletion causes a loss of epithelial polarity, with disrupted villi and crypt organization. In the intestinal crypts, RHOA deficiency results in reduced cell proliferation, increased apoptosis, and a loss of intestinal stem cells (ISCs) that mimic effects of radiation damage. Mechanistically, RHOA loss reduces YAP signaling of the Hippo pathway and affects YAP effector epiregulin (EREG) expression in the crypts. Expression of an active YAP (S112A) mutant rescues ISC marker expression, ISC regeneration, and ISC-associated Wnt signaling, but not defective epithelial polarity, in RhoA knockout mice, implicating YAP in RHOA-regulated ISC function. EREG treatment or active β-catenin Catnblox(ex3) mutant expression rescues the RhoA KO ISC phenotypes. Thus, RHOA controls YAP-EREG signaling to regulate intestinal homeostasis and ISC regeneration.
Collapse
Affiliation(s)
- Ming Liu
- Division of Experimental Hematology & Cancer Biology, Cincinnati Children's Hospital Research Foundation, 3333 Burnet Avenue, Cincinnati, OH 45229, USA; Department of Medical Oncology, West China Hospital, Sichuan University, Chengdu, Sichuan Province 610041, China
| | - Zheng Zhang
- Division of Experimental Hematology & Cancer Biology, Cincinnati Children's Hospital Research Foundation, 3333 Burnet Avenue, Cincinnati, OH 45229, USA
| | - Leesa Sampson
- Division of Experimental Hematology & Cancer Biology, Cincinnati Children's Hospital Research Foundation, 3333 Burnet Avenue, Cincinnati, OH 45229, USA
| | - Xuan Zhou
- Division of Experimental Hematology & Cancer Biology, Cincinnati Children's Hospital Research Foundation, 3333 Burnet Avenue, Cincinnati, OH 45229, USA
| | - Kodandaramireddy Nalapareddy
- Division of Experimental Hematology & Cancer Biology, Cincinnati Children's Hospital Research Foundation, 3333 Burnet Avenue, Cincinnati, OH 45229, USA
| | - Yuxin Feng
- Division of Experimental Hematology & Cancer Biology, Cincinnati Children's Hospital Research Foundation, 3333 Burnet Avenue, Cincinnati, OH 45229, USA
| | - Shailaja Akunuru
- Division of Experimental Hematology & Cancer Biology, Cincinnati Children's Hospital Research Foundation, 3333 Burnet Avenue, Cincinnati, OH 45229, USA
| | - Jaime Melendez
- Division of Experimental Hematology & Cancer Biology, Cincinnati Children's Hospital Research Foundation, 3333 Burnet Avenue, Cincinnati, OH 45229, USA; Laboratorio de Bioquímica y Biología Molecular Depto. Farmacia Facultad de Química, P. Universidad Católica de Chile, Santiago, Chile
| | - Ashley Kuenzi Davis
- Division of Experimental Hematology & Cancer Biology, Cincinnati Children's Hospital Research Foundation, 3333 Burnet Avenue, Cincinnati, OH 45229, USA
| | - Feng Bi
- Department of Medical Oncology, West China Hospital, Sichuan University, Chengdu, Sichuan Province 610041, China
| | - Hartmut Geiger
- Division of Experimental Hematology & Cancer Biology, Cincinnati Children's Hospital Research Foundation, 3333 Burnet Avenue, Cincinnati, OH 45229, USA
| | - Mei Xin
- Division of Experimental Hematology & Cancer Biology, Cincinnati Children's Hospital Research Foundation, 3333 Burnet Avenue, Cincinnati, OH 45229, USA
| | - Yi Zheng
- Division of Experimental Hematology & Cancer Biology, Cincinnati Children's Hospital Research Foundation, 3333 Burnet Avenue, Cincinnati, OH 45229, USA.
| |
Collapse
|
38
|
Cytoskeletal Stability in the Auditory Organ In Vivo: RhoA Is Dispensable for Wound Healing but Essential for Hair Cell Development. eNeuro 2017; 4:eN-NWR-0149-17. [PMID: 28929130 PMCID: PMC5602105 DOI: 10.1523/eneuro.0149-17.2017] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Revised: 08/24/2017] [Accepted: 08/28/2017] [Indexed: 01/03/2023] Open
Abstract
Wound healing in the inner ear sensory epithelia is performed by the apical domains of supporting cells (SCs). Junctional F-actin belts of SCs are thin during development but become exceptionally thick during maturation. The functional significance of the thick belts is not fully understood. We have studied the role of F-actin belts during wound healing in the developing and adult cochlea of mice in vivo. We show that the thick belts serve as intracellular scaffolds that preserve the positions of surviving cells in the cochlear sensory epithelium. Junctions associated with the thick F-actin belts did not readily disassemble during wound healing. To compensate for this, basolateral membranes of SCs participated in the closure of surface breach. Because not only neighboring but also distant SCs contributed to wound healing by basolateral protrusions, this event appears to be triggered by contact-independent diffusible signals. In the search for regulators of wound healing, we inactivated RhoA in SCs, which, however, did not limit wound healing. RhoA inactivation in developing outer hair cells (OHCs) caused myosin II delocalization from the perijunctional domain and apical cell-surface enlargement. These abnormalities led to the extrusion of OHCs from the epithelium. These results demonstrate the importance of stability of the apical domain, both in wound repair by SCs and in development of OHCs, and that only this latter function is regulated by RhoA. Because the correct cytoarchitecture of the cochlear sensory epithelium is required for normal hearing, the stability of cell apices should be maintained in regenerative and protective interventions.
Collapse
|
39
|
Racicot K, VanOeveren S, Alberts A. Viral Hijacking of Formins in Neurodevelopmental Pathologies. Trends Mol Med 2017; 23:778-785. [DOI: 10.1016/j.molmed.2017.07.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Revised: 07/12/2017] [Accepted: 07/16/2017] [Indexed: 01/19/2023]
|
40
|
Johnson CA, Wright CE, Ghashghaei HT. Regulation of cytokinesis during corticogenesis: focus on the midbody. FEBS Lett 2017; 591:4009-4026. [PMID: 28493553 DOI: 10.1002/1873-3468.12676] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Revised: 04/23/2017] [Accepted: 05/07/2017] [Indexed: 12/21/2022]
Abstract
Development of the cerebral cortices depends on tight regulation of cell divisions. In this system, stem and progenitor cells undergo symmetric and asymmetric divisions to ultimately produce neurons that establish the layers of the cortex. Cell division culminates with the formation of the midbody, a transient organelle that establishes the site of abscission between nascent daughter cells. During cytokinetic abscission, the final stage of cell division, one daughter cell will inherit the midbody remnant, which can then maintain or expel the remnant, but mechanisms and circumstances influencing this decision are unclear. This review describes the midbody and its constituent proteins, as well as the known consequences of their manipulation during cortical development. The potential functional relevance of midbody mechanisms is discussed.
Collapse
Affiliation(s)
- Caroline A Johnson
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, USA.,Comparative Biomedical Sciences Graduate Program, Neurosciences Concentration Area, North Carolina State University, Raleigh, NC, USA
| | - Catherine E Wright
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, USA
| | - H Troy Ghashghaei
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, USA.,Comparative Biomedical Sciences Graduate Program, Neurosciences Concentration Area, North Carolina State University, Raleigh, NC, USA.,Program in Genetics, North Carolina State University, Raleigh, NC, USA.,Keck Center for Behavioral Biology, North Carolina State University, Raleigh, NC, USA
| |
Collapse
|
41
|
Loss of Usp9x disrupts cell adhesion, and components of the Wnt and Notch signaling pathways in neural progenitors. Sci Rep 2017; 7:8109. [PMID: 28808228 PMCID: PMC5556043 DOI: 10.1038/s41598-017-05451-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Accepted: 06/02/2017] [Indexed: 12/31/2022] Open
Abstract
Development of neural progenitors depends upon the coordination of appropriate intrinsic responses to extrinsic signalling pathways. Here we show the deubiquitylating enzyme, Usp9x regulates components of both intrinsic and extrinsic fate determinants. Nestin-cre mediated ablation of Usp9x from embryonic neural progenitors in vivo resulted in a transient disruption of cell adhesion and apical-basal polarity and, an increased number and ectopic localisation of intermediate neural progenitors. In contrast to other adhesion and polarity proteins, levels of β-catenin protein, especially S33/S37/T41 phospho-β-catenin, were markedly increased in Usp9x−/Y embryonic cortices. Loss of Usp9x altered composition of the β-catenin destruction complex possibly impeding degradation of S33/S37/T41 phospho-β-catenin. Pathway analysis of transcriptomic data identified Wnt signalling as significantly affected in Usp9x−/Y embryonic brains. Depletion of Usp9x in cultured human neural progenitors resulted in Wnt-reporter activation. Usp9x also regulated components of the Notch signalling pathway. Usp9x co-localized and associated with both Itch and Numb in embryonic neocortices. Loss of Usp9x led to decreased Itch and Numb levels, and a concomitant increase in levels of the Notch intracellular domain as well as, increased expression of the Notch target gene Hes5. Therefore Usp9x modulates and potentially coordinates multiple fate determinants in neural progenitors.
Collapse
|
42
|
El Zowalaty AE, Li R, Zheng Y, Lydon JP, DeMayo FJ, Ye X. Deletion of RhoA in Progesterone Receptor-Expressing Cells Leads to Luteal Insufficiency and Infertility in Female Mice. Endocrinology 2017; 158:2168-2178. [PMID: 28498971 PMCID: PMC5505209 DOI: 10.1210/en.2016-1796] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Accepted: 05/08/2017] [Indexed: 12/22/2022]
Abstract
Ras homolog gene family, member A (RhoA) is widely expressed throughout the female reproductive system. To assess its role in progesterone receptor-expressing cells, we generated RhoA conditional knockout mice RhoAd/d (RhoAf/f-Pgr-Cre+/-). RhoAd/d female mice had comparable mating activity, serum luteinizing hormone, prolactin, and estradiol levels and ovulation with control but were infertile with progesterone insufficiency, indicating impaired steroidogenesis in RhoAd/d corpus luteum (CL). RhoA was highly expressed in wild-type luteal cells and conditionally deleted in RhoAd/d CL. Gestation day 3.5 (D3.5) RhoAd/d ovaries had reduced numbers of CL, less defined corpus luteal cord formation, and disorganized CL collagen IV staining. RhoAd/d CL had lipid droplet and free cholesterol accumulation, indicating the availability of cholesterol for steroidogenesis, but disorganized β-actin and vimentin staining, indicating disrupted cytoskeleton integrity. Cytoskeleton is important for cytoplasmic cholesterol movement to mitochondria and for regulating mitochondria. Dramatically reduced expression of mitochondrial markers heat shock protein 60 (HSP60), voltage-dependent anion channel, and StAR was detected in RhoAd/d CL. StAR carries out the rate-limiting step of steroidogenesis. StAR messenger RNA expression was reduced in RU486-treated D3.5 wild-type CL and tended to be induced in progesterone-treated D3.5 RhoAd/d CL, with parallel changes of HSP60 expression. These data demonstrated the in vivo function of RhoA in CL luteal cell cytoskeleton integrity, cholesterol transport, StAR expression, and progesterone synthesis, and a positive feedback on StAR expression in CL by progesterone signaling. These findings provide insights into mechanisms of progesterone insufficiency.
Collapse
Affiliation(s)
- Ahmed E. El Zowalaty
- Department of Physiology and Pharmacology, College of Veterinary Medicine, University of Georgia, Athens, Georgia 30602
- Interdisciplinary Toxicology Program, University of Georgia, Athens, Georgia 30602
| | - Rong Li
- Department of Physiology and Pharmacology, College of Veterinary Medicine, University of Georgia, Athens, Georgia 30602
- Interdisciplinary Toxicology Program, University of Georgia, Athens, Georgia 30602
| | - Yi Zheng
- Division of Experimental Hematology and Cancer Biology, Children’s Hospital Research Foundation, Cincinnati, Ohio 45229
| | - John P. Lydon
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas 77030
| | - Francesco J. DeMayo
- Reproductive and Developmental Biology Laboratory/Pregnancy and Female Reproduction Group, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina 27709
| | - Xiaoqin Ye
- Department of Physiology and Pharmacology, College of Veterinary Medicine, University of Georgia, Athens, Georgia 30602
- Interdisciplinary Toxicology Program, University of Georgia, Athens, Georgia 30602
| |
Collapse
|
43
|
Jossin Y, Lee M, Klezovitch O, Kon E, Cossard A, Lien WH, Fernandez TE, Cooper JA, Vasioukhin V. Llgl1 Connects Cell Polarity with Cell-Cell Adhesion in Embryonic Neural Stem Cells. Dev Cell 2017; 41:481-495.e5. [PMID: 28552558 DOI: 10.1016/j.devcel.2017.05.002] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Revised: 04/11/2017] [Accepted: 05/01/2017] [Indexed: 10/19/2022]
Abstract
Malformations of the cerebral cortex (MCCs) are devastating developmental disorders. We report here that mice with embryonic neural stem-cell-specific deletion of Llgl1 (Nestin-Cre/Llgl1fl/fl), a mammalian ortholog of the Drosophila cell polarity gene lgl, exhibit MCCs resembling severe periventricular heterotopia (PH). Immunohistochemical analyses and live cortical imaging of PH formation revealed that disruption of apical junctional complexes (AJCs) was responsible for PH in Nestin-Cre/Llgl1fl/fl brains. While it is well known that cell polarity proteins govern the formation of AJCs, the exact mechanisms remain unclear. We show that LLGL1 directly binds to and promotes internalization of N-cadherin, and N-cadherin/LLGL1 interaction is inhibited by atypical protein kinase C-mediated phosphorylation of LLGL1, restricting the accumulation of AJCs to the basolateral-apical boundary. Disruption of the N-cadherin-LLGL1 interaction during cortical development in vivo is sufficient for PH. These findings reveal a mechanism responsible for the physical and functional connection between cell polarity and cell-cell adhesion machineries in mammalian cells.
Collapse
Affiliation(s)
- Yves Jossin
- Division of Basic Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; Mammalian Development & Cell Biology Unit, Institute of Neuroscience, Université Catholique de Louvain, 1200 Brussels, Belgium.
| | - Minhui Lee
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; Molecular and Cellular Biology Program, University of Washington, Seattle, WA 98195, USA
| | - Olga Klezovitch
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Elif Kon
- Mammalian Development & Cell Biology Unit, Institute of Neuroscience, Université Catholique de Louvain, 1200 Brussels, Belgium
| | - Alexia Cossard
- Mammalian Development & Cell Biology Unit, Institute of Neuroscience, Université Catholique de Louvain, 1200 Brussels, Belgium
| | - Wen-Hui Lien
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; Molecular and Cellular Biology Program, University of Washington, Seattle, WA 98195, USA
| | - Tania E Fernandez
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Jonathan A Cooper
- Division of Basic Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; Molecular and Cellular Biology Program, University of Washington, Seattle, WA 98195, USA
| | - Valera Vasioukhin
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; Molecular and Cellular Biology Program, University of Washington, Seattle, WA 98195, USA; Department of Pathology, Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98195, USA.
| |
Collapse
|
44
|
Ravindran E, Hu H, Yuzwa SA, Hernandez-Miranda LR, Kraemer N, Ninnemann O, Musante L, Boltshauser E, Schindler D, Hübner A, Reinecker HC, Ropers HH, Birchmeier C, Miller FD, Wienker TF, Hübner C, Kaindl AM. Homozygous ARHGEF2 mutation causes intellectual disability and midbrain-hindbrain malformation. PLoS Genet 2017; 13:e1006746. [PMID: 28453519 PMCID: PMC5428974 DOI: 10.1371/journal.pgen.1006746] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Revised: 05/12/2017] [Accepted: 04/05/2017] [Indexed: 11/18/2022] Open
Abstract
Mid-hindbrain malformations can occur during embryogenesis through a disturbance of transient and localized gene expression patterns within these distinct brain structures. Rho guanine nucleotide exchange factor (ARHGEF) family members are key for controlling the spatiotemporal activation of Rho GTPase, to modulate cytoskeleton dynamics, cell division, and cell migration. We identified, by means of whole exome sequencing, a homozygous frameshift mutation in the ARHGEF2 as a cause of intellectual disability, a midbrain-hindbrain malformation, and mild microcephaly in a consanguineous pedigree of Kurdish-Turkish descent. We show that loss of ARHGEF2 perturbs progenitor cell differentiation and that this is associated with a shift of mitotic spindle plane orientation, putatively favoring more symmetric divisions. The ARHGEF2 mutation leads to reduction in the activation of the RhoA/ROCK/MLC pathway crucial for cell migration. We demonstrate that the human brain malformation is recapitulated in Arhgef2 mutant mice and identify an aberrant migration of distinct components of the precerebellar system as a pathomechanism underlying the midbrain-hindbrain phenotype. Our results highlight the crucial function of ARHGEF2 in human brain development and identify a mutation in ARHGEF2 as novel cause of a neurodevelopmental disorder.
Collapse
Affiliation(s)
- Ethiraj Ravindran
- Institute of Cell Biology and Neurobiology, Charité University Medicine Berlin, Berlin, Germany
- Department of Pediatric Neurology, Charité University Medicine Berlin, Berlin, Germany
- Sozialpädiatrisches Zentrum (SPZ), Center for Chronic Sick Children, Charité University, Berlin, Germany
| | - Hao Hu
- Max Planck Institute for Molecular Genetics, Berlin, Germany
- Guangzhou Women and Children's Medical Center, Guangzhou, China
| | - Scott A. Yuzwa
- Department of Molecular Genetics, University of Toronto, Toronto, Canada
- Program in Neurosciences & Mental Health, Hospital for Sick Children, Toronto, Canada
| | | | - Nadine Kraemer
- Institute of Cell Biology and Neurobiology, Charité University Medicine Berlin, Berlin, Germany
- Department of Pediatric Neurology, Charité University Medicine Berlin, Berlin, Germany
- Sozialpädiatrisches Zentrum (SPZ), Center for Chronic Sick Children, Charité University, Berlin, Germany
| | - Olaf Ninnemann
- Institute of Cell Biology and Neurobiology, Charité University Medicine Berlin, Berlin, Germany
| | - Luciana Musante
- Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - Eugen Boltshauser
- Department of Pediatric Neurology, University Children's Hospital of Zurich, Zurich, Switzerland
| | - Detlev Schindler
- Department of Human Genetics, University of Würzburg, Würzburg, Germany
| | - Angela Hübner
- Pediatrics, University Hospital, Technical University Dresden, Dresden, Germany
| | - Hans-Christian Reinecker
- Gastrointestinal Unit and Center for the Study of Inflammatory Bowel Disease, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | | | | | - Freda D. Miller
- Department of Molecular Genetics, University of Toronto, Toronto, Canada
- Program in Neurosciences & Mental Health, Hospital for Sick Children, Toronto, Canada
| | | | - Christoph Hübner
- Department of Pediatric Neurology, Charité University Medicine Berlin, Berlin, Germany
| | - Angela M. Kaindl
- Institute of Cell Biology and Neurobiology, Charité University Medicine Berlin, Berlin, Germany
- Department of Pediatric Neurology, Charité University Medicine Berlin, Berlin, Germany
- Sozialpädiatrisches Zentrum (SPZ), Center for Chronic Sick Children, Charité University, Berlin, Germany
- * E-mail:
| |
Collapse
|
45
|
Li XW, Li F, Liu J, Wang Y, Fu W. [Effect of antepartum taurine supplementation in regulating the activity of Rho family factors and promoting the proliferation of neural stem cells in neonatal rats with fetal growth restriction]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2016. [PMID: 27817785 DOI: 10.7499/j.issn.1008-8830.2016.11.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
OBJECTIVE To study the possible effect of antepartum taurine supplementation in regulating the activity of Rho family factors and promoting the proliferation of neural stem cells in neonatal rats with fetal growth restriction (FGR), and to provide a basis for antepartum taurine supplementation to promote brain development in children with FGR. METHODS A total of 24 pregnant Sprague-Dawley rats were randomly divided into three groups: control, FGR, and taurine (n=8 each ). A rat model of FGR was established by food restriction throughout pregnancy. RT-PCR, immunohistochemistry, and Western blot were used to measure the expression of the specific intracellular markers for neural stem cells fatty acid binding protein 7 (FABP7), Rho-associated coiled-coil containing protein kinase 2 (ROCK2), ras homolog gene family, member A (RhoA), and Ras-related C3 botulinum toxin substrate (Rac). RESULTS The FGR group had significantly lower OD value of FABP7-positive cells and mRNA and protein expression of FABP7 than the control group, and the taurine group had significantly higher OD value of FABP7-positive cells and mRNA and protein expression of FABP7 than the FGR group (P<0.05). The FGR group had significantly higher mRNA expression of RhoA and ROCK2 than the control group. The taurine group had significantly higher mRNA expression of RhoA and ROCK2 than the control group and significantly lower expression than the FGR group (P<0.05). The FGR group had significantly lower mRNA expression of Rac than the control group. The taurine group had significantly higher mRNA expression of Rac than the FGR and control groups (P<0.05). The FGR group had significantly higher protein expression of RhoA and ROCK2 than the control group. The taurine group had significantly lower protein expression of RhoA and ROCK2 than the FGR group (P<0.05). CONCLUSIONS Antepartum taurine supplementation can promote the proliferation of neural stem cells in rats with FGR, and its mechanism may be related to the regulation of the activity of Rho family factors.
Collapse
Affiliation(s)
- Xiang-Wen Li
- Department of Neonatology, Rocket Army General Hospital of the Chinese People's Liberation Army, Jinzhou Medical University, Beijing 100700, China.
| | | | | | | | | |
Collapse
|
46
|
Mooney MA, McWeeney SK, Faraone SV, Hinney A, Hebebrand J, Nigg JT, Wilmot B. Pathway analysis in attention deficit hyperactivity disorder: An ensemble approach. Am J Med Genet B Neuropsychiatr Genet 2016; 171:815-26. [PMID: 27004716 PMCID: PMC4983253 DOI: 10.1002/ajmg.b.32446] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Accepted: 03/07/2016] [Indexed: 12/21/2022]
Abstract
Despite a wealth of evidence for the role of genetics in attention deficit hyperactivity disorder (ADHD), specific and definitive genetic mechanisms have not been identified. Pathway analyses, a subset of gene-set analyses, extend the knowledge gained from genome-wide association studies (GWAS) by providing functional context for genetic associations. However, there are numerous methods for association testing of gene sets and no real consensus regarding the best approach. The present study applied six pathway analysis methods to identify pathways associated with ADHD in two GWAS datasets from the Psychiatric Genomics Consortium. Methods that utilize genotypes to model pathway-level effects identified more replicable pathway associations than methods using summary statistics. In addition, pathways implicated by more than one method were significantly more likely to replicate. A number of brain-relevant pathways, such as RhoA signaling, glycosaminoglycan biosynthesis, fibroblast growth factor receptor activity, and pathways containing potassium channel genes, were nominally significant by multiple methods in both datasets. These results support previous hypotheses about the role of regulation of neurotransmitter release, neurite outgrowth and axon guidance in contributing to the ADHD phenotype and suggest the value of cross-method convergence in evaluating pathway analysis results. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Michael A. Mooney
- Division of Bioinformatics & Computational Biology, Department of Medical Informatics & Clinical Epidemiology, Oregon Health & Science University, Portland, Oregon,OHSU Knight Cancer Institute, Portland, Oregon
| | - Shannon K. McWeeney
- Division of Bioinformatics & Computational Biology, Department of Medical Informatics & Clinical Epidemiology, Oregon Health & Science University, Portland, Oregon,OHSU Knight Cancer Institute, Portland, Oregon,Oregon Clinical and Translational Research Institute, Portland, Oregon
| | - Stephen V. Faraone
- Departments of Psychiatry and Neuroscience & Physiology, State University of New York, Syracuse, New York,K.G. Jebsen Centre for Neuropsychiatric Disorders, Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Anke Hinney
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Johannes Hebebrand
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | | | | | - Joel T. Nigg
- Division of Psychology, Department of Psychiatry, Oregon Health & Science University, Portland, Oregon,Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, Oregon
| | - Beth Wilmot
- Division of Bioinformatics & Computational Biology, Department of Medical Informatics & Clinical Epidemiology, Oregon Health & Science University, Portland, Oregon,OHSU Knight Cancer Institute, Portland, Oregon,Oregon Clinical and Translational Research Institute, Portland, Oregon,Correspondence to: Beth Wilmot, Ph.D., Division of Bioinformatics & Computational Biology, Department of Medical Informatics & Clinical Epidemiology, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd., Mail code: CR145, Portland, OR 97239.
| |
Collapse
|
47
|
Ju XC, Hou QQ, Sheng AL, Wu KY, Zhou Y, Jin Y, Wen T, Yang Z, Wang X, Luo ZG. The hominoid-specific gene TBC1D3 promotes generation of basal neural progenitors and induces cortical folding in mice. eLife 2016; 5. [PMID: 27504805 PMCID: PMC5028191 DOI: 10.7554/elife.18197] [Citation(s) in RCA: 113] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Accepted: 08/08/2016] [Indexed: 12/31/2022] Open
Abstract
Cortical expansion and folding are often linked to the evolution of higher intelligence, but molecular and cellular mechanisms underlying cortical folding remain poorly understood. The hominoid-specific gene TBC1D3 undergoes segmental duplications during hominoid evolution, but its role in brain development has not been explored. Here, we found that expression of TBC1D3 in ventricular cortical progenitors of mice via in utero electroporation caused delamination of ventricular radial glia cells (vRGs) and promoted generation of self-renewing basal progenitors with typical morphology of outer radial glia (oRG), which are most abundant in primates. Furthermore, down-regulation of TBC1D3 in cultured human brain slices decreased generation of oRGs. Interestingly, localized oRG proliferation resulting from either in utero electroporation or transgenic expression of TBC1D3, was often found to underlie cortical regions exhibiting folding. Thus, we have identified a hominoid gene that is required for oRG generation in regulating the cortical expansion and folding. DOI:http://dx.doi.org/10.7554/eLife.18197.001 The outer layer of the mammalian brain the cerebral cortex plays a key role in memory, attention, awareness and thought. While rodents have a smooth cortical surface, the cortex of larger mammals such as primates is organized into folds and furrows. These folds increase the amount of cortex that can fit inside the confines of the skull, and are thought to have allowed the evolution of more advanced thought processes. Mutations in various genes are likely to have contributed to the expansion and folding of the cortex. These mutations may not always have involved changes in the instructions encoded within the genes, but might instead have involved changes in the number of copies of a gene. One plausible candidate gene is TBC1D3, which is only found in the great apes and is active in the cortex. The chimpanzee genome contains a single copy of TBC1D3 whereas the human genome contains multiple copies. Ju, Hou et al. have now shown that introducing the TBC1D3 gene into mouse embryos triggers changes in the embryonic cortex. Specifically, this gene increases the number of a type of cell called the outer radial glial cell in the cortex. These cells give rise to new neurons, and are usually rare in mice but abundant in the brains of animals with a folded cortex. Additional experiments using samples of human brain tissue confirmed that TBC1D3 is required for the outer radial glial cells to form. The samples were collected from miscarried fetuses with the informed consent of the patients and following approved protocols and ethical guidelines. Finally, introducing the TBC1D3 gene into the mouse genome also gave rise to animals with a folded cortex, rather than their usual smooth brain surface. Further work is now required to identify how TBC1D3 helps to generate outer radial glial cells, and to work out how these cells cause the cortex to expand. Testing the behavior of mice with the TBC1D3 gene could also uncover the links between cortical folding and thought processes. DOI:http://dx.doi.org/10.7554/eLife.18197.002
Collapse
Affiliation(s)
- Xiang-Chun Ju
- Institute of Neuroscience, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China.,State Key Laboratory of Neuroscience, Chinese Academy of Sciences, Shanghai, China.,Chinese Academy of Sciences University, Beijing, China
| | - Qiong-Qiong Hou
- Institute of Neuroscience, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China.,State Key Laboratory of Neuroscience, Chinese Academy of Sciences, Shanghai, China.,Chinese Academy of Sciences University, Beijing, China
| | - Ai-Li Sheng
- Institute of Neuroscience, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China.,State Key Laboratory of Neuroscience, Chinese Academy of Sciences, Shanghai, China
| | - Kong-Yan Wu
- Institute of Neuroscience, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China.,State Key Laboratory of Neuroscience, Chinese Academy of Sciences, Shanghai, China
| | - Yang Zhou
- The Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Ying Jin
- The Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Tieqiao Wen
- School of Life Sciences, Shanghai University, Shanghai, China
| | - Zhengang Yang
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China
| | - Xiaoqun Wang
- CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai, China.,Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Zhen-Ge Luo
- Institute of Neuroscience, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China.,State Key Laboratory of Neuroscience, Chinese Academy of Sciences, Shanghai, China.,Chinese Academy of Sciences University, Beijing, China.,CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai, China.,ShanghaiTech University, Shanghai, China
| |
Collapse
|
48
|
Kobayashi K, Sano H, Kato S, Kuroda K, Nakamuta S, Isa T, Nambu A, Kaibuchi K, Kobayashi K. Survival of corticostriatal neurons by Rho/Rho-kinase signaling pathway. Neurosci Lett 2016; 630:45-52. [PMID: 27424794 DOI: 10.1016/j.neulet.2016.07.020] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2016] [Revised: 07/07/2016] [Accepted: 07/13/2016] [Indexed: 01/10/2023]
Abstract
Developing cortical neurons undergo a number of sequential developmental events including neuronal survival/apoptosis, and the molecular mechanism underlying each characteristic process has been studied in detail. However, the survival pathway of cortical neurons at mature stages remains largely uninvestigated. We herein focused on mature corticostriatal neurons because of their important roles in various higher brain functions and the spectrum of neurological and neuropsychiatric disorders. The small GTPase Rho is known to control diverse and essential cellular functions through some effector molecules, including Rho-kinase, during neural development. In the present study, we investigated the role of Rho signaling through Rho-kinase in the survival of corticostriatal neurons. We performed the conditional expression of Clostridium botulinum C3 ADP-ribosyltransferase (C3 transferase) or dominant-negative form for Rho-kinase (Rho-K DN), a well-known inhibitor of Rho or Rho-kinase, respectively, in corticostriatal neurons using a dual viral vector approach combining a neuron-specific retrograde gene transfer lentiviral vector and an adeno-associated virus vector. C3 transferase markedly decreased the number of corticostriatal neurons, which was attributed to caspase-3-dependent enhanced apoptosis. In addition, Rho-K DN produced phenotypic defects similar to those caused by C3 transferase. These results indicate that the Rho/Rho-kinase signaling pathway plays a crucial role in the survival of corticostriatal neurons.
Collapse
Affiliation(s)
- Kenta Kobayashi
- Section of Viral Vector Development, National Institute for Physiological Sciences, Okazaki 444-8585, Japan; SOKENDAI (The Graduate University for Advanced Studies), Hayama 240-0193, Japan.
| | - Hiromi Sano
- SOKENDAI (The Graduate University for Advanced Studies), Hayama 240-0193, Japan; Division of System Neurophysiology, National Institute for Physiological Sciences, Okazaki 444-8585, Japan
| | - Shigeki Kato
- Department of Molecular Genetics, Institute of Biomedical Sciences, Fukushima Medical University School of Medicine, Fukushima 960-1295, Japan
| | - Keisuke Kuroda
- Department of Cell Pharmacology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Shinichi Nakamuta
- Department of Cell Pharmacology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Tadashi Isa
- Section of Viral Vector Development, National Institute for Physiological Sciences, Okazaki 444-8585, Japan; SOKENDAI (The Graduate University for Advanced Studies), Hayama 240-0193, Japan; Department of Developmental Physiology, National Institute for Physiological Sciences, Okazaki 444-8585, Japan
| | - Atsushi Nambu
- Section of Viral Vector Development, National Institute for Physiological Sciences, Okazaki 444-8585, Japan; SOKENDAI (The Graduate University for Advanced Studies), Hayama 240-0193, Japan; Division of System Neurophysiology, National Institute for Physiological Sciences, Okazaki 444-8585, Japan
| | - Kozo Kaibuchi
- Department of Cell Pharmacology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Kazuto Kobayashi
- Department of Molecular Genetics, Institute of Biomedical Sciences, Fukushima Medical University School of Medicine, Fukushima 960-1295, Japan
| |
Collapse
|
49
|
Lin SC, Gou GH, Hsia CW, Ho CW, Huang KL, Wu YF, Lee SY, Chen YH. Simulated Microgravity Disrupts Cytoskeleton Organization and Increases Apoptosis of Rat Neural Crest Stem Cells Via Upregulating CXCR4 Expression and RhoA-ROCK1-p38 MAPK-p53 Signaling. Stem Cells Dev 2016; 25:1172-93. [PMID: 27269634 DOI: 10.1089/scd.2016.0040] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Neural crest stem cells (NCSCs) are a population of multipotent stem cells that are distributed broadly in many tissues and organs and are capable of differentiating into a variety of cell types that are dispersed throughout three germ layers. We are interested in studying the effects of simulated microgravity on the survival and self-renewal of NCSCs. NCSCs extracted from the hair follicle bulge region of the rat whisker pad were cultured in vitro, respectively, in a 2D adherent environment and a 3D suspension environment using the rotatory cell culture system (RCCS) to simulate microgravity. We found that rat NCSCs (rNCSCs) cultured in the RCCS for 24 h showed disrupted organization of filamentous actin, increased globular actin level, formation of plasma membrane blebbing and neurite-like artifact, as well as decreased levels of cortactin and vimentin. Interestingly, ∼70% of RCCS-cultured rNCSCs co-expressed cleaved (active) caspase-3 and neuronal markers microtubule-associated protein 2 (MAP2) and Tuj1 instead of NCSC markers, suggesting stress-induced formation of neurite-like artifact in rNCSCs. In addition, rNCSCs showed increased C-X-C chemokine receptor 4 (CXCR4) expression, RhoA GTPase activation, Rho-associated kinase 1 (ROCK1) and p38 mitogen-activated protein kinase (MAPK) phosphorylation, and p53 expression in the nucleus. Incubation of rNCSCs with the Gα protein inhibitor pertussis toxin or CXCR4 siRNA during RCCS-culturing prevented cytoskeleton disorganization and plasma membrane blebbing, and it suppressed apoptosis of rNCSCs. Taken together, we demonstrate for the first time that simulated microgravity disrupts cytoskeleton organization and increases apoptosis of rNCSCs via upregulating CXCR4 expression and the RhoA-ROCK1-p38 MAPK-p53 signaling pathway.
Collapse
Affiliation(s)
- Shing-Chen Lin
- 1 Graduate Institute of Aerospace and Undersea Medicine, National Defense Medical Center , Neihu District, Taipei City, Taiwan
| | - Guo-Hau Gou
- 2 Graduate Institute of Medical Sciences, National Defense Medical Center , Neihu District, Taipei City, Taiwan
| | - Ching-Wu Hsia
- 2 Graduate Institute of Medical Sciences, National Defense Medical Center , Neihu District, Taipei City, Taiwan
| | - Cheng-Wen Ho
- 1 Graduate Institute of Aerospace and Undersea Medicine, National Defense Medical Center , Neihu District, Taipei City, Taiwan .,3 Division of Rehabilitation Medicine, Taoyuan Armed Forces General Hospital , Longtan Township, Taoyuan County, Taiwan
| | - Kun-Lun Huang
- 1 Graduate Institute of Aerospace and Undersea Medicine, National Defense Medical Center , Neihu District, Taipei City, Taiwan .,4 Department of Undersea and Hyperbaric Medicine, Tri-Service General Hospital , Neihu District, Taipei City, Taiwan
| | - Yung-Fu Wu
- 5 Department of Medical Research, Tri-Service General Hospital , Neihu District, Taipei City, Taiwan
| | - Shih-Yu Lee
- 1 Graduate Institute of Aerospace and Undersea Medicine, National Defense Medical Center , Neihu District, Taipei City, Taiwan
| | - Yi-Hui Chen
- 1 Graduate Institute of Aerospace and Undersea Medicine, National Defense Medical Center , Neihu District, Taipei City, Taiwan
| |
Collapse
|
50
|
Akizu N, García MA, Estarás C, Fueyo R, Badosa C, de la Cruz X, Martínez-Balbás MA. EZH2 regulates neuroepithelium structure and neuroblast proliferation by repressing p21. Open Biol 2016; 6:150227. [PMID: 27248655 PMCID: PMC4852452 DOI: 10.1098/rsob.150227] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Accepted: 03/17/2016] [Indexed: 11/12/2022] Open
Abstract
The function of EZH2 as a transcription repressor is well characterized. However, its role during vertebrate development is still poorly understood, particularly in neurogenesis. Here, we uncover the role of EZH2 in controlling the integrity of the neural tube and allowing proper progenitor proliferation. We demonstrate that knocking down the EZH2 in chick embryo neural tubes unexpectedly disrupts the neuroepithelium (NE) structure, correlating with alteration of the Rho pathway, and reduces neural progenitor proliferation. Moreover, we use transcriptional profiling and functional assays to show that EZH2-mediated repression of p21(WAF1/CIP1) contributes to both processes. Accordingly, overexpression of cytoplasmic p21(WAF1/CIP1) induces NE structural alterations and p21(WAF1/CIP1) suppression rescues proliferation defects and partially compensates for the structural alterations and the Rho activity. Overall, our findings describe a new role of EZH2 in controlling the NE integrity in the neural tube to allow proper progenitor proliferation.
Collapse
Affiliation(s)
- Naiara Akizu
- Department of Molecular Genomics, Instituto de Biología Molecular de Barcelona (IBMB), Consejo Superior de Investigaciones Científicas (CSIC), Barcelona 08028, Spain
| | - María Alejandra García
- Department of Molecular Genomics, Instituto de Biología Molecular de Barcelona (IBMB), Consejo Superior de Investigaciones Científicas (CSIC), Barcelona 08028, Spain
| | - Conchi Estarás
- Department of Molecular Genomics, Instituto de Biología Molecular de Barcelona (IBMB), Consejo Superior de Investigaciones Científicas (CSIC), Barcelona 08028, Spain
| | - Raquel Fueyo
- Department of Molecular Genomics, Instituto de Biología Molecular de Barcelona (IBMB), Consejo Superior de Investigaciones Científicas (CSIC), Barcelona 08028, Spain
| | - Carmen Badosa
- Department of Molecular Genomics, Instituto de Biología Molecular de Barcelona (IBMB), Consejo Superior de Investigaciones Científicas (CSIC), Barcelona 08028, Spain
| | - Xavier de la Cruz
- Vall d'Hebron Institute of Research (VHIR), Passeig de la Vall d'Hebron, 119, Barcelona 08035, Spain Institut Català per la Recerca i Estudis Avançats (ICREA), Barcelona 08018, Spain
| | - Marian A Martínez-Balbás
- Department of Molecular Genomics, Instituto de Biología Molecular de Barcelona (IBMB), Consejo Superior de Investigaciones Científicas (CSIC), Barcelona 08028, Spain
| |
Collapse
|