1
|
Han KA, Ko J. Orchestration of synaptic functions by WAVE regulatory complex-mediated actin reorganization. Exp Mol Med 2023; 55:1065-1075. [PMID: 37258575 PMCID: PMC10318009 DOI: 10.1038/s12276-023-01004-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 03/13/2023] [Accepted: 03/13/2023] [Indexed: 06/02/2023] Open
Abstract
The WAVE regulatory complex (WRC), composed of five components-Cyfip1/Sra1, WAVE/Scar, Abi, Nap1/Nckap1, and Brk1/HSPC300-is essential for proper actin cytoskeletal dynamics and remodeling in eukaryotic cells, likely by matching various patterned signals to Arp2/3-mediated actin nucleation. Accumulating evidence from recent studies has revealed diverse functions of the WRC in neurons, demonstrating its crucial role in dictating the assembly of molecular complexes for the patterning of various trans-synaptic signals. In this review, we discuss recent exciting findings on the physiological role of the WRC in regulating synaptic properties and highlight the involvement of WRC dysfunction in various brain disorders.
Collapse
Affiliation(s)
- Kyung Ah Han
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), 333 Techno Jungangdae-Ro, Hyeonpoong-Eup, Dalseong-Gun, Daegu, 42988, Korea
- Center for Synapse Diversity and Specificity, DGIST, Daegu, 42988, Korea
| | - Jaewon Ko
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), 333 Techno Jungangdae-Ro, Hyeonpoong-Eup, Dalseong-Gun, Daegu, 42988, Korea.
- Center for Synapse Diversity and Specificity, DGIST, Daegu, 42988, Korea.
| |
Collapse
|
2
|
Wang S, Zhang H, Liu H, Guo X, Ma R, Zhu W, Gao P. ELK1-induced up-regulation of KIF26B promotes cell cycle progression in breast cancer. Med Oncol 2021; 39:15. [PMID: 34817735 DOI: 10.1007/s12032-021-01607-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 11/01/2021] [Indexed: 10/19/2022]
Abstract
KIF26B is a member of the kinesin superfamily that is up-regulated in various tumors, including breast cancer (BC), which can promote tumor progression. This study aimed to investigate the potential function of KIF26B in BC, and the underlying mechanisms, focusing mainly on cell proliferation. KIF26B expression was examined in BC tissue samples obtained from 99 patients. Then, we performed MTS, EdU and flow cytometry assays to detect cell proliferation, and western blotting to measure the expression of cell cycle-related proteins in MDA-MB-231 and MDA-MB-468 cells following KIF26B knockdown. Promoter analysis was used to study the upstream regulatory mechanism of KIF26B. KIF26B was upregulated in BC tissues. High expression of KIF26B was associated with clinicopathological parameters, such as positive lymph node metastasis, higher tumor grade, and higher proliferative index in BC. Furthermore, knockdown of KIF26B expression inhibited MDA-MB-231 and MDA-MB-468 cell proliferation, arresting cells in the G1 phase of the cell cycle in vitro. Similarly, KIF26B silencing decreased the expression levels of Wnt, β-catenin, and cell cycle-related proteins such as c-Myc, cyclin D1, and cyclin-dependent kinase 4, while increasing the expression of p27. Moreover, ELK1 could bind to the core promoter region of KIF26B and activate its transcription. KIF26B acts as an oncogene in BC by regulating multiple proteins involved in the cell cycle. ELK1 activates KIF26B transcription.
Collapse
Affiliation(s)
- SuXia Wang
- Key Laboratory for Experimental Teratology of Ministry of Education, Department of Pathology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan Wen Hua Xi Road 44, Jinan, 250012, Shandong, China.,Department of Pathology, Yantai Yuhuangding Hospital of Qingdao University, Yantai, 264000, Shandong, China.,Department of Pathology, Qilu Hospital, Shandong University Jinan, Shandong, 250012, China
| | - Hui Zhang
- Key Laboratory for Experimental Teratology of Ministry of Education, Department of Pathology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan Wen Hua Xi Road 44, Jinan, 250012, Shandong, China.,Department of Pathology, Qilu Hospital, Shandong University Jinan, Shandong, 250012, China
| | - HaiTing Liu
- Key Laboratory for Experimental Teratology of Ministry of Education, Department of Pathology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan Wen Hua Xi Road 44, Jinan, 250012, Shandong, China.,Department of Pathology, Qilu Hospital, Shandong University Jinan, Shandong, 250012, China
| | - XiangYu Guo
- Key Laboratory for Experimental Teratology of Ministry of Education, Department of Pathology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan Wen Hua Xi Road 44, Jinan, 250012, Shandong, China
| | - RanRan Ma
- Key Laboratory for Experimental Teratology of Ministry of Education, Department of Pathology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan Wen Hua Xi Road 44, Jinan, 250012, Shandong, China.,Department of Pathology, Qilu Hospital, Shandong University Jinan, Shandong, 250012, China
| | - WenJie Zhu
- Key Laboratory for Experimental Teratology of Ministry of Education, Department of Pathology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan Wen Hua Xi Road 44, Jinan, 250012, Shandong, China.
| | - P Gao
- Key Laboratory for Experimental Teratology of Ministry of Education, Department of Pathology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan Wen Hua Xi Road 44, Jinan, 250012, Shandong, China. .,Department of Pathology, Qilu Hospital, Shandong University Jinan, Shandong, 250012, China.
| |
Collapse
|
3
|
SARGAZI S, HEIDARI NIA M, MIRINEJAD S, MOUDI M, JAFARI SHAHROUDI M, SARAVANI R, VALIAN-BOROJENI S. Association of a Novel KIF26B Gene Polymorphism with Susceptibility to Schizophrenia and Breast Cancer: A Case-Control Study. IRANIAN JOURNAL OF PUBLIC HEALTH 2021; 50:397-406. [PMID: 33748005 PMCID: PMC7956084 DOI: 10.18502/ijph.v50i2.5359] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 07/21/2020] [Indexed: 12/30/2022]
Abstract
BACKGROUND KIF26B gene is found to play essential roles in regulating different aspects of cell proliferation and development of the nervous system. We aimed to determine if rs12407427 T/C polymorphism could affect susceptibility to schizophrenia (SZN) and breast cancer (BC), the two genetically correlated diseases. METHODS The current case-control study was performed from Aug 2018 to Dec 2018. Briefly, 159 female pathologically confirmed BC cases referring to Alzahra Hospital, Isfahan, Iran, and 102 psychologically confirmed SZN patients (60 males and 42 females) admitted to Baharan Hospital, Zahedan, Iran, were enrolled. Using the salting-out method, genomic DNA was extracted, and variants were genotyped using allele-specific amplification refractory mutation system polymerase chain reaction (ARMS-PCR) method. RESULTS The results revealed a significant association between the KIF26B rs12407427 codominant CT (P=0.001), CC (P=0.0001), dominant CT+CC, and recessive CC (P=0.001) genotypes with the risk of developing SZN. Significant correlations were also found regarding rs12407427 and BC susceptibility in different inheritance models, including over-dominant CT (P=0.026), dominant CT+CC (P=0.001), recessive CC (P=0.009), and codominant CT and CC (P=0.001) genotypes. The over-presence of the C allele was also correlated with an increased risk for SZN (P=0.0001) and BC (P=0.0001). Finally, computational analysis predicted that T/C variation in this polymorphism could change the binding sites in proteins involved in splicing. CONCLUSION rs12407427 T/C as a de novo KIF26B variant might be a novel genetic biomarker for SZN and/or BC susceptibility in a sample of the Iranian population.
Collapse
Affiliation(s)
- Saman SARGAZI
- Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Milad HEIDARI NIA
- Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran
- Department of Biology, Faculty of Science, Isfahan University, Isfahan, Iran
| | - Shekoufeh MIRINEJAD
- Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Mahdiyeh MOUDI
- Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Mahdiyeh JAFARI SHAHROUDI
- Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Ramin SARAVANI
- Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran
- Department of Clinical Biochemistry, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran
| | | |
Collapse
|
4
|
Mawaribuchi S, Aiki Y, Ikeda N, Ito Y. mRNA and miRNA expression profiles in an ectoderm-biased substate of human pluripotent stem cells. Sci Rep 2019; 9:11910. [PMID: 31417139 PMCID: PMC6695399 DOI: 10.1038/s41598-019-48447-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Accepted: 08/05/2019] [Indexed: 12/27/2022] Open
Abstract
The potential applications of human pluripotent stem cells, embryonic stem (ES) cells, and induced pluripotent stem (iPS) cells in cell therapy and regenerative medicine have been widely studied. The precise definition of pluripotent stem cell status during culture using biomarkers is essential for basic research and regenerative medicine. Culture conditions, including extracellular matrices, influence the balance between self-renewal and differentiation. Accordingly, to explore biomarkers for defining and monitoring the pluripotent substates during culture, we established different substates in H9 human ES cells by changing the extracellular matrix from vitronectin to Matrigel. The substate was characterised by low and high expression of the pluripotency marker R-10G epitope and the mesenchymal marker vimentin, respectively. Immunohistochemistry, induction of the three germ layers, and exhaustive expression analysis showed that the substate was ectoderm-biased, tended to differentiate into nerves, but retained the potential to differentiate into the three germ layers. Further integrated analyses of mRNA and miRNA microarrays and qPCR analysis showed that nine genes (COL9A2, DGKI, GBX2, KIF26B, MARCH1, PLXNA4, SLC24A4, TLR4, and ZHX3) were upregulated in the ectoderm-biased cells as ectoderm-biased biomarker candidates in pluripotent stem cells. Our findings provide important insights into ectoderm-biased substates of human pluripotent stem cells in the fields of basic research and regenerative medicine.
Collapse
Affiliation(s)
- Shuuji Mawaribuchi
- Biotechnology Research Institute for Drug Discovery, National Institute of Advanced Industrial Science and Technology (AIST), Central 5, 1-1-1 Higashi, Tsukuba, Ibaraki, 305-8565, Japan
| | - Yasuhiko Aiki
- Biotechnology Research Institute for Drug Discovery, National Institute of Advanced Industrial Science and Technology (AIST), Central 5, 1-1-1 Higashi, Tsukuba, Ibaraki, 305-8565, Japan
| | - Nozomi Ikeda
- Biotechnology Research Institute for Drug Discovery, National Institute of Advanced Industrial Science and Technology (AIST), Central 5, 1-1-1 Higashi, Tsukuba, Ibaraki, 305-8565, Japan
| | - Yuzuru Ito
- Biotechnology Research Institute for Drug Discovery, National Institute of Advanced Industrial Science and Technology (AIST), Central 5, 1-1-1 Higashi, Tsukuba, Ibaraki, 305-8565, Japan.
| |
Collapse
|
5
|
Li H, Shen S, Chen X, Ren Z, Li Z, Yu Z. miR-450b-5p loss mediated KIF26B activation promoted hepatocellular carcinoma progression by activating PI3K/AKT pathway. Cancer Cell Int 2019; 19:205. [PMID: 31388332 PMCID: PMC6670205 DOI: 10.1186/s12935-019-0923-x] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Accepted: 07/22/2019] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Kinesin family member 26B (KIF26B) is unveiled acted as important role in many solid tumors, however, the function of KIF26B in hepatocellular carcinoma (HCC) is unclear. METHODS The expression of KIF26B in HCC tissues and cell lines were measured with immunochemistry, real-time PCR and western blotting. The correlation between KIF26B expression and clinicopathological characteristics were analyzed by SPSS19.0. Functional experiments of KIF26B was conducted by CCK-8, transwell, EDU, colony formation in vitro and tumorigenesis in vivo. The gene set enrichment analysis was used to search the downstream pathway, luciferase reporter experiment was used to find the upstream regulatory factor of KIF26B. RESULTS In this study, we found that KIF26B was overexpressed both in HCC tissues and cell lines. High expression of KIF26B was associated with poor overall survival (OS), late TNM stage and poor differentiation. Loss of function experiments showed that suppression of KIF26B could inhibit cell viability, proliferation rate and invasion ability of HCC cells. KEGG and GO analysis showed that expression of KIF26B was highly relevant with PI3K/AKT signal pathway, and suppression of KIF26B could decrease the expression of m-TOR, p-PI3K and p-AKT. Further study demonstrated that expression of KIF26B was negative correlated with miR-450b-5p level in HCC tissues, and miR-450b-5p could inhibit cell viability, proliferation rate and invasion ability of HCC cells via targeted inhibiting KIF26B. CONCLUSION Our study demonstrated that miR-450-5p/KIF26B/AKT axis is critical for progression of HCC, and might provide novel prognostic biomarker and therapeutic target for HCC.
Collapse
Affiliation(s)
- Hua Li
- Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, 1# Jianshe East Road, Zhengzhou, 450052 China
- Gene Hospital of Henan Province, Precision Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052 China
| | - Shen Shen
- Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, 1# Jianshe East Road, Zhengzhou, 450052 China
| | - Xiaolong Chen
- Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, 1# Jianshe East Road, Zhengzhou, 450052 China
| | - Zhigang Ren
- Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, 1# Jianshe East Road, Zhengzhou, 450052 China
- Gene Hospital of Henan Province, Precision Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052 China
| | - Zhiqin Li
- Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, 1# Jianshe East Road, Zhengzhou, 450052 China
| | - Zujiang Yu
- Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, 1# Jianshe East Road, Zhengzhou, 450052 China
- Gene Hospital of Henan Province, Precision Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052 China
| |
Collapse
|
6
|
Wojcik MH, Okada K, Prabhu SP, Nowakowski DW, Ramsey K, Balak C, Rangasamy S, Brownstein CA, Schmitz-Abe K, Cohen JS, Fatemi A, Shi J, Grant EP, Narayanan V, Ho HYH, Agrawal PB. De novo variant in KIF26B is associated with pontocerebellar hypoplasia with infantile spinal muscular atrophy. Am J Med Genet A 2018; 176:2623-2629. [PMID: 30151950 DOI: 10.1002/ajmg.a.40493] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Revised: 06/13/2018] [Accepted: 07/02/2018] [Indexed: 12/14/2022]
Abstract
KIF26B is a member of the kinesin superfamily with evolutionarily conserved functions in controlling aspects of embryogenesis, including the development of the nervous system, though its function is incompletely understood. We describe an infant with progressive microcephaly, pontocerebellar hypoplasia, and arthrogryposis secondary to the involvement of anterior horn cells and ventral (motor) nerves. We performed whole exome sequencing on the trio and identified a de novo KIF26B missense variant, p.Gly546Ser, in the proband. This variant alters a highly conserved amino acid residue that is part of the phosphate-binding loop motif and motor-like domain and is deemed pathogenic by several in silico methods. Functional analysis of the variant protein in cultured cells revealed a reduction in the KIF26B protein's ability to promote cell adhesion, a defect that potentially contributes to its pathogenicity. Overall, KIF26B may play a critical role in the brain development and, when mutated, cause pontocerebellar hypoplasia with arthrogryposis.
Collapse
Affiliation(s)
- Monica H Wojcik
- Division of Newborn Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts.,Division of Genetics and Genomics, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts.,The Manton Center for Orphan Disease Research, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Kyoko Okada
- Department of Cell Biology and Human Anatomy, University of California Davis School of Medicine, Davis, California
| | - Sanjay P Prabhu
- Neuroradiology Division, Department of Radiology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | | | - Keri Ramsey
- Center for Rare Childhood Disorders, Translational Genomic Research Institute, Phoenix, Arizona
| | - Chris Balak
- Center for Rare Childhood Disorders, Translational Genomic Research Institute, Phoenix, Arizona
| | - Sampath Rangasamy
- Center for Rare Childhood Disorders, Translational Genomic Research Institute, Phoenix, Arizona
| | - Catherine A Brownstein
- Division of Genetics and Genomics, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts.,The Manton Center for Orphan Disease Research, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Klaus Schmitz-Abe
- Division of Newborn Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts.,The Manton Center for Orphan Disease Research, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Julie S Cohen
- Division of Neurogenetics, Hugo W. Moser Research Institute, Kennedy Krieger Institute, Baltimore, Maryland
| | - Ali Fatemi
- Division of Neurogenetics, Hugo W. Moser Research Institute, Kennedy Krieger Institute, Baltimore, Maryland.,Departments of Neurology and Pediatrics, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Jiahai Shi
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, Hong Kong SAR
| | - Ellen P Grant
- Division of Newborn Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts.,Neuroradiology Division, Department of Radiology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Vinodh Narayanan
- Center for Rare Childhood Disorders, Translational Genomic Research Institute, Phoenix, Arizona
| | - Hsin-Yi Henry Ho
- Department of Cell Biology and Human Anatomy, University of California Davis School of Medicine, Davis, California
| | - Pankaj B Agrawal
- Division of Newborn Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts.,Division of Genetics and Genomics, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts.,The Manton Center for Orphan Disease Research, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
7
|
Gu S, Liang H, Qi D, Mao L, Mao G, Qian L, Zhang S. Knockdown of KIF26B inhibits breast cancer cell proliferation, migration, and invasion. Onco Targets Ther 2018; 11:3195-3203. [PMID: 29881293 PMCID: PMC5985780 DOI: 10.2147/ott.s163346] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Background Kinesin family member 26B (KIF26B) plays a key role in the development and progression of many human cancers. However, the role and underlying mechanisms of KIF26B in breast cancer cells remain unknown. Materials and methods In this study, we inhibited the expression of KIF26B in MDA-MB-231 and MCF-7 cells using lentivirus-delivered shRNA. Results Lentivirus-mediated KIF26B knockdown significantly suppressed cell proliferation, colony formation, migration, and invasion. Furthermore, cell cycle analyses revealed that the percentage of cells in the G0/G1 phase was significantly increased in KIF26B knockdown cells. Moreover, the knockdown of KIF26B significantly promoted cell apoptosis via the upregulation of cleaved caspase-3 and Bax. Conclusion Our data indicate that KIF26B plays a pivotal role in tumor growth and metastasis in breast cancer cells and may be a potential therapeutic target for treating breast cancer.
Collapse
Affiliation(s)
- Shudong Gu
- Department of Oncology, Affiliated Hospital of Nantong University, Nantong 226001, China
| | - Haibin Liang
- Department of General Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Donghui Qi
- Medical College of Nantong University, Nantong 226001, China
| | - Liyan Mao
- Department of Endoscopic Diagnosis and Treatment of Digestive Diseases, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Guoxin Mao
- Department of Oncology, Affiliated Hospital of Nantong University, Nantong 226001, China
| | - Li Qian
- Department of Oncology, Affiliated Hospital of Nantong University, Nantong 226001, China
| | - Shu Zhang
- Department of Pathology, Affiliated Hospital of Nantong University, Nantong 226001, China
| |
Collapse
|
8
|
Guillabert-Gourgues A, Jaspard-Vinassa B, Bats ML, Sewduth RN, Franzl N, Peghaire C, Jeanningros S, Moreau C, Roux E, Larrieu-Lahargue F, Dufourcq P, Couffinhal T, Duplàa C. Kif26b controls endothelial cell polarity through the Dishevelled/Daam1-dependent planar cell polarity-signaling pathway. Mol Biol Cell 2016; 27:941-53. [PMID: 26792835 PMCID: PMC4791138 DOI: 10.1091/mbc.e14-08-1332] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2014] [Accepted: 01/13/2016] [Indexed: 11/11/2022] Open
Abstract
Angiogenesis involves the coordinated growth and migration of endothelial cells (ECs) toward a proangiogenic signal. The Wnt planar cell polarity (PCP) pathway, through the recruitment of Dishevelled (Dvl) and Dvl-associated activator of morphogenesis (Daam1), has been proposed to regulate cell actin cytoskeleton and microtubule (MT) reorganization for oriented cell migration. Here we report that Kif26b--a kinesin--and Daam1 cooperatively regulate initiation of EC sprouting and directional migration via MT reorganization. First, we find that Kif26b is recruited within the Dvl3/Daam1 complex. Using a three-dimensional in vitro angiogenesis assay, we show that Kif26b and Daam1 depletion impairs tip cell polarization and destabilizes extended vascular processes. Kif26b depletion specifically alters EC directional migration and mislocalized MT organizing center (MTOC)/Golgi and myosin IIB cell rear enrichment. Therefore the cell fails to establish a proper front-rear polarity. Of interest, Kif26b ectopic expression rescues the siDaam1 polarization defect phenotype. Finally, we show that Kif26b functions in MT stabilization, which is indispensable for asymmetrical cell structure reorganization. These data demonstrate that Kif26b, together with Dvl3/Daam1, initiates cell polarity through the control of PCP signaling pathway-dependent activation.
Collapse
Affiliation(s)
| | - Beatrice Jaspard-Vinassa
- Adaptation Cardiovasculaire à l'Ischémie, INSERM, U1034, F-33600 Pessac, France Adaptation Cardiovasculaire à l'Ischémie, U1034, Université de Bordeaux, F-33600 Pessac, France
| | - Marie-Lise Bats
- Adaptation Cardiovasculaire à l'Ischémie, INSERM, U1034, F-33600 Pessac, France Adaptation Cardiovasculaire à l'Ischémie, U1034, Université de Bordeaux, F-33600 Pessac, France
| | - Raj N Sewduth
- Adaptation Cardiovasculaire à l'Ischémie, INSERM, U1034, F-33600 Pessac, France
| | - Nathalie Franzl
- Adaptation Cardiovasculaire à l'Ischémie, INSERM, U1034, F-33600 Pessac, France
| | - Claire Peghaire
- Adaptation Cardiovasculaire à l'Ischémie, INSERM, U1034, F-33600 Pessac, France
| | - Sylvie Jeanningros
- Adaptation Cardiovasculaire à l'Ischémie, INSERM, U1034, F-33600 Pessac, France
| | - Catherine Moreau
- Adaptation Cardiovasculaire à l'Ischémie, INSERM, U1034, F-33600 Pessac, France
| | - Etienne Roux
- Adaptation Cardiovasculaire à l'Ischémie, INSERM, U1034, F-33600 Pessac, France Adaptation Cardiovasculaire à l'Ischémie, U1034, Université de Bordeaux, F-33600 Pessac, France
| | | | - Pascale Dufourcq
- Adaptation Cardiovasculaire à l'Ischémie, INSERM, U1034, F-33600 Pessac, France Adaptation Cardiovasculaire à l'Ischémie, U1034, Université de Bordeaux, F-33600 Pessac, France
| | - Thierry Couffinhal
- Adaptation Cardiovasculaire à l'Ischémie, INSERM, U1034, F-33600 Pessac, France Adaptation Cardiovasculaire à l'Ischémie, U1034, Université de Bordeaux, F-33600 Pessac, France Service des Maladies Cardiaques et Vasculaires, Centre Hospitalier Universitaire de Bordeaux, F-33000 Bordeaux, France
| | - Cecile Duplàa
- Adaptation Cardiovasculaire à l'Ischémie, INSERM, U1034, F-33600 Pessac, France Adaptation Cardiovasculaire à l'Ischémie, U1034, Université de Bordeaux, F-33600 Pessac, France
| |
Collapse
|
9
|
Maccani JZJ, Koestler DC, Lester B, Houseman EA, Armstrong DA, Kelsey KT, Marsit CJ. Placental DNA Methylation Related to Both Infant Toenail Mercury and Adverse Neurobehavioral Outcomes. ENVIRONMENTAL HEALTH PERSPECTIVES 2015; 123:723-9. [PMID: 25748564 PMCID: PMC4492267 DOI: 10.1289/ehp.1408561] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/15/2014] [Accepted: 03/04/2015] [Indexed: 05/19/2023]
Abstract
BACKGROUND Prenatal mercury (Hg) exposure is associated with adverse child neurobehavioral outcomes. Because Hg can interfere with placental functioning and cross the placenta to target the fetal brain, prenatal Hg exposure can inhibit fetal growth and development directly and indirectly. OBJECTIVES We examined potential associations between prenatal Hg exposure assessed through infant toenail Hg, placental DNA methylation changes, and newborn neurobehavioral outcomes. METHODS The methylation status of > 485,000 CpG loci was interrogated in 192 placental samples using Illumina's Infinium HumanMethylation450 BeadArray. Hg concentrations were analyzed in toenail clippings from a subset of 41 infants; neurobehavior was assessed using the NICU Network Neurobehavioral Scales (NNNS) in an independent subset of 151 infants. RESULTS We identified 339 loci with an average methylation difference > 0.125 between any two toenail Hg tertiles. Variation among these loci was subsequently found to be associated with a high-risk neurodevelopmental profile (omnibus p-value = 0.007) characterized by the NNNS. Ten loci had p < 0.01 for the association between methylation and the high-risk NNNS profile. Six of 10 loci reside in the EMID2 gene and were hypomethylated in the 16 high-risk profile infants' placentas. Methylation at these loci was moderately correlated (correlation coefficients range, -0.33 to -0.45) with EMID2 expression. CONCLUSIONS EMID2 hypomethylation may represent a novel mechanism linking in utero Hg exposure and adverse infant neurobehavioral outcomes.
Collapse
Affiliation(s)
- Jennifer Z J Maccani
- Penn State Tobacco Center of Regulatory Science, Department of Public Health Sciences, College of Medicine, Penn State University, Hershey, Pennsylvania, USA
| | | | | | | | | | | | | |
Collapse
|
10
|
Elevated kinesin family member 26B is a prognostic biomarker and a potential therapeutic target for colorectal cancer. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2015; 34:13. [PMID: 25652119 PMCID: PMC4322797 DOI: 10.1186/s13046-015-0129-6] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Accepted: 01/26/2015] [Indexed: 12/30/2022]
Abstract
Background Kinesins play a key role in the development and progression of many human cancers. The present study investigated the expression and clinical significance of kinesin family member 26B (KIF26B) in colorectal cancer (CRC). Methods Using quantitative real-time PCR and Western blot analyses as well as immunohistochemical staining of a tissue microarray we examined KIF26B mRNA and protein levels in CRC tumor tissues and paired adjacent normal mucosa. Moreover, the effect of KIF26B knockdown on CRC cell proliferation was investigated using Cell Counting Kit-8 assays. Results Expression of KIF26B was found to be elevated in CRC. Suppression of KIF26B inhibited CRC cell proliferation. Furthermore, upregulated expression of KIF26B was significantly correlated with tumor size (P = 0.020), American Joint Committee on Cancer (AJCC) stage (P = 0.018), T stage (P = 0.026), N stage (P = 0.013), and differentiation histology (P = 0.047). KIF26B was also shown to be an independent prognostic indicator of overall survival for CRC patients (HR 5.621; 95% CI 2.302–13.730; P < 0.001). Conclusion Our data indicate that KIF26B plays an important role in colorectal carcinogenesis and functions as a novel prognostic indicator and a potential therapeutic target for CRC.
Collapse
|