1
|
Sun L, Liu F, He M, Xu J, Wu C, Zhagn S, Gao J, Dai J. Preserving Life: How Retinoic Acid (RA) Enhances Cell Viability and Reduces Apoptosis in Cryopreserved Blastocyst Cells of Pudong Chickens. Cells 2025; 14:504. [PMID: 40214458 PMCID: PMC11988042 DOI: 10.3390/cells14070504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Revised: 03/18/2025] [Accepted: 03/27/2025] [Indexed: 04/14/2025] Open
Abstract
The preservation of chicken embryonic cells is essential for protecting avian genetic resources and enhancing breeding programs. This study investigates the effects of retinoic acid (RA) on the viability, functionality, and adhesion of thawed chicken blastoderm cells (BCs) following cryopreservation. After thawing and culturing the cells for 24 h, RA treatment resulted in significantly higher cell viability and adhesion rates compared to the control group, with the 2.0 μM RA group demonstrating the best outcomes. After 48 and 72 h of culture, similar trends were observed, with the 2.0 μM RA group consistently maintaining the highest cell viability and adhesion rates. Furthermore, immunofluorescence TUNEL assays revealed that RA significantly reduced both early and late apoptosis rates, particularly at a concentration of 2.0 μM, which exhibited a strong protective effect. Flow cytometry analysis indicated that RA treatment enhanced the mitochondrial membrane potential (MMP), reflecting improved cellular health. Analysis of the apoptosis-related genes BAX, BCL-2, and Caspase-3 revealed that moderate RA concentrations promoted the expression of anti-apoptotic factors while also upregulating pro-apoptotic factors, with the 2.0 μM RA group exhibiting the highest expression levels. Cell cycle analysis showed that RA significantly influenced the distribution of BCs across different phases, with the 4.0 μM RA group exhibiting the highest proportion of cells in the G1/G0 phase, suggesting an enhanced tolerance to cryopreservation stress. Conversely, the S phase cell population was notably reduced at higher RA concentrations, indicating potential inhibition of cell proliferation. These results suggest that RA not only significantly enhances the survival rates and mitochondrial function of BCs, but also regulates the cell cycle, providing better conditions for BC cryopreservation. Overall, the addition of RA represents a valuable strategy for optimizing cryopreservation techniques in chicken embryonic cells, with implications for avian genetic resource conservation and breeding strategies.
Collapse
Affiliation(s)
- Lingwei Sun
- Shanghai Municipal Key Laboratory of Agri-Genetics and Breeding, Institute of Animal Science and Veterinary Medicine, Shanghai Academy of Agricultural Sciences, Shanghai 201106, China; (L.S.); (F.L.); (M.H.); (J.X.); (C.W.); (S.Z.)
- Key Laboratory of Livestock and Poultry Resources (Pig) Evaluation and Utilization, Ministry of Agriculture and Rural Affairs, Shanghai 201106, China
| | - Fuqin Liu
- Shanghai Municipal Key Laboratory of Agri-Genetics and Breeding, Institute of Animal Science and Veterinary Medicine, Shanghai Academy of Agricultural Sciences, Shanghai 201106, China; (L.S.); (F.L.); (M.H.); (J.X.); (C.W.); (S.Z.)
- Key Laboratory of Livestock and Poultry Resources (Pig) Evaluation and Utilization, Ministry of Agriculture and Rural Affairs, Shanghai 201106, China
| | - Mengqian He
- Shanghai Municipal Key Laboratory of Agri-Genetics and Breeding, Institute of Animal Science and Veterinary Medicine, Shanghai Academy of Agricultural Sciences, Shanghai 201106, China; (L.S.); (F.L.); (M.H.); (J.X.); (C.W.); (S.Z.)
- Key Laboratory of Livestock and Poultry Resources (Pig) Evaluation and Utilization, Ministry of Agriculture and Rural Affairs, Shanghai 201106, China
| | - Jiehuan Xu
- Shanghai Municipal Key Laboratory of Agri-Genetics and Breeding, Institute of Animal Science and Veterinary Medicine, Shanghai Academy of Agricultural Sciences, Shanghai 201106, China; (L.S.); (F.L.); (M.H.); (J.X.); (C.W.); (S.Z.)
- Key Laboratory of Livestock and Poultry Resources (Pig) Evaluation and Utilization, Ministry of Agriculture and Rural Affairs, Shanghai 201106, China
| | - Caifeng Wu
- Shanghai Municipal Key Laboratory of Agri-Genetics and Breeding, Institute of Animal Science and Veterinary Medicine, Shanghai Academy of Agricultural Sciences, Shanghai 201106, China; (L.S.); (F.L.); (M.H.); (J.X.); (C.W.); (S.Z.)
- Key Laboratory of Livestock and Poultry Resources (Pig) Evaluation and Utilization, Ministry of Agriculture and Rural Affairs, Shanghai 201106, China
| | - Shushan Zhagn
- Shanghai Municipal Key Laboratory of Agri-Genetics and Breeding, Institute of Animal Science and Veterinary Medicine, Shanghai Academy of Agricultural Sciences, Shanghai 201106, China; (L.S.); (F.L.); (M.H.); (J.X.); (C.W.); (S.Z.)
- Key Laboratory of Livestock and Poultry Resources (Pig) Evaluation and Utilization, Ministry of Agriculture and Rural Affairs, Shanghai 201106, China
| | - Jun Gao
- Shanghai Municipal Key Laboratory of Agri-Genetics and Breeding, Institute of Animal Science and Veterinary Medicine, Shanghai Academy of Agricultural Sciences, Shanghai 201106, China; (L.S.); (F.L.); (M.H.); (J.X.); (C.W.); (S.Z.)
- Key Laboratory of Livestock and Poultry Resources (Pig) Evaluation and Utilization, Ministry of Agriculture and Rural Affairs, Shanghai 201106, China
| | - Jianjun Dai
- Shanghai Municipal Key Laboratory of Agri-Genetics and Breeding, Institute of Animal Science and Veterinary Medicine, Shanghai Academy of Agricultural Sciences, Shanghai 201106, China; (L.S.); (F.L.); (M.H.); (J.X.); (C.W.); (S.Z.)
- Key Laboratory of Livestock and Poultry Resources (Pig) Evaluation and Utilization, Ministry of Agriculture and Rural Affairs, Shanghai 201106, China
| |
Collapse
|
2
|
Lodge DJ, Elam HB, Boley AM, Donegan JJ. Discrete hippocampal projections are differentially regulated by parvalbumin and somatostatin interneurons. Nat Commun 2023; 14:6653. [PMID: 37863893 PMCID: PMC10589277 DOI: 10.1038/s41467-023-42484-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 10/12/2023] [Indexed: 10/22/2023] Open
Abstract
People with schizophrenia show hyperactivity in the ventral hippocampus (vHipp) and we have previously demonstrated distinct behavioral roles for vHipp cell populations. Here, we test the hypothesis that parvalbumin (PV) and somatostatin (SST) interneurons differentially innervate and regulate hippocampal pyramidal neurons based on their projection target. First, we use eGRASP to show that PV-positive interneurons form a similar number of synaptic connections with pyramidal cells regardless of their projection target while SST-positive interneurons preferentially target nucleus accumbens (NAc) projections. To determine if these anatomical differences result in functional changes, we used in vivo opto-electrophysiology to show that SST cells also preferentially regulate the activity of NAc-projecting cells. These results suggest vHipp interneurons differentially regulate that vHipp neurons that project to the medial prefrontal cortex (mPFC) and NAc. Characterization of these cell populations may provide potential molecular targets for the treatment schizophrenia and other psychiatric disorders associated with vHipp dysfunction.
Collapse
Affiliation(s)
- Daniel J Lodge
- Department of Pharmacology and Center for Biomedical Neuroscience, University of Texas Health Science Center, San Antonio, TX, 78229, USA
- South Texas Veterans Health Care System, Audie L. Murphy Division, San Antonio, TX, USA
| | - Hannah B Elam
- Department of Pharmacology and Center for Biomedical Neuroscience, University of Texas Health Science Center, San Antonio, TX, 78229, USA
- South Texas Veterans Health Care System, Audie L. Murphy Division, San Antonio, TX, USA
| | - Angela M Boley
- Department of Pharmacology and Center for Biomedical Neuroscience, University of Texas Health Science Center, San Antonio, TX, 78229, USA
- South Texas Veterans Health Care System, Audie L. Murphy Division, San Antonio, TX, USA
| | - Jennifer J Donegan
- Department of Pharmacology and Center for Biomedical Neuroscience, University of Texas Health Science Center, San Antonio, TX, 78229, USA.
- Department of Psychiatry and Behavioral Sciences and Center for Early Life Adversity, Department of Neuroscience, Dell Medical School at the University of Texas at Austin, Austin, TX, 78712, USA.
| |
Collapse
|
3
|
Saito J, Kojima T, Tanifuji S, Kato Y, Oka S, Ichikawa Y, Miyagi E, Tachibana T, Asou T, Yokoyama U. Transcriptome Analysis Reveals Differential Gene Expression between the Closing Ductus Arteriosus and the Patent Ductus Arteriosus in Humans. J Cardiovasc Dev Dis 2021; 8:jcdd8040045. [PMID: 33923468 PMCID: PMC8073410 DOI: 10.3390/jcdd8040045] [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: 03/08/2021] [Revised: 04/08/2021] [Accepted: 04/14/2021] [Indexed: 12/14/2022] Open
Abstract
The ductus arteriosus (DA) immediately starts closing after birth. This dynamic process involves DA-specific properties, including highly differentiated smooth muscle, sparse elastic fibers, and intimal thickening (IT). Although several studies have demonstrated DA-specific gene expressions using animal tissues and human fetuses, the transcriptional profiles of the closing DA and the patent DA remain largely unknown. We performed transcriptome analysis using four human DA samples. The three closing DA samples exhibited typical DA morphology, but the patent DA exhibited aorta-like elastic lamellae and poorly formed IT. A cluster analysis revealed that samples were clearly divided into two major clusters, the closing DA and patent DA clusters, and showed distinct gene expression profiles in IT and the tunica media of the closing DA samples. Cardiac neural crest-related genes such as JAG1 were highly expressed in the tunica media and IT of the closing DA samples compared to the patent DA sample. Abundant protein expressions of jagged 1 and the differentiated smooth muscle marker calponin were observed in the closing DA samples but not in the patent DA sample. Second heart field-related genes such as ISL1 were enriched in the patent DA sample. These data indicate that the patent DA may have different cell lineages compared to the closing DA.
Collapse
Affiliation(s)
- Junichi Saito
- Department of Physiology, Tokyo Medical University, 6-1-1 Shinjuku, Shinjuku-ku, Tokyo 160-8402, Japan; (J.S.); (T.K.); (S.T.); (Y.K.); (S.O.)
| | - Tomoyuki Kojima
- Department of Physiology, Tokyo Medical University, 6-1-1 Shinjuku, Shinjuku-ku, Tokyo 160-8402, Japan; (J.S.); (T.K.); (S.T.); (Y.K.); (S.O.)
- Department of Obstetrics and Gynecology, Yokohama City University, 3-9 Fukuura, Kanazawa-ku, Yokohama, Kanagawa 236-0004, Japan;
| | - Shota Tanifuji
- Department of Physiology, Tokyo Medical University, 6-1-1 Shinjuku, Shinjuku-ku, Tokyo 160-8402, Japan; (J.S.); (T.K.); (S.T.); (Y.K.); (S.O.)
| | - Yuko Kato
- Department of Physiology, Tokyo Medical University, 6-1-1 Shinjuku, Shinjuku-ku, Tokyo 160-8402, Japan; (J.S.); (T.K.); (S.T.); (Y.K.); (S.O.)
| | - Sayuki Oka
- Department of Physiology, Tokyo Medical University, 6-1-1 Shinjuku, Shinjuku-ku, Tokyo 160-8402, Japan; (J.S.); (T.K.); (S.T.); (Y.K.); (S.O.)
| | - Yasuhiro Ichikawa
- Department of Cardiovascular Surgery, Kanagawa Children’s Medical Center, 2-138-4 Mutsukawa, Minami-ku, Yokohama, Kanagawa 232-8555, Japan; (Y.I.); (T.T.); (T.A.)
| | - Etsuko Miyagi
- Department of Obstetrics and Gynecology, Yokohama City University, 3-9 Fukuura, Kanazawa-ku, Yokohama, Kanagawa 236-0004, Japan;
| | - Tsuyoshi Tachibana
- Department of Cardiovascular Surgery, Kanagawa Children’s Medical Center, 2-138-4 Mutsukawa, Minami-ku, Yokohama, Kanagawa 232-8555, Japan; (Y.I.); (T.T.); (T.A.)
| | - Toshihide Asou
- Department of Cardiovascular Surgery, Kanagawa Children’s Medical Center, 2-138-4 Mutsukawa, Minami-ku, Yokohama, Kanagawa 232-8555, Japan; (Y.I.); (T.T.); (T.A.)
| | - Utako Yokoyama
- Department of Physiology, Tokyo Medical University, 6-1-1 Shinjuku, Shinjuku-ku, Tokyo 160-8402, Japan; (J.S.); (T.K.); (S.T.); (Y.K.); (S.O.)
- Correspondence: ; Tel.: +81-3-3351-6141
| |
Collapse
|
4
|
Espina JA, Marchant CL, Barriga EH. Durotaxis: the mechanical control of directed cell migration. FEBS J 2021; 289:2736-2754. [PMID: 33811732 PMCID: PMC9292038 DOI: 10.1111/febs.15862] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 02/23/2021] [Accepted: 04/01/2021] [Indexed: 11/28/2022]
Abstract
Directed cell migration is essential for cells to efficiently migrate in physiological and pathological processes. While migrating in their native environment, cells interact with multiple types of cues, such as mechanical and chemical signals. The role of chemical guidance via chemotaxis has been studied in the past, the understanding of mechanical guidance of cell migration via durotaxis remained unclear until very recently. Nonetheless, durotaxis has become a topic of intensive research and several advances have been made in the study of mechanically guided cell migration across multiple fields. Thus, in this article we provide a state of the art about durotaxis by discussing in silico, in vitro and in vivo data. We also present insights on the general mechanisms by which cells sense, transduce and respond to environmental mechanics, to then contextualize these mechanisms in the process of durotaxis and explain how cells bias their migration in anisotropic substrates. Furthermore, we discuss what is known about durotaxis in vivo and we comment on how haptotaxis could arise from integrating durotaxis and chemotaxis in native environments.
Collapse
Affiliation(s)
- Jaime A Espina
- Mechanisms of Morphogenesis Lab, Gulbenkian Institute of Science (IGC), Oeiras, Portugal
| | - Cristian L Marchant
- Mechanisms of Morphogenesis Lab, Gulbenkian Institute of Science (IGC), Oeiras, Portugal
| | - Elias H Barriga
- Mechanisms of Morphogenesis Lab, Gulbenkian Institute of Science (IGC), Oeiras, Portugal
| |
Collapse
|
5
|
Kukreja S, Udaykumar N, Yogesh B, Sen J. Retinoic acid signaling regulates proliferation and lamina formation in the developing chick optic tectum. Dev Biol 2020; 467:95-107. [PMID: 32919944 DOI: 10.1016/j.ydbio.2020.09.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 09/03/2020] [Accepted: 09/06/2020] [Indexed: 01/05/2023]
Abstract
The retinotectal system has been extensively studied for investigating the mechanism(s) for topographic map formation. The optic tectum, which is composed of multiple laminae, is the major retino recipient structure in the developing avian brain. Laminar development of the tectum results from cell proliferation, differentiation and migration, coordinated in strict temporal and spatial patterns. However, the molecular mechanisms that orchestrate these complex developmental events, have not been fully elucidated. In this study, we have identified the presence of differential retinoic acid (RA) signaling along the rostro-caudal and dorsoventral axis of the tectum. We show for the first time that loss of RA signaling in the anterior optic tectum, leads to an increase in cell proliferation and gross changes in the morphology manifested as defects in lamination. Detailed analysis points to delayed migration of cells as the plausible cause for the defects in lamina formation. Thus, we conclude that in the optic tectum, RA signaling is involved in maintaining cell proliferation and in regulating the formation of the tectal laminae.
Collapse
Affiliation(s)
- Shweta Kukreja
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, 208016, Uttar Pradesh, India; Present address: Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, USA
| | - Niveda Udaykumar
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, 208016, Uttar Pradesh, India
| | - Baba Yogesh
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, 208016, Uttar Pradesh, India; Present address: Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland
| | - Jonaki Sen
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, 208016, Uttar Pradesh, India.
| |
Collapse
|
6
|
Yang X, Liang R, Liu C, Liu JA, Cheung MPL, Liu X, Man OY, Guan XY, Lung HL, Cheung M. SOX9 is a dose-dependent metastatic fate determinant in melanoma. J Exp Clin Cancer Res 2019; 38:17. [PMID: 30642390 PMCID: PMC6330758 DOI: 10.1186/s13046-018-0998-6] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 12/06/2018] [Indexed: 12/03/2022] Open
Abstract
BACKGROUND In this research, we aimed to resolve contradictory results whether SOX9 plays a positive or negative role in melanoma progression and determine whether SOX9 and its closely related member SOX10 share the same or distinct targets in mediating their functions in melanoma. METHODS Immunofluorescence, TCGA database and qPCR were used to analyze the correlation between the expression patterns and levels of SOX9, SOX10 and NEDD9 in melanoma patient samples. AlamarBlue, transwell invasion and colony formation assays in melanoma cell lines were conducted to investigate the epistatic relationship between SOX10 and NEDD9, as well as the effects of graded SOX9 expression levels. Lung metastasis was determined by tail vein injection assay. Live cell imaging was conducted to monitor dynamics of melanoma migratory behavior. RHOA and RAC1 activation assays measured the activity of Rho GTPases. RESULTS High SOX9 expression was predominantly detected in patients with distant melanoma metastases whereas SOX10 was present in the different stages of melanoma. Both SOX9 and SOX10 exhibited distinct but overlapping expression patterns with metastatic marker NEDD9. Accordingly, SOX10 was required for NEDD9 expression, which partly mediated its oncogenic functions in melanoma cells. Compensatory upregulation of SOX9 expression in SOX10-inhibited melanoma cells reduced growth and migratory capacity, partly due to elevated expression of cyclin-dependent kinase inhibitor p21 and lack of NEDD9 induction. Conversely, opposite phenomenon was observed when SOX9 expression was further elevated to a range of high SOX9 expression levels in metastatic melanoma specimens, and that high levels of SOX9 can restore melanoma progression in the absence of SOX10 both in vitro and in vivo. In addition, overexpression of SOX9 can also promote invasiveness of the parental melanoma cells by modulating the expression of various matrix metalloproteinases. SOX10 or high SOX9 expression regulates melanoma mesenchymal migration through the NEDD9-mediated focal adhesion dynamics and Rho GTPase signaling. CONCLUSIONS These results unravel NEDD9 as a common target for SOX10 or high SOX9 to partly mediate their oncogenic events, and most importantly, reconcile previous discrepancies that suboptimal level of SOX9 expression is anti-metastatic whereas high level of SOX9 is metastatic in a heterogeneous population of melanoma.
Collapse
Affiliation(s)
- Xintao Yang
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 21 Sassoon Road, Hong Kong, China
| | - Rui Liang
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 21 Sassoon Road, Hong Kong, China
| | - Chunxi Liu
- Department of Anesthesiology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang China
| | - Jessica Aijia Liu
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 21 Sassoon Road, Hong Kong, China
| | - May Pui Lai Cheung
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 21 Sassoon Road, Hong Kong, China
| | - Xuelai Liu
- Department of Pediatric Surgery, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei China
| | - On Ying Man
- Department of Biology, Faculty of Science, Hong Kong Baptist University, Hong Kong, China
| | - Xin-Yuan Guan
- Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Hong Lok Lung
- Department of Biology, Faculty of Science, Hong Kong Baptist University, Hong Kong, China
| | - Martin Cheung
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 21 Sassoon Road, Hong Kong, China
| |
Collapse
|
7
|
Haworth S, Shungin D, van der Tas JT, Vucic S, Medina-Gomez C, Yakimov V, Feenstra B, Shaffer JR, Lee MK, Standl M, Thiering E, Wang C, Bønnelykke K, Waage J, Jessen LE, Nørrisgaard PE, Joro R, Seppälä I, Raitakari O, Dudding T, Grgic O, Ongkosuwito E, Vierola A, Eloranta AM, West NX, Thomas SJ, McNeil DW, Levy SM, Slayton R, Nohr EA, Lehtimäki T, Lakka T, Bisgaard H, Pennell C, Kühnisch J, Marazita ML, Melbye M, Geller F, Rivadeneira F, Wolvius EB, Franks PW, Johansson I, Timpson NJ. Consortium-based genome-wide meta-analysis for childhood dental caries traits. Hum Mol Genet 2018; 27:3113-3127. [PMID: 29931343 PMCID: PMC6097157 DOI: 10.1093/hmg/ddy237] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Revised: 05/29/2018] [Accepted: 06/14/2018] [Indexed: 12/26/2022] Open
Abstract
Prior studies suggest dental caries traits in children and adolescents are partially heritable, but there has been no large-scale consortium genome-wide association study (GWAS) to date. We therefore performed GWAS for caries in participants aged 2.5-18.0 years from nine contributing centres. Phenotype definitions were created for the presence or absence of treated or untreated caries, stratified by primary and permanent dentition. All studies tested for association between caries and genotype dosage and the results were combined using fixed-effects meta-analysis. Analysis included up to 19 003 individuals (7530 affected) for primary teeth and 13 353 individuals (5875 affected) for permanent teeth. Evidence for association with caries status was observed at rs1594318-C for primary teeth [intronic within ALLC, odds ratio (OR) 0.85, effect allele frequency (EAF) 0.60, P 4.13e-8] and rs7738851-A (intronic within NEDD9, OR 1.28, EAF 0.85, P 1.63e-8) for permanent teeth. Consortium-wide estimated heritability of caries was low [h2 of 1% (95% CI: 0%: 7%) and 6% (95% CI 0%: 13%) for primary and permanent dentitions, respectively] compared with corresponding within-study estimates [h2 of 28% (95% CI: 9%: 48%) and 17% (95% CI: 2%: 31%)] or previously published estimates. This study was designed to identify common genetic variants with modest effects which are consistent across different populations. We found few single variants associated with caries status under these assumptions. Phenotypic heterogeneity between cohorts and limited statistical power will have contributed; these findings could also reflect complexity not captured by our study design, such as genetic effects which are conditional on environmental exposure.
Collapse
Affiliation(s)
- Simon Haworth
- Medical Research Council Integrative Epidemiology Unit at Bristol Medical School, University of Bristol, Bristol BS8 2BN, UK
| | - Dmitry Shungin
- Department of Odontology, Umeå University, Umeå 901 87, Sweden
- Broad Institute of the Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02142, USA
| | - Justin T van der Tas
- Department of Oral and Maxillofacial Surgery, Special Dental Care and Orthodontics
| | - Strahinja Vucic
- Department of Oral and Maxillofacial Surgery, Special Dental Care and Orthodontics
| | - Carolina Medina-Gomez
- The Generation R Study Group
- Department of Internal Medicine
- Department of Epidemiology, Erasmus Medical Center, University Medical Center Rotterdam, Rotterdam 3015 CN, The Netherlands
| | - Victor Yakimov
- Department of Epidemiology Research, Statens Serum Institut, Copenhagen DK-2300, Denmark
| | - Bjarke Feenstra
- Department of Epidemiology Research, Statens Serum Institut, Copenhagen DK-2300, Denmark
| | - John R Shaffer
- Department of Human Genetics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Center for Craniofacial and Dental Genetics, Department of Oral Biology, School of Dental Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Myoung Keun Lee
- Center for Craniofacial and Dental Genetics, Department of Oral Biology, School of Dental Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Marie Standl
- Institute of Epidemiology I, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg D-85764, Germany
| | - Elisabeth Thiering
- Institute of Epidemiology I, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg D-85764, Germany
- Division of Metabolic and Nutritional Medicine, Dr von Hauner Children's Hospital, University of Munich Medical Center, Munich 80337, Germany
| | - Carol Wang
- Division of Obstetrics and Gynaecology, The University of Western Australia, Perth WA 6009, Australia
| | - Klaus Bønnelykke
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofe Hospital, University of Copenhagen, Copenhagen 2730, Denmark
| | - Johannes Waage
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofe Hospital, University of Copenhagen, Copenhagen 2730, Denmark
| | - Leon Eyrich Jessen
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofe Hospital, University of Copenhagen, Copenhagen 2730, Denmark
| | - Pia Elisabeth Nørrisgaard
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofe Hospital, University of Copenhagen, Copenhagen 2730, Denmark
| | - Raimo Joro
- Institute of Biomedicine, School of Medicine, University of Eastern Finland Kuopio Campus, 70211 Kuopio, Finland
| | - Ilkka Seppälä
- Department of Clinical Chemistry, Fimlab Laboratories and Finnish Cardiovascular Research Center Tampere - Faculty of Medicine and Life Sciences, University of Tampere, Tampere 33520, Finland
| | - Olli Raitakari
- Department of Clinical Physiology and Nuclear Medicine, Turku University Hospital, Turku 20520, Finland
- Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku 20520, Finland
| | - Tom Dudding
- Medical Research Council Integrative Epidemiology Unit at Bristol Medical School, University of Bristol, Bristol BS8 2BN, UK
| | - Olja Grgic
- Department of Oral and Maxillofacial Surgery, Special Dental Care and Orthodontics
- The Generation R Study Group
| | | | - Anu Vierola
- Institute of Biomedicine, School of Medicine, University of Eastern Finland Kuopio Campus, 70211 Kuopio, Finland
| | - Aino-Maija Eloranta
- Institute of Biomedicine, School of Medicine, University of Eastern Finland Kuopio Campus, 70211 Kuopio, Finland
| | - Nicola X West
- Bristol Dental School, University of Bristol, Bristol BS1 2LY, UK
| | - Steven J Thomas
- Bristol Dental School, University of Bristol, Bristol BS1 2LY, UK
| | - Daniel W McNeil
- Department of Psychology, Eberly College of Arts and Sciences, West Virginia University, Morgantown, WA 26506-6286, USA
| | - Steven M Levy
- Department of Preventive and Community Dentistry, College of Dentistry, University of Iowa, Cedar Rapids, IA 52242-1010, USA
| | - Rebecca Slayton
- Department of Pediatric Dentistry (Retired), School of Dentistry, University of Washington, Seattle, WA 98195, USA
| | - Ellen A Nohr
- Research Unit for Gynaecology and Obstetrics, Department of Clinical Research, University of Southern Denmark, Odense 5000, Denmark
| | - Terho Lehtimäki
- Department of Clinical Chemistry, Fimlab Laboratories and Finnish Cardiovascular Research Center Tampere - Faculty of Medicine and Life Sciences, University of Tampere, Tampere 33520, Finland
| | - Timo Lakka
- Institute of Biomedicine, School of Medicine, University of Eastern Finland Kuopio Campus, 70211 Kuopio, Finland
- Department of Clinical Physiology and Nuclear Medicine, Kuopio University Hospital, Kuopio 70210, Finland
- Kuopio Research Institute of Exercise Medicine, Kuopio 70100, Finland
| | - Hans Bisgaard
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofe Hospital, University of Copenhagen, Copenhagen 2730, Denmark
| | - Craig Pennell
- Division of Obstetrics and Gynaecology, The University of Western Australia, Perth WA 6009, Australia
| | - Jan Kühnisch
- Department of Conservative Dentistry and Periodontology, University Hospital, Ludwig-Maximilians-Universität München, Munich 80336, Germany
| | - Mary L Marazita
- Department of Human Genetics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Center for Craniofacial and Dental Genetics, Department of Oral Biology, School of Dental Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Mads Melbye
- Department of Epidemiology Research, Statens Serum Institut, Copenhagen DK-2300, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen 2200, Denmark
- Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Frank Geller
- Department of Epidemiology Research, Statens Serum Institut, Copenhagen DK-2300, Denmark
| | - Fernando Rivadeneira
- The Generation R Study Group
- Department of Internal Medicine
- Department of Epidemiology, Erasmus Medical Center, University Medical Center Rotterdam, Rotterdam 3015 CN, The Netherlands
| | - Eppo B Wolvius
- Department of Oral and Maxillofacial Surgery, Special Dental Care and Orthodontics
| | - Paul W Franks
- Department of Clinical Sciences, Genetic and Molecular Epidemiology Unit, Lund University, Malmö 202 13, Sweden
- Department of Public Health and Clinical Medicine, Umeå University, Umeå 901 85, Sweden
- Department of Nutrition, Harvard T. H. Chan School of Public Health, Boston, MA 02115, USA
| | | | - Nicholas J Timpson
- Medical Research Council Integrative Epidemiology Unit at Bristol Medical School, University of Bristol, Bristol BS8 2BN, UK
| |
Collapse
|
8
|
Solanki A, Yanez DC, Ross S, Lau CI, Papaioannou E, Li J, Saldaña JI, Crompton T. Gli3 in fetal thymic epithelial cells promotes thymocyte positive selection and differentiation by repression of Shh. Development 2018; 145:dev.146910. [PMID: 29361554 PMCID: PMC5817998 DOI: 10.1242/dev.146910] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Accepted: 01/03/2018] [Indexed: 12/15/2022]
Abstract
Gli3 is a Hedgehog (Hh)-responsive transcription factor that can function as a transcriptional repressor or activator. We show that Gli3 activity in mouse thymic epithelial cells (TECs) promotes positive selection and differentiation from CD4+ CD8+ to CD4+ CD8- single-positive (SP4) cells in the fetal thymus and that Gli3 represses Shh Constitutive deletion of Gli3, and conditional deletion of Gli3 from TECs, reduced differentiation to SP4, whereas conditional deletion of Gli3 from thymocytes did not. Conditional deletion of Shh from TECs increased differentiation to SP4, and expression of Shh was upregulated in the Gli3-deficient thymus. Use of a transgenic Hh reporter showed that the Hh pathway was active in thymocytes, and increased in the Gli3-deficient fetal thymus. Neutralisation of endogenous Hh proteins in the Gli3-/- thymus restored SP4 differentiation, indicating that Gli3 in TECs promotes SP4 differentiation by repression of Shh Transcriptome analysis showed that Hh-mediated transcription was increased whereas TCR-mediated transcription was decreased in Gli3-/- thymocytes compared with wild type.
Collapse
Affiliation(s)
- Anisha Solanki
- UCL GOS Institute of Child Health, 30 Guilford Street, London WC1N 1EH, UK
| | - Diana C Yanez
- UCL GOS Institute of Child Health, 30 Guilford Street, London WC1N 1EH, UK
| | - Susan Ross
- UCL GOS Institute of Child Health, 30 Guilford Street, London WC1N 1EH, UK
| | - Ching-In Lau
- UCL GOS Institute of Child Health, 30 Guilford Street, London WC1N 1EH, UK
| | | | - Jiawei Li
- UCL GOS Institute of Child Health, 30 Guilford Street, London WC1N 1EH, UK
| | - José Ignacio Saldaña
- UCL GOS Institute of Child Health, 30 Guilford Street, London WC1N 1EH, UK.,School of Health, Sport and Bioscience, University of East London, London E15 4LZ, UK
| | - Tessa Crompton
- UCL GOS Institute of Child Health, 30 Guilford Street, London WC1N 1EH, UK
| |
Collapse
|
9
|
Liu JA, Rao Y, Cheung MPL, Hui MN, Wu MH, Chan LK, Ng IOL, Niu B, Cheah KSE, Sharma R, Hodgson L, Cheung M. Asymmetric localization of DLC1 defines avian trunk neural crest polarity for directional delamination and migration. Nat Commun 2017; 8:1185. [PMID: 29084958 PMCID: PMC5662599 DOI: 10.1038/s41467-017-01107-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Accepted: 08/18/2017] [Indexed: 11/08/2022] Open
Abstract
Following epithelial-mesenchymal transition, acquisition of avian trunk neural crest cell (NCC) polarity is prerequisite for directional delamination and migration, which in turn is essential for peripheral nervous system development. However, how this cell polarization is established and regulated remains unknown. Here we demonstrate that, using the RHOA biosensor in vivo and in vitro, the initiation of NCC polarization is accompanied by highly activated RHOA in the cytoplasm at the cell rear and its fluctuating activity at the front edge. This differential RHOA activity determines polarized NC morphology and motility, and is regulated by the asymmetrically localized RhoGAP Deleted in liver cancer (DLC1) in the cytoplasm at the cell front. Importantly, the association of DLC1 with NEDD9 is crucial for its asymmetric localization and differential RHOA activity. Moreover, NC specifiers, SOX9 and SOX10, regulate NEDD9 and DLC1 expression, respectively. These results present a SOX9/SOX10-NEDD9/DLC1-RHOA regulatory axis to govern NCC migratory polarization.
Collapse
Affiliation(s)
- Jessica Aijia Liu
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Yanxia Rao
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - May Pui Lai Cheung
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Man-Ning Hui
- Department of Obstetrics and Gynaecology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Ming-Hoi Wu
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Lo-Kong Chan
- State Key Laboratory for Liver Research and Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Irene Oi-Lin Ng
- State Key Laboratory for Liver Research and Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Ben Niu
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Kathryn S E Cheah
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Rakesh Sharma
- Proteomics and Metabolomics Core Facility, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Louis Hodgson
- Department of Anatomy and Structural Biology, Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Martin Cheung
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China.
| |
Collapse
|
10
|
|
11
|
Jones BC, Kelley LC, Loskutov YV, Marinak KM, Kozyreva VK, Smolkin MB, Pugacheva EN. Dual Targeting of Mesenchymal and Amoeboid Motility Hinders Metastatic Behavior. Mol Cancer Res 2017; 15:670-682. [PMID: 28235899 DOI: 10.1158/1541-7786.mcr-16-0411] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2016] [Revised: 12/14/2016] [Accepted: 02/04/2017] [Indexed: 01/22/2023]
Abstract
Commonly upregulated in human cancers, the scaffolding protein NEDD9/HEF1 is a known regulator of mesenchymal migration and cancer cell plasticity. However, the functional role of NEDD9 as a regulator of different migration/invasion modes in the context of breast cancer metastasis is currently unknown. Here, it is reported that NEDD9 is necessary for both mesenchymal and amoeboid individual cell migration/invasion in triple-negative breast cancer (TNBC). NEDD9 deficiency results in acquisition of the amoeboid morphology, but severely limits all types of cell motility. Mechanistically, NEDD9 promotes mesenchymal migration via VAV2-dependent Rac1 activation, and depletion of VAV2 impairs the ability of NEDD9 to activate Rac1. In addition, NEDD9 supports a mesenchymal phenotype through stimulating polymerization of actin via promoting CTTN phosphorylation in an AURKA-dependent manner. Interestingly, an increase in RhoA activity in NEDD9-depleted cells does not facilitate a switch to functional amoeboid motility, indicating a role of NEDD9 in the regulation of downstream RhoA signaling effectors. Simultaneous depletion of NEDD9 or inhibition of AURKA in combination with inhibition of the amoeboid driver ROCK results in an additional decrease in cancer cell migration/invasion. Finally, we confirmed that a dual targeting strategy is a viable and efficient therapeutic approach to hinder the metastasis of breast cancer in xenograft models, showcasing the important need for further clinical evaluation of this regimen to impede the spread of disease and improve patient survival.Implications: This study provides new insight into the therapeutic benefit of combining NEDD9 depletion with ROCK inhibition to reduce tumor cell dissemination and discovers a new regulatory role of NEDD9 in the modulation of VAV2-dependent activation of Rac1 and actin polymerization. Mol Cancer Res; 15(6); 670-82. ©2017 AACR.
Collapse
Affiliation(s)
- Brandon C Jones
- Department of Biochemistry, West Virginia University School of Medicine, Morgantown, West Virginia
| | - Laura C Kelley
- West Virginia University Cancer Institute, West Virginia University School of Medicine, Morgantown, West Virginia
| | - Yuriy V Loskutov
- West Virginia University Cancer Institute, West Virginia University School of Medicine, Morgantown, West Virginia
| | - Kristina M Marinak
- West Virginia University Cancer Institute, West Virginia University School of Medicine, Morgantown, West Virginia
| | - Varvara K Kozyreva
- West Virginia University Cancer Institute, West Virginia University School of Medicine, Morgantown, West Virginia
| | - Matthew B Smolkin
- Department of Pathology, West Virginia University School of Medicine, Morgantown, West Virginia
| | - Elena N Pugacheva
- Department of Biochemistry, West Virginia University School of Medicine, Morgantown, West Virginia.
- West Virginia University Cancer Institute, West Virginia University School of Medicine, Morgantown, West Virginia
| |
Collapse
|
12
|
New Insights Into the Roles of Retinoic Acid Signaling in Nervous System Development and the Establishment of Neurotransmitter Systems. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2016; 330:1-84. [PMID: 28215529 DOI: 10.1016/bs.ircmb.2016.09.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Secreted chiefly from the underlying mesoderm, the morphogen retinoic acid (RA) is well known to contribute to the specification, patterning, and differentiation of neural progenitors in the developing vertebrate nervous system. Furthermore, RA influences the subtype identity and neurotransmitter phenotype of subsets of maturing neurons, although relatively little is known about how these functions are mediated. This review provides a comprehensive overview of the roles played by RA signaling during the formation of the central and peripheral nervous systems of vertebrates and highlights its effects on the differentiation of several neurotransmitter systems. In addition, the evolutionary history of the RA signaling system is discussed, revealing both conserved properties and alternate modes of RA action. It is proposed that comparative approaches should be employed systematically to expand our knowledge of the context-dependent cellular mechanisms controlled by the multifunctional signaling molecule RA.
Collapse
|
13
|
Santos LC, Blair DA, Kumari S, Cammer M, Iskratsch T, Herbin O, Alexandropoulos K, Dustin ML, Sheetz MP. Actin polymerization-dependent activation of Cas-L promotes immunological synapse stability. Immunol Cell Biol 2016; 94:981-993. [PMID: 27359298 PMCID: PMC5121033 DOI: 10.1038/icb.2016.61] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Revised: 06/06/2016] [Accepted: 06/20/2016] [Indexed: 02/07/2023]
Abstract
The immunological synapse formed between a T-cell and an antigen-presenting cell is important for cell-cell communication during T-cell-mediated immune responses. Immunological synapse formation begins with stimulation of the T-cell receptor (TCR). TCR microclusters are assembled and transported to the center of the immunological synapse in an actin polymerization-dependent process. However, the physical link between TCR and actin remains elusive. Here we show that lymphocyte-specific Crk-associated substrate (Cas-L), a member of a force sensing protein family, is required for transport of TCR microclusters and for establishing synapse stability. We found that Cas-L is phosphorylated at TCR microclusters in an actin polymerization-dependent fashion. Furthermore, Cas-L participates in a positive feedback loop leading to amplification of Ca2+ signaling, inside-out integrin activation, and actomyosin contraction. We propose a new role for Cas-L in T-cell activation as a mechanical transducer linking TCR microclusters to the underlying actin network and coordinating multiple actin-dependent structures in the immunological synapse. Our studies highlight the importance of mechanotransduction processes in T-cell-mediated immune responses.
Collapse
Affiliation(s)
- Luís C Santos
- Department of Biological Sciences, Columbia UniversityNew YorkNYUSA
- Skirball Institute of Biomolecular Medicine, New York School of MedicineNew YorkNYUSA
- Icahn Medical Institute, Mount Sinai School of MedicineNew YorkNYUSA
| | - David A Blair
- Skirball Institute of Biomolecular Medicine, New York School of MedicineNew YorkNYUSA
| | - Sudha Kumari
- Skirball Institute of Biomolecular Medicine, New York School of MedicineNew YorkNYUSA
| | - Michael Cammer
- Skirball Institute of Biomolecular Medicine, New York School of MedicineNew YorkNYUSA
| | - Thomas Iskratsch
- Department of Biological Sciences, Columbia UniversityNew YorkNYUSA
| | - Olivier Herbin
- Icahn Medical Institute, Mount Sinai School of MedicineNew YorkNYUSA
| | | | - Michael L Dustin
- Skirball Institute of Biomolecular Medicine, New York School of MedicineNew YorkNYUSA
- Kennedy Institute of Rheumatology, University of OxfordHeadingtonUK
| | - Michael P Sheetz
- Department of Biological Sciences, Columbia UniversityNew YorkNYUSA
| |
Collapse
|
14
|
Latasa MJ, Jiménez-Lara AM, Cosgaya JM. Retinoic acid regulates Schwann cell migration via NEDD9 induction by transcriptional and post-translational mechanisms. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2016; 1863:1510-8. [PMID: 27085739 DOI: 10.1016/j.bbamcr.2016.04.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Revised: 03/30/2016] [Accepted: 04/11/2016] [Indexed: 12/14/2022]
Abstract
Schwann cell migration is essential during the regenerative response to nerve injury, however, the factors that regulate this phenomenon are not yet clear. Here we describe that retinoic acid (RA), whose production and signaling activity are greatly enhanced during nerve regeneration, increases Schwann cell migration. This is accompanied by the up-regulation of NEDD9, a member of the CAS family of scaffold proteins previously implicated in migratory and invasive behavior in gliomas, melanomas and the neural crest cells from which Schwann cells derive. This RA-induced NEDD9 accumulation is due to augmented mRNA levels, as well as an increase of NEDD9 protein stability. Although all NEDD9 phospho-isoforms present in Schwann cells are induced by the retinoid, the hormone also changes its phosphorylation status, thus altering the ratio between the different isoforms. Silencing NEDD9 in Schwann cells had no effect on basal migratory ability, but completely abrogated RA-induced enhanced migration. Collectively, our results indicate that RA could be a major regulator of Schwann cell migration after nerve injury, thus offering a new insight into peripheral nerve repair.
Collapse
Affiliation(s)
- Maria-Jesus Latasa
- Instituto de Investigaciones Biomédicas, Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, Spain; Department of Endocrine and Nervous System Physiopathology, Arturo Duperier, 4, 28029 Madrid, Spain
| | - Ana María Jiménez-Lara
- Instituto de Investigaciones Biomédicas, Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, Spain; Department of Endocrine and Nervous System Physiopathology, Arturo Duperier, 4, 28029 Madrid, Spain
| | - Jose Miguel Cosgaya
- Instituto de Investigaciones Biomédicas, Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, Spain; Department of Endocrine and Nervous System Physiopathology, Arturo Duperier, 4, 28029 Madrid, Spain.
| |
Collapse
|
15
|
Knutson DC, Mitzey AM, Talton LE, Clagett-Dame M. Mice null for NEDD9 (HEF1α) display extensive hippocampal dendritic spine loss and cognitive impairment. Brain Res 2015; 1632:141-55. [PMID: 26683084 DOI: 10.1016/j.brainres.2015.12.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Revised: 11/23/2015] [Accepted: 12/01/2015] [Indexed: 01/28/2023]
Abstract
NEDD9 (neural precursor cell expressed, developmentally down-regulated 9) is a member of the CAS (Crk-associated substrate) family of scaffolding proteins that regulate cell adhesion and migration. A Nedd9 knock-out/lacZ knock-in mouse (Nedd9(-/)(-)) was developed in order to study Nedd9 expression and function in the nervous system. Herein we show that NEDD9 is expressed in the adult brain and is prominently expressed in the hippocampus. Behavioral testing uncovered functional deficits in Nedd9(-)(/)(-) mice. In the Morris water maze test, Nedd9(-)(/)(-) mice showed deficits in both the ability to learn the task as well as in their ability to recall the platform location. There was no change in the gross morphology of the hippocampus, and stereological analysis of BrdU-labeled newly formed hippocampal cells suggested that this defect is not secondary to altered neurogenesis. However, analysis of the hippocampus revealed extensive loss of dendritic spine density in both the dentate gyrus (DG) and CA1 regions. Spine loss occurred equally across all branch orders and regions of the dendrite. Analysis of spine density in Nedd9(-)(/)(-) mice at 1.5, 6 and 10 months revealed an age-dependent spine loss. This work shows that NEDD9 is required for the maintenance of dendritic spines in the hippocampus, and suggests it could play a role in learning and memory.
Collapse
Affiliation(s)
- D C Knutson
- Department of Biochemistry, University of Wisconsin-Madison, 433 Babcock Drive, Madison, WI 53706, USA
| | - A M Mitzey
- Department of Biochemistry, University of Wisconsin-Madison, 433 Babcock Drive, Madison, WI 53706, USA
| | - L E Talton
- Behavioral Testing Core Facility, University of California, Los Angeles, CA 90095, USA
| | - M Clagett-Dame
- Department of Biochemistry, University of Wisconsin-Madison, 433 Babcock Drive, Madison, WI 53706, USA; Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, 777 Highland Avenue, Madison, WI 53705, USA.
| |
Collapse
|
16
|
Deneka A, Korobeynikov V, Golemis EA. Embryonal Fyn-associated substrate (EFS) and CASS4: The lesser-known CAS protein family members. Gene 2015; 570:25-35. [PMID: 26119091 DOI: 10.1016/j.gene.2015.06.062] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Accepted: 06/23/2015] [Indexed: 01/15/2023]
Abstract
The CAS (Crk-associated substrate) adaptor protein family consists of four members: CASS1/BCAR1/p130Cas, CASS2/NEDD9/HEF1/Cas-L, CASS3/EFS/Sin and CASS4/HEPL. While CAS proteins lack enzymatic activity, they contain specific recognition and binding sites for assembly of larger signaling complexes that are essential for cell proliferation, survival, migration, and other processes. All family members are intermediates in integrin-dependent signaling pathways mediated at focal adhesions, and associate with FAK and SRC family kinases to activate downstream effectors regulating the actin cytoskeleton. Most studies of CAS proteins to date have been focused on the first two members, BCAR1 and NEDD9, with altered expression of these proteins now appreciated as influencing disease development and prognosis for cancer and other serious pathological conditions. For these family members, additional mechanisms of action have been defined in receptor tyrosine kinase (RTK) signaling, estrogen receptor signaling or cell cycle progression, involving discrete partner proteins such as SHC, NSP proteins, or AURKA. By contrast, EFS and CASS4 have been less studied, although structure-function analyses indicate they conserve many elements with the better-known family members. Intriguingly, a number of recent studies have implicated these proteins in immune system function, and the pathogenesis of developmental disorders, autoimmune disorders including Crohn's disease, Alzheimer's disease, cancer and other diseases. In this review, we summarize the current understanding of EFS and CASS4 protein function in the context of the larger CAS family group.
Collapse
Affiliation(s)
- Alexander Deneka
- Molecular Therapeutics Program, Fox Chase Cancer Center, Philadelphia, PA, 19111, United States; Kazan Federal University, 420000, Kazan, Russian Federation
| | - Vladislav Korobeynikov
- Molecular Therapeutics Program, Fox Chase Cancer Center, Philadelphia, PA, 19111, United States; Novosibirsk State University, Medical Department, 630090, Novosibirsk, Russian Federation
| | - Erica A Golemis
- Molecular Therapeutics Program, Fox Chase Cancer Center, Philadelphia, PA, 19111, United States.
| |
Collapse
|
17
|
Knutson DC, Clagett-Dame M. A complex RARE is required for the majority of Nedd9 embryonic expression. Transgenic Res 2014; 24:123-34. [PMID: 25120220 PMCID: PMC4274375 DOI: 10.1007/s11248-014-9825-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2014] [Accepted: 08/01/2014] [Indexed: 11/16/2022]
Abstract
Neural precursor cell expressed, developmentally down-regulated 9 (Nedd9, Casl, Hef1, p105cas, Ef1) is a scaffolding protein that assembles complexes involved in regulating cell adhesion, migration, division, and survival. Nedd9 is found very early in the developing embryonic nervous system. A highly conserved complex retinoic acid response element (RARE) is located 485 base pairs (bp) upstream of exon 2B in the promoter of the Nedd9 gene. Mice transgenic for a 5.2 kilobase (kb) region of the 2B Nedd9 promoter containing the RARE upstream of a lacZ reporter gene [Nedd9(RARE)-lacZ] show a large subset of the normal endogenous Nedd9 expression including that in the caudal hindbrain neuroepithelium, spinal cord, dorsal root ganglia (drg) and migrating neural crest (ncc). However, the transgenic mice do not recapitulate the native Nedd9 expression pattern in presumptive rhombomeres (pr) 3 and 5 of the early hindbrain, the base of the neuroepithelium in the midbrain, nor the forebrain telencephalon. Thus, the 5.2 kb region containing the intact RARE drives a large subset of Nedd9 expression, with additional sequences outside of this region needed to define the full complement of expression. When the 5.2 kb construct is modified (eight point mutations) to eliminate responsiveness of the RARE to all-trans retinoic acid (atRA) [Nedd9(mutRARE)-lacZ], virtually all β-galactosidase (β-gal, lacZ) expression is lost. Exposure of Nedd9(RARE)-lacZ transgenic embryos to excess atRA at embryonic day 8.0 (E8.0) leads to rostral ectopic transgene expression within 6 h whereas the Nedd9(mutRARE)-lacZ mutant does not show this effect. Thus the RARE upstream of the Nedd9 2B promoter is necessary for much of the endogenous gene expression during early development as well as ectopic expression in response to atRA.
Collapse
Affiliation(s)
- Danielle C Knutson
- Department of Biochemistry, University of Wisconsin-Madison, 433 Babcock Drive, Madison, WI, 53706, USA
| | | |
Collapse
|
18
|
Adaptors for disorders of the brain? The cancer signaling proteins NEDD9, CASS4, and PTK2B in Alzheimer's disease. Oncoscience 2014; 1:486-503. [PMID: 25594051 PMCID: PMC4278314 DOI: 10.18632/oncoscience.64] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2014] [Accepted: 07/23/2014] [Indexed: 12/19/2022] Open
Abstract
No treatment strategies effectively limit the progression of Alzheimer's disease (AD), a common and debilitating neurodegenerative disorder. The absence of viable treatment options reflects the fact that the pathophysiology and genotypic causes of the disease are not well understood. The advent of genome-wide association studies (GWAS) has made it possible to broadly investigate genotypic alterations driving phenotypic occurrences. Recent studies have associated single nucleotide polymorphisms (SNPs) in two paralogous scaffolding proteins, NEDD9 and CASS4, and the kinase PTK2B, with susceptibility to late-onset AD (LOAD). Intriguingly, NEDD9, CASS4, and PTK2B have been much studied as interacting partners regulating oncogenesis and metastasis, and all three are known to be active in the brain during development and in cancer. However, to date, the majority of studies of these proteins have emphasized their roles in the directly cancer relevant processes of migration and survival signaling. We here discuss evidence for roles of NEDD9, CASS4 and PTK2B in additional processes, including hypoxia, vascular changes, inflammation, microtubule stabilization and calcium signaling, as potentially relevant to the pathogenesis of LOAD. Reciprocally, these functions can better inform our understanding of the action of NEDD9, CASS4 and PTK2B in cancer.
Collapse
|
19
|
|
20
|
Nikonova AS, Gaponova AV, Kudinov AE, Golemis EA. CAS proteins in health and disease: an update. IUBMB Life 2014; 66:387-95. [PMID: 24962474 DOI: 10.1002/iub.1282] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2014] [Accepted: 06/07/2014] [Indexed: 12/30/2022]
Abstract
The CAS family of scaffolding proteins has increasingly attracted scrutiny as important for regulation of cancer-associated signaling. BCAR1 (also known as p130Cas), NEDD9 (HEF1, Cas-L), EFS (Sin), and CASS4 (HEPL) are regulated by and mediate cell attachment, growth factor, and chemokine signaling. Altered expression and activity of CAS proteins are now known to promote metastasis and drug resistance in cancer, influence normal development, and contribute to the pathogenesis of heart and pulmonary disease. In this article, we provide an update on recently published studies describing signals regulating and regulated by CAS proteins, and evidence for biological activity of CAS proteins in normal development, cancer, and other pathological conditions.
Collapse
Affiliation(s)
- Anna S Nikonova
- Program in Developmental Therapeutics, Fox Chase Cancer Center, Philadelphia, PA, USA
| | | | | | | |
Collapse
|
21
|
Kipanyula MJ, Kimaro WH, Yepnjio FN, Aldebasi YH, Farahna M, Nwabo Kamdje AH, Abdel-Magied EM, Seke Etet PF. Signaling pathways bridging fate determination of neural crest cells to glial lineages in the developing peripheral nervous system. Cell Signal 2014; 26:673-682. [PMID: 24378534 DOI: 10.1016/j.cellsig.2013.12.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2013] [Revised: 12/13/2013] [Accepted: 12/22/2013] [Indexed: 11/29/2022]
Abstract
Fate determination of neural crest cells is an essential step for the development of different crest cell derivatives. Peripheral glia development is marked by the choice of the neural crest cells to differentiate along glial lineages. The molecular mechanism underlying fate acquisition is poorly understood. However, recent advances have identified different transcription factors and genes required for the complex instructive signaling process that comprise both local environmental and cell intrinsic cues. Among others, at least the roles of Sox10, Notch, and neuregulin 1 have been documented in both in vivo and in vitro models. Cooperative interactions of such factors appear to be necessary for the switch from multipotent neural crest cells to glial lineage precursors in the peripheral nervous system. This review summarizes recent advances in the understanding of fate determination of neural crest cells into different glia subtypes, together with the potential implications in regenerative medicine.
Collapse
Affiliation(s)
- Maulilio John Kipanyula
- Department of Veterinary Anatomy, Faculty of Veterinary Medicine, Sokoine University of Agriculture, P.O. Box 3016, Chuo Kikuu, Morogoro, Tanzania.
| | - Wahabu Hamisi Kimaro
- Department of Veterinary Anatomy, Faculty of Veterinary Medicine, Sokoine University of Agriculture, P.O. Box 3016, Chuo Kikuu, Morogoro, Tanzania
| | - Faustin N Yepnjio
- Neurology Department, Yaoundé Central Hospital, Department of Internal Medicine and Specialties, University of Yaoundé I, P.O. Box 1937, Yaoundé, Cameroon
| | - Yousef H Aldebasi
- Department of Optometry, College of Applied Medical Sciences, Qassim University, 51452 Buraydah, Saudi Arabia
| | - Mohammed Farahna
- Department of Basic Health Sciences, College of Applied Medical Sciences, Qassim University, 51452 Buraydah, Saudi Arabia
| | | | - Eltuhami M Abdel-Magied
- Department of Anatomy and Histology, College of Medicine, Qassim University, 51452 Buraydah, Saudi Arabia
| | - Paul Faustin Seke Etet
- Department of Basic Health Sciences, College of Applied Medical Sciences, Qassim University, 51452 Buraydah, Saudi Arabia.
| |
Collapse
|
22
|
NEDD9 Regulates 3D Migratory Activity Independent of the Rac1 Morphology Switch in Glioma and Neuroblastoma. Mol Cancer Res 2013; 12:264-73. [DOI: 10.1158/1541-7786.mcr-13-0513] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
23
|
The overexpression of scaffolding protein NEDD9 promotes migration and invasion in cervical cancer via tyrosine phosphorylated FAK and SRC. PLoS One 2013; 8:e74594. [PMID: 24058594 PMCID: PMC3776827 DOI: 10.1371/journal.pone.0074594] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2013] [Accepted: 08/05/2013] [Indexed: 01/01/2023] Open
Abstract
NEDD9, a focal adhesion scaffolding protein, has been recently proposed to regulate invasion and metastasis in some cancer types, but unknown in cervical cancer. The aim of this study was to determine if NEDD9 was involved in the progression and metastasis of cervical cancer. The experimental results showed NEDD9 protein was overexpressed in cervical cancer compared with normal cervical epithelium tissues. Overexpression of NEDD9 was correlated with histological grading, lymph node metastasis, and FIGO stage of cervical cancer. Silencing NEDD9 resulted in tyrosine dephosphorylation of FAK and SRC oncoproteins, and decreased cell migration and invasion in the cervical carcinoma SiHa and HeLa cells. Overexpression of NEDD9 led to tyrosine phosphorylation of FAK and SRC oncoproteins, and increased cell migration and invasion. Moreover, tyrosine phosphorylation of NEDD9 was significantly decreased via suppressing tyrosine phosphorylation of FAK or SRC, suggesting a positive feedback loop of tyrosine phosphorylation between NEDD9 and FAK or SRC. In addition, our data showed that silencing NEDD9 decreased Vimentin expression and increased E-cadherin expression in cervical cancer cells, and vice versa. E-cadherin was subject to regulation of NEDD9, FAK and SRC, but altered neither tyrosine-phosphorylated nor total NEDD9. Our findings suggest that NEDD9 is overexpressed in cervical cancer tissues and cells, and overexpressed NEDD9 promotes migration and invasion in cervical carcinoma cells, probably via a positive feedback loop of tyrosine phosphorylation between NEDD9 and FAK or SRC.
Collapse
|
24
|
Malmquist SJ, Abramsson A, McGraw HF, Linbo TH, Raible DW. Modulation of dorsal root ganglion development by ErbB signaling and the scaffold protein Sorbs3. Development 2013; 140:3986-96. [PMID: 24004948 DOI: 10.1242/dev.084640] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
The multipotent cells of the vertebrate neural crest (NC) arise at the dorsal aspect of the neural tube, then migrate throughout the developing embryo and differentiate into diverse cell types, including the sensory neurons and glia of the dorsal root ganglia (DRG). As multiple cell types are derived from this lineage, it is ideal for examining mechanisms of fate restriction during development. We have isolated a mutant, ouchless, that specifically fails to develop DRG neurons, although other NC derivatives develop normally. This mutation affects the expression of Sorbs3, a scaffold protein known to interact with proteins involved in focal adhesions and several signaling pathways. ouchless mutants share some phenotypic similarities with mutants in ErbB receptors, EGFR homologs that are implicated in diverse developmental processes and associated with several cancers; and ouchless interacts genetically with an allele of erbb3 in DRG neurogenesis. However, the defect in ouchless DRG neurogenesis is distinct from ErbB loss of function in that it is not associated with a loss of glia. Both ouchless and neurogenin1 heterozygous fish are sensitized to the effects of ErbB chemical inhibitors, which block the development of DRG in a dose-dependent manner. Inhibitors of MEK show similar effects on DRG neurogenesis. We propose a model in which Sorbs3 helps to integrate ErbB signals to promote DRG neurogenesis through the activation of MAPK and upregulation of neurogenin1.
Collapse
Affiliation(s)
- Sarah J Malmquist
- Department of Biological Structure, University of Washington, 1959 NE Pacific Street, Seattle, WA 98195, USA
| | | | | | | | | |
Collapse
|
25
|
Simkin JE, Zhang D, Rollo BN, Newgreen DF. Retinoic acid upregulates ret and induces chain migration and population expansion in vagal neural crest cells to colonise the embryonic gut. PLoS One 2013; 8:e64077. [PMID: 23717535 PMCID: PMC3661488 DOI: 10.1371/journal.pone.0064077] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2013] [Accepted: 04/11/2013] [Indexed: 11/23/2022] Open
Abstract
Vagal neural crest cells (VNCCs) arise in the hindbrain, and at (avian) embryonic day (E) 1.5 commence migration through paraxial tissues to reach the foregut as chains of cells 1–2 days later. They then colonise the rest of the gut in a rostrocaudal wave. The chains of migrating cells later resolve into the ganglia of the enteric nervous system. In organ culture, E4.5 VNCCs resident in the gut (termed enteric or ENCC) which have previously encountered vagal paraxial tissues, rapidly colonised aneural gut tissue in large numbers as chains of cells. Within the same timeframe, E1.5 VNCCs not previously exposed to paraxial tissues provided very few cells that entered the gut mesenchyme, and these never formed chains, despite their ability to migrate in paraxial tissue and in conventional cell culture. Exposing VNCCs in vitro to paraxial tissue normally encountered en route to the foregut conferred enteric migratory ability. VNCC after passage through paraxial tissue developed elements of retinoic acid signalling such as Retinoic Acid Binding Protein 1 expression. The paraxial tissue's ability to promote gut colonisation was reproduced by the addition of retinoic acid, or the synthetic retinoid Am80, to VNCCs (but not to trunk NCCs) in organ culture. The retinoic acid receptor antagonist CD 2665 strongly reduced enteric colonisation by E1.5 VNCC and E4.5 ENCCs, at a concentration suggesting RARα signalling. By FACS analysis, retinoic acid application to vagal neural tube and NCCs in vitro upregulated Ret; a Glial-derived-neurotrophic-factor receptor expressed by ENCCs which is necessary for normal enteric colonisation. This shows that early VNCC, although migratory, are incapable of migrating in appropriate chains in gut mesenchyme, but can be primed for this by retinoic acid. This is the first instance of the characteristic form of NCC migration, chain migration, being attributed to the application of a morphogen.
Collapse
Affiliation(s)
- Johanna E. Simkin
- Embryology Laboratory, Murdoch Children’s Research Institute, Royal Children’s Hospital, Parkville VIC, Australia
| | - Dongcheng Zhang
- Embryology Laboratory, Murdoch Children’s Research Institute, Royal Children’s Hospital, Parkville VIC, Australia
| | - Benjamin N. Rollo
- Embryology Laboratory, Murdoch Children’s Research Institute, Royal Children’s Hospital, Parkville VIC, Australia
| | - Donald F. Newgreen
- Embryology Laboratory, Murdoch Children’s Research Institute, Royal Children’s Hospital, Parkville VIC, Australia
- * E-mail:
| |
Collapse
|
26
|
Wang W, Xu G, Ding CL, Zhao LJ, Zhao P, Ren H, Qi ZT. All-trans retinoic acid protects hepatocellular carcinoma cells against serum-starvation-induced cell death by upregulating collagen 8A2. FEBS J 2013; 280:1308-19. [PMID: 23298258 DOI: 10.1111/febs.12122] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2012] [Revised: 12/11/2012] [Accepted: 01/01/2013] [Indexed: 01/14/2023]
Abstract
As a therapeutic or chemopreventative agent for various cancers, all-trans retinoic acid (atRA) has been reported to inhibit growth, induce apoptosis or cause differentiation. It was found that atRA could protect hepatocellular carcinoma (HCC) cells against cell death induced by serum starvation. Furthermore, it was found that atRA could enhance cell adhesion, but had no effect on the cell cycle and apoptosis. Using an Illumina Human HT-12 v4 expression microarray, 207 upregulated and 173 downregulated genes were identified in HepG2 cells treated with atRA. The most upregulated genes are cytochrome P450 family 26 subfamily A polypeptide 1 (CYP26A1), histidine triad nucleotide binding protein 3 (HINT3), miR-1282 and cytochrome P450 family 26 subfamily B polypeptide 1 (CYP26B1), which showed more than fivefold greater expression. Using Gene Ontology analysis, the greatest significance was found in extracellular-matrix-related molecular functions and the cellular component in upregulated genes. The upregulation of collagen 8A2 (COL8A2) was further confirmed using quantitative RT-PCR and western blotting. Knockdown of COL8A2 blocked enhancement in the early stage of cell adhesion by atRA treatment. Re-expression of COL8A2 in COL8A2-knocked-down HCC cells reversed the effect of small interfering RNA-COL8A2. In addition, COL8A2 could increase HCC cell migration and invasion. Thus, COL8A2 was identified as the key protein involved in the enhancement of cell adhesion of atRA under serum-free conditions. In conclusion, atRA protects HCC cells against serum-starvation-induced cell death by enhancing cell adhesion, and COL8A2 plays an important role in HCC cell migration and invasion.
Collapse
Affiliation(s)
- Wen Wang
- Department of Microbiology, Shanghai Key Laboratory of Medical Biodefense, Second Military Medical University, Shanghai, China
| | | | | | | | | | | | | |
Collapse
|
27
|
A requirement for Nedd9 in luminal progenitor cells prior to mammary tumorigenesis in MMTV-HER2/ErbB2 mice. Oncogene 2013; 33:411-20. [PMID: 23318423 DOI: 10.1038/onc.2012.607] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2012] [Revised: 10/22/2012] [Accepted: 11/12/2012] [Indexed: 02/08/2023]
Abstract
Overexpression of the NEDD9/HEF1/Cas-L scaffolding protein is frequent, and drives invasion and metastasis in breast, head and neck, colorectal, melanoma, lung and other types of cancer. We have examined the consequences of genetic ablation of Nedd9 in the MMTV-HER2/ERBB2/neu mouse mammary tumor model. Unexpectedly, we found that only a limited effect on metastasis in MMTV-neu;Nedd9(-/-) mice compared with MMTV-neu;Nedd9(+/+) mice, but instead a dramatic reduction in tumor incidence (18 versus 80%), and a significantly increased latency until tumor appearance. Orthotopic reinjection and tail-vein injection of cells arising from tumors, coupled with in vivo analysis, indicated tumors arising in MMTV-neu;Nedd9(-/-) mice had undergone mutational selection that overcame the initial requirement for Nedd9. To better understand the defects in early tumor growth, we compared mammary progenitor cell pools from MMTV-neu;Nedd9(-/-) versus MMTV-neu;Nedd9(+/+) mice. The MMTV-neu;Nedd9(-/-) genotype selectively reduced both the number and colony-forming potential of mammary luminal epithelial progenitor cells, while not affecting basal epithelial progenitors. MMTV-neu;Nedd9(-/-) mammospheres had striking defects in morphology and cell polarity. All of these defects were seen predominantly in the context of the HER2/neu oncogene, and were not associated with randomization of the plane of mitotic division, but rather with depressed expression the cell attachment protein FAK, accompanied by increased sensitivity to small molecule inhibitors of FAK and SRC. Surprisingly, in spite of these significant differences, only minimal changes were observed in the gene expression profile of Nedd9(-/-) mice, indicating critical Nedd9-dependent differences in cell growth properties were mediated via post-transcriptional regulation of cell signaling. Coupled with emerging data indicating a role for NEDD9 in progenitor cell populations during the morphogenesis of other tissues, these results indicate a functional requirement for NEDD9 in the growth of mammary cancer progenitor cells.
Collapse
|
28
|
McKeown SJ, Wallace AS, Anderson RB. Expression and function of cell adhesion molecules during neural crest migration. Dev Biol 2012; 373:244-57. [PMID: 23123967 DOI: 10.1016/j.ydbio.2012.10.028] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2012] [Revised: 10/18/2012] [Accepted: 10/25/2012] [Indexed: 01/13/2023]
Abstract
Neural crest cells are highly migratory cells that give rise to many derivatives including peripheral ganglia, craniofacial structures and melanocytes. Neural crest cells migrate along defined pathways to their target sites, interacting with each other and their environment as they migrate. Cell adhesion molecules are critical during this process. In this review we discuss the expression and function of cell adhesion molecules during the process of neural crest migration, in particular cadherins, integrins, members of the immunoglobulin superfamily of cell adhesion molecules, and the proteolytic enzymes that cleave these cell adhesion molecules. The expression and function of these cell adhesion molecules and proteases are compared across neural crest emigrating from different axial levels, and across different species of vertebrates.
Collapse
Affiliation(s)
- Sonja J McKeown
- Department of Anatomy and Neuroscience, University of Melbourne, 3010 VIC, Australia.
| | | | | |
Collapse
|
29
|
Wang Y, Bi L, Wang H, Li Y, Di Q, Xu W, Qian Y. NEDD9 rs760678 polymorphism and the risk of Alzheimer's disease: a meta-analysis. Neurosci Lett 2012; 527:121-5. [PMID: 22963925 DOI: 10.1016/j.neulet.2012.08.044] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2012] [Revised: 08/16/2012] [Accepted: 08/24/2012] [Indexed: 01/22/2023]
Abstract
The NEDD9 rs760678 polymorphism has been extensively investigated for association to Alzheimer's disease (AD), however, results of different studies have been inconsistent. The objective of this study is to assess the relationship of NEDD9 rs760678 polymorphism and AD risk by using meta-analysis. Systematic searches of electronic databases Pubmed and Embase, as well as hand searching of the references of identified articles were performed. Statistical analyses were performed using software Revman 4.2 and STATA 11.0. A total of 4436 cases and 4420 controls in 11 case-control studies were included. The results indicated that the homozygote GG had a 13% decreased risk of AD, when compared with the C allele carriers (CC+CG) (OR=0.87, 95%CI=0.77-0.99, P=0.04 for GG vs. CG+CC). In the subgroup analysis by ethnicity, significant decreased risk was associated with homozygote GG or G allele carriers in Caucasians (OR=0.84, 95%CI=0.74-0.96, P=0.008 for GG vs. CG+CC; OR=0.79, 95%CI=0.69-0.91, P=0.001 for GG vs. CC; OR=0.90, 95%CI=0.84-0.96, P=0.002 for G vs. C), but not in Asians. This meta-analysis suggests that the GG genotype of NEDD9 rs760678 polymorphism would be a protective factor for AD in Caucasians but not in Asians. To further evaluate the effect of gene-gene and gene-environmental interactions between NEDD9 rs760678 polymorphism and the risk of AD, more studies with larger number of subjects are required.
Collapse
Affiliation(s)
- Yucheng Wang
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Nanjing Medical University, No. 300, Guangzhou Road, Nanjing, Jiangsu Province 210029, PR China
| | | | | | | | | | | | | |
Collapse
|
30
|
Xing YY, Yu JT, Yan WJ, Chen W, Zhong XL, Jiang H, Wang P, Tan L. NEDD9 is genetically associated with Alzheimer's disease in a Han Chinese population. Brain Res 2010; 1369:230-4. [PMID: 21059344 DOI: 10.1016/j.brainres.2010.10.113] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2010] [Revised: 10/27/2010] [Accepted: 10/30/2010] [Indexed: 10/18/2022]
Abstract
Neural precursor cell-expressed, developmentally downregulated 9 (NEDD9) has been suspected to be associated with Alzheimer's disease (AD) through participating in the formation of neurite-like membrane extensions and neurite outgrowth to affect the number of neuronal cells/synapses in the brain under stressful conditions. A recent large-scale, multi-tiered association study has identified significant association of a common single nucleotide polymorphism (SNP) rs760678 in the NEDD9 gene with predisposition to late-onset Alzheimer's disease (LOAD) in Caucasians. In order to evaluate the involvement of the NEDD9 polymorphism in the risk of sporadic LOAD, we performed an independent case-control association study to analyze the genotype and allele distributions of the NEDD9 rs760678 polymorphism in a Han Chinese population (383 LOAD cases and 369 healthy controls). There were significant differences in genotype and allele frequencies between LOAD cases and controls (genotype P=0.003, allele P=0.002). After stratification by APOE ε4-carrying status, the C allele of rs760678 was only significantly associated with LOAD in non-APOE ε4 allele carriers (OR=1.43, 95%, CI=1.06-1.94, P=0.024). In addition, a logistic regression analysis also conferred positive association between the SNP rs760678 and LOAD (dominant model: OR=2.10, 95% CI=1.23-3.58, P=0.007; additive model: OR=1.37, 95% CI=1.09-1.74, P=0.008) after adjustment for age, gender, and the APOE ε4 carrier status. The study demonstrated a significant association between the tested SNP and LOAD, indicating that NEDD9 polymorphism has a possible role in changing the genetic susceptibility to LOAD in a Han Chinese population.
Collapse
Affiliation(s)
- Yao-Yao Xing
- Department of Neurology, Qingdao Municipal Hospital, School of Medicine, Qingdao University, No.5 Donghai Middle Road, Qingdao, Shandong Province 266071, PR China
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Tikhmyanova N, Tulin AV, Roegiers F, Golemis EA. Dcas supports cell polarization and cell-cell adhesion complexes in development. PLoS One 2010; 5:e12369. [PMID: 20808771 PMCID: PMC2927436 DOI: 10.1371/journal.pone.0012369] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2010] [Accepted: 07/29/2010] [Indexed: 01/17/2023] Open
Abstract
Mammalian Cas proteins regulate cell migration, division and survival, and are often deregulated in cancer. However, the presence of four paralogous Cas family members in mammals (BCAR1/p130Cas, EFS/Sin1, NEDD9/HEF1/Cas-L, and CASS4/HEPL) has limited their analysis in development. We deleted the single Drosophila Cas gene, Dcas, to probe the developmental function of Dcas. Loss of Dcas had limited effect on embryonal development. However, we found that Dcas is an important modulator of the severity of the developmental phenotypes of mutations affecting integrins (If and mew) and their downstream effectors Fak56D or Src42A. Strikingly, embryonic lethal Fak56D-Dcas double mutant embryos had extensive cell polarity defects, including mislocalization and reduced expression of E-cadherin. Further genetic analysis established that loss of Dcas modified the embryonal lethal phenotypes of embryos with mutations in E-cadherin (Shg) or its signaling partners p120- and beta-catenin (Arm). These results support an important role for Cas proteins in cell-cell adhesion signaling in development.
Collapse
Affiliation(s)
- Nadezhda Tikhmyanova
- Fox Chase Cancer Center, Philadelphia, Pennsylvania, United States of America
- Department of Biochemistry, Drexel University Medical School, Philadelphia, Pennsylvania, United States of America
| | - Alexei V. Tulin
- Fox Chase Cancer Center, Philadelphia, Pennsylvania, United States of America
| | - Fabrice Roegiers
- Fox Chase Cancer Center, Philadelphia, Pennsylvania, United States of America
| | - Erica A. Golemis
- Fox Chase Cancer Center, Philadelphia, Pennsylvania, United States of America
| |
Collapse
|
32
|
Duband JL. Diversity in the molecular and cellular strategies of epithelium-to-mesenchyme transitions: Insights from the neural crest. Cell Adh Migr 2010; 4:458-82. [PMID: 20559020 DOI: 10.4161/cam.4.3.12501] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Although epithelial to mesenchymal transitions (EMT) are often viewed as a unique event, they are characterized by a great diversity of cellular processes resulting in strikingly different outcomes. They may be complete or partial, massive or progressive, and lead to the complete disruption of the epithelium or leave it intact. Although the molecular and cellular mechanisms of EMT are being elucidated owing chiefly from studies on transformed epithelial cell lines cultured in vitro or from cancer cells, the basis of the diversity of EMT processes remains poorly understood. Clues can be collected from EMT occuring during embryonic development and which affect equally tissues of ectodermal, endodermal or mesodermal origins. Here, based on our current knowledge of the diversity of processes underlying EMT of neural crest cells in the vertebrate embryo, we propose that the time course and extent of EMT do not depend merely on the identity of the EMT transcriptional regulators and their cellular effectors but rather on the combination of molecular players recruited and on the possible coordination of EMT with other cellular processes.
Collapse
|
33
|
Tikhmyanova N, Little JL, Golemis EA. CAS proteins in normal and pathological cell growth control. Cell Mol Life Sci 2010; 67:1025-48. [PMID: 19937461 PMCID: PMC2836406 DOI: 10.1007/s00018-009-0213-1] [Citation(s) in RCA: 150] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2009] [Revised: 11/03/2009] [Accepted: 11/09/2009] [Indexed: 12/20/2022]
Abstract
Proteins of the CAS (Crk-associated substrate) family (BCAR1/p130Cas, NEDD9/HEF1/Cas-L, EFS/SIN and CASS4/HEPL) are integral players in normal and pathological cell biology. CAS proteins act as scaffolds to regulate protein complexes controlling migration and chemotaxis, apoptosis, cell cycle, and differentiation, and have more recently been linked to a role in progenitor cell function. Reflecting these complex functions, over-expression of CAS proteins has now been strongly linked to poor prognosis and increased metastasis in cancer, as well as resistance to first-line chemotherapeutics in multiple tumor types including breast and lung cancers, glioblastoma, and melanoma. Further, CAS proteins have also been linked to additional pathological conditions including inflammatory disorders, Alzheimer's and Parkinson's disease, as well as developmental defects. This review will explore the roles of the CAS proteins in normal and pathological states in the context of the many mechanistic insights into CAS protein function that have emerged in the past decade.
Collapse
Affiliation(s)
- Nadezhda Tikhmyanova
- Fox Chase Cancer Center, 333 Cottman Ave., Philadelphia, PA 19111 USA
- Department of Biochemistry, Drexel University Medical School, Philadelphia, PA 19102 USA
| | - Joy L. Little
- Fox Chase Cancer Center, 333 Cottman Ave., Philadelphia, PA 19111 USA
| | - Erica A. Golemis
- Fox Chase Cancer Center, 333 Cottman Ave., Philadelphia, PA 19111 USA
| |
Collapse
|
34
|
Human cytomegalovirus infection causes premature and abnormal differentiation of human neural progenitor cells. J Virol 2010; 84:3528-41. [PMID: 20071566 DOI: 10.1128/jvi.02161-09] [Citation(s) in RCA: 86] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Congenital human cytomegalovirus (HCMV) infection is a leading cause of birth defects, largely manifested as central nervous system (CNS) disorders. The principal site of manifestations in the mouse model is the fetal brain's neural progenitor cell (NPC)-rich subventricular zone. Our previous human NPC studies found these cells to be fully permissive for HCMV and a useful in vitro model system. In continuing work, we observed that under culture conditions favoring maintenance of multipotency, infection caused NPCs to quickly and abnormally differentiate. This phenotypic change required active viral transcription. Whole-genome expression analysis found rapid downregulation of genes that maintain multipotency and establish NPCs' neural identity. Quantitative PCR, Western blot, and immunofluorescence assays confirmed that the mRNA and protein levels of four hallmark NPC proteins (nestin, doublecortin, sex-determining homeobox 2, and glial fibrillary acidic protein) were decreased by HCMV infection. The decreases required active viral replication and were due, at least in part, to proteasomal degradation. Our results suggest that HCMV infection causes in utero CNS defects by inducing both premature and abnormal differentiation of NPCs.
Collapse
|
35
|
Barouki R, Coumoul X. Cell migration and metastasis markers as targets of environmental pollutants and the Aryl hydrocarbon receptor. Cell Adh Migr 2010; 4:72-6. [PMID: 20009531 DOI: 10.4161/cam.4.1.10313] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
During the last few years, several studies have pointed to a surprising link between environmental pollutants cellular signaling and important cell functions such as plasticity, adhesion and migration. This unexpected link could be related to endogenous functions of pollutants receptors that may be disrupted by environmental factors, which is supported by observations in invertebrate species. It could also reveal novel toxic end-points and mechanisms of those pollutants, such as teratogenesis and cancer metastasis that are highly relevant from a public health point of view. In the present short article, we will review our recent observations on the aryl hydrocarbon receptor and its new molecular and cellular targets. We identified HEF1/NEDD9/CAS-L, a multifunctional protein involved in integrin-based signaling as a transcriptional target of the receptor, and showed that its induction was critical for cell plasticity mediated by environmental pollutants. We will put our studies in perspective with other observations made by several groups.
Collapse
|