1
|
Nishida M, Sato A, Shimizu A, Rahman N, Wada A, Kageyama S, Ogita H. EphA-Mediated Regulation of Stomatin Expression in Prostate Cancer Cells. Cancer Med 2024; 13:e70276. [PMID: 39377541 PMCID: PMC11459579 DOI: 10.1002/cam4.70276] [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: 06/14/2024] [Revised: 09/13/2024] [Accepted: 09/20/2024] [Indexed: 10/09/2024] Open
Abstract
BACKGROUND AND AIMS Tumor growth and progression are affected by interactions between tumor cells and stromal cells within the tumor microenvironment. We previously showed that the expression of an integral membrane protein, called stomatin, was increased in cancer cells following their association with stromal cells. Additionally, stomatin impaired the Akt signaling pathway to suppress tumor growth. However, it remains unclear how stomatin expression is regulated. To explore this, we examined the cell surface molecules that can transduce the intercellular communication signals between cancer cells and stromal cells. RESULTS Among these molecules, EphA3 and EphA7 receptors and their ligand ephrin-A5 were found to be expressed in prostate cancer cells, but not in prostate stromal cells. Cell-to-cell contact of prostate cancer cells through the EphA-ephrin-A interaction suppressed stomatin expression, while knockdown of EphA3/7 or ephrin-A5 increased stomatin expression. This increase contributed to an inhibition of prostate cancer cell proliferation. Intracellularly, the binding of ephrin-A to EphA attenuated extracellular signaling-regulated kinase (ERK) activation that promoted stomatin expression. Furthermore, ELK1 and ELK4, which are Ets family transcription factors phosphorylated by ERK, were involved in the induction of stomatin expression. We also found that higher Gleason score prostate cancer tissue samples had increased activation of EphA, while the stomatin expression and activated ERK and ELK levels were all low. In the mouse xenograft tumor samples generated by implantation of prostate cancer cells, EphA3 phosphorylation was attenuated and the ERK-ELK signaling and stomatin expression were enhanced in the area where stromal cells infiltrated the tumor. CONCLUSION The EphA-mediated signaling suppresses the ERK-ELK pathway, leading to the reduction of stomatin expression that affects prostate cancer malignancy.
Collapse
Affiliation(s)
- Masanari Nishida
- Division of Molecular Medical Biochemistry, Department of Biochemistry and Molecular BiologyShiga University of Medical ScienceOtsuJapan
- Department of UrologyShiga University of Medical ScienceOtsuJapan
| | - Akira Sato
- Division of Molecular Medical Biochemistry, Department of Biochemistry and Molecular BiologyShiga University of Medical ScienceOtsuJapan
| | - Akio Shimizu
- Division of Molecular Medical Biochemistry, Department of Biochemistry and Molecular BiologyShiga University of Medical ScienceOtsuJapan
| | - Nor Idayu A. Rahman
- Division of Molecular Medical Biochemistry, Department of Biochemistry and Molecular BiologyShiga University of Medical ScienceOtsuJapan
| | - Akinori Wada
- Department of UrologyShiga University of Medical ScienceOtsuJapan
| | - Susumu Kageyama
- Department of UrologyShiga University of Medical ScienceOtsuJapan
| | - Hisakazu Ogita
- Division of Molecular Medical Biochemistry, Department of Biochemistry and Molecular BiologyShiga University of Medical ScienceOtsuJapan
| |
Collapse
|
2
|
Ziranu P, Pretta A, Aimola V, Cau F, Mariani S, D’Agata AP, Codipietro C, Rizzo D, Dell’Utri V, Sanna G, Moledda G, Cadoni A, Lai E, Puzzoni M, Pusceddu V, Castagnola M, Scartozzi M, Faa G. CD44: A New Prognostic Marker in Colorectal Cancer? Cancers (Basel) 2024; 16:1569. [PMID: 38672650 PMCID: PMC11048923 DOI: 10.3390/cancers16081569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 03/19/2024] [Accepted: 04/13/2024] [Indexed: 04/28/2024] Open
Abstract
Cluster of differentiation 44 (CD44) is a non-kinase cell surface glycoprotein. It is overexpressed in several cell types, including cancer stem cells (CSCs). Cells overexpressing CD44 exhibit several CSC traits, such as self-renewal, epithelial-mesenchymal transition (EMT) capability, and resistance to chemo- and radiotherapy. The role of CD44 in maintaining stemness and the CSC function in tumor progression is accomplished by binding to its main ligand, hyaluronan (HA). The HA-CD44 complex activates several signaling pathways that lead to cell proliferation, adhesion, migration, and invasion. The CD44 gene regularly undergoes alternative splicing, resulting in the standard (CD44s) and variant (CD44v) isoforms. The different functional roles of CD44s and specific CD44v isoforms still need to be fully understood. The clinicopathological impact of CD44 and its isoforms in promoting tumorigenesis suggests that CD44 could be a molecular target for cancer therapy. Furthermore, the recent association observed between CD44 and KRAS-dependent carcinomas and the potential correlations between CD44 and tumor mutational burden (TMB) and microsatellite instability (MSI) open new research scenarios for developing new strategies in cancer treatment. This review summarises current research regarding the different CD44 isoform structures, their roles, and functions in supporting tumorigenesis and discusses its therapeutic implications.
Collapse
Affiliation(s)
- Pina Ziranu
- Medical Oncology Unit, University Hospital and University of Cagliari, SS 554 km 4500 Bivio per Sestu, Monserrato, 09042 Cagliari, Italy; (A.P.); (S.M.); (A.P.D.); (C.C.); (D.R.); (V.D.); (G.S.); (G.M.); (A.C.); (E.L.); (M.P.); (V.P.); (M.S.)
| | - Andrea Pretta
- Medical Oncology Unit, University Hospital and University of Cagliari, SS 554 km 4500 Bivio per Sestu, Monserrato, 09042 Cagliari, Italy; (A.P.); (S.M.); (A.P.D.); (C.C.); (D.R.); (V.D.); (G.S.); (G.M.); (A.C.); (E.L.); (M.P.); (V.P.); (M.S.)
| | - Valentina Aimola
- Division of Pathology, Department of Medical Sciences and Public Health, AOU Cagliari, University of Cagliari, 09124 Cagliari, Italy; (V.A.); (F.C.)
| | - Flaviana Cau
- Division of Pathology, Department of Medical Sciences and Public Health, AOU Cagliari, University of Cagliari, 09124 Cagliari, Italy; (V.A.); (F.C.)
| | - Stefano Mariani
- Medical Oncology Unit, University Hospital and University of Cagliari, SS 554 km 4500 Bivio per Sestu, Monserrato, 09042 Cagliari, Italy; (A.P.); (S.M.); (A.P.D.); (C.C.); (D.R.); (V.D.); (G.S.); (G.M.); (A.C.); (E.L.); (M.P.); (V.P.); (M.S.)
| | - Alessandra Pia D’Agata
- Medical Oncology Unit, University Hospital and University of Cagliari, SS 554 km 4500 Bivio per Sestu, Monserrato, 09042 Cagliari, Italy; (A.P.); (S.M.); (A.P.D.); (C.C.); (D.R.); (V.D.); (G.S.); (G.M.); (A.C.); (E.L.); (M.P.); (V.P.); (M.S.)
| | - Claudia Codipietro
- Medical Oncology Unit, University Hospital and University of Cagliari, SS 554 km 4500 Bivio per Sestu, Monserrato, 09042 Cagliari, Italy; (A.P.); (S.M.); (A.P.D.); (C.C.); (D.R.); (V.D.); (G.S.); (G.M.); (A.C.); (E.L.); (M.P.); (V.P.); (M.S.)
| | - Daiana Rizzo
- Medical Oncology Unit, University Hospital and University of Cagliari, SS 554 km 4500 Bivio per Sestu, Monserrato, 09042 Cagliari, Italy; (A.P.); (S.M.); (A.P.D.); (C.C.); (D.R.); (V.D.); (G.S.); (G.M.); (A.C.); (E.L.); (M.P.); (V.P.); (M.S.)
| | - Veronica Dell’Utri
- Medical Oncology Unit, University Hospital and University of Cagliari, SS 554 km 4500 Bivio per Sestu, Monserrato, 09042 Cagliari, Italy; (A.P.); (S.M.); (A.P.D.); (C.C.); (D.R.); (V.D.); (G.S.); (G.M.); (A.C.); (E.L.); (M.P.); (V.P.); (M.S.)
| | - Giorgia Sanna
- Medical Oncology Unit, University Hospital and University of Cagliari, SS 554 km 4500 Bivio per Sestu, Monserrato, 09042 Cagliari, Italy; (A.P.); (S.M.); (A.P.D.); (C.C.); (D.R.); (V.D.); (G.S.); (G.M.); (A.C.); (E.L.); (M.P.); (V.P.); (M.S.)
| | - Giusy Moledda
- Medical Oncology Unit, University Hospital and University of Cagliari, SS 554 km 4500 Bivio per Sestu, Monserrato, 09042 Cagliari, Italy; (A.P.); (S.M.); (A.P.D.); (C.C.); (D.R.); (V.D.); (G.S.); (G.M.); (A.C.); (E.L.); (M.P.); (V.P.); (M.S.)
| | - Andrea Cadoni
- Medical Oncology Unit, University Hospital and University of Cagliari, SS 554 km 4500 Bivio per Sestu, Monserrato, 09042 Cagliari, Italy; (A.P.); (S.M.); (A.P.D.); (C.C.); (D.R.); (V.D.); (G.S.); (G.M.); (A.C.); (E.L.); (M.P.); (V.P.); (M.S.)
| | - Eleonora Lai
- Medical Oncology Unit, University Hospital and University of Cagliari, SS 554 km 4500 Bivio per Sestu, Monserrato, 09042 Cagliari, Italy; (A.P.); (S.M.); (A.P.D.); (C.C.); (D.R.); (V.D.); (G.S.); (G.M.); (A.C.); (E.L.); (M.P.); (V.P.); (M.S.)
| | - Marco Puzzoni
- Medical Oncology Unit, University Hospital and University of Cagliari, SS 554 km 4500 Bivio per Sestu, Monserrato, 09042 Cagliari, Italy; (A.P.); (S.M.); (A.P.D.); (C.C.); (D.R.); (V.D.); (G.S.); (G.M.); (A.C.); (E.L.); (M.P.); (V.P.); (M.S.)
| | - Valeria Pusceddu
- Medical Oncology Unit, University Hospital and University of Cagliari, SS 554 km 4500 Bivio per Sestu, Monserrato, 09042 Cagliari, Italy; (A.P.); (S.M.); (A.P.D.); (C.C.); (D.R.); (V.D.); (G.S.); (G.M.); (A.C.); (E.L.); (M.P.); (V.P.); (M.S.)
| | - Massimo Castagnola
- Proteomics Laboratory, Centro Europeo di Ricerca sul Cervello, IRCCS Fondazione Santa Lucia, 00013 Rome, Italy;
| | - Mario Scartozzi
- Medical Oncology Unit, University Hospital and University of Cagliari, SS 554 km 4500 Bivio per Sestu, Monserrato, 09042 Cagliari, Italy; (A.P.); (S.M.); (A.P.D.); (C.C.); (D.R.); (V.D.); (G.S.); (G.M.); (A.C.); (E.L.); (M.P.); (V.P.); (M.S.)
| | - Gavino Faa
- Department of Medical Sciences and Public Health, AOU Cagliari, University of Cagliari, 09124 Cagliari, Italy;
- Department of Biology, College of Science and Technology, Temple University, Philadelphia, PA 19122, USA
| |
Collapse
|
3
|
Wang T, Ding G, Wang X, Cui Y, Ma X, Ma J, Wu J. Expression of EPB41L2 in Cancer-Associated Fibroblasts: Prognostic Implications for Bladder Cancer and Response to Immunotherapy. Arch Med Res 2024; 55:102927. [PMID: 38154234 DOI: 10.1016/j.arcmed.2023.102927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 10/25/2023] [Accepted: 11/23/2023] [Indexed: 12/30/2023]
Abstract
BACKGROUND Immunotherapy response in patients with bladder cancer (BLCA) treated with immune checkpoint inhibitors (ICIs) is variable. The accurate evaluation of immunotherapy efficacy may be facilitated by the tumor microenvironment (TME). Erythrocyte membrane protein band 4.1 like 2 (EPB41L2), a cytoskeletal protein with a regulatory role in the TME was intensively investigated to determine its biological characterization, clinical relevance, and predictive value for immunotherapy in BLCA. METHODS Comprehensive bioinformatics and statistical analyses were conducted to examine gene expression profile, TME components, immune contexture, molecular features, and prediction of immunotherapy response. Immunohistochemistry (IHC) validated the results of the bioinformatics analysis. Association between immune checkpoint genes (ICGs) and EPB41L2-based risk stratification was validated in the IMvigor210 cohort, and their association with ICI response was assessed. RESULTS EPB41L2 mRNA levels were decreased in BLCA compared to normal tissue. IHC showed reduced EPB41L2 staining intensity in early BLCA tissue. Nevertheless, elevated EPB41L2 expression was observed in cancer-associated fibroblasts (CAFs) with higher histological grade and pathological stage. High EPB41L2 expression served as a poor prognostic factor for BLCA. Single-cell RNA-seq and further analyses revealed that EPB41L2 was mainly expressed in CAFs and promoted TME remodeling. EPB41L2low/ICGshigh patients showed greater benefit from immunotherapy. Gene mutation analysis revealed a close relationship between EPB41L2 and the frequency of oncogenic mutations, including TP53 and FGFR3. CONCLUSION Comprehensive analysis and IHC confirmed the upregulation of EPB41L2 in BLCA CAFs and its association with TME remodeling. EPB41L2 and ICG expression were identified as combinatorial biomarkers to predict the response to immunotherapy.
Collapse
Affiliation(s)
- Tianqi Wang
- Department of Urology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong, China
| | - Guixin Ding
- Department of Urology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong, China
| | - Xiaoyu Wang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yuanshan Cui
- Department of Urology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong, China
| | - Xiaohong Ma
- Department of Urology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong, China
| | - Jian Ma
- Department of Urology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong, China
| | - Jitao Wu
- Department of Urology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong, China.
| |
Collapse
|
4
|
Jaramillo-Rangel G, Chávez-Briones MDL, Ancer-Arellano A, Miranda-Maldonado I, Ortega-Martínez M. Back to the Basics: Usefulness of Naturally Aged Mouse Models and Immunohistochemical and Quantitative Morphologic Methods in Studying Mechanisms of Lung Aging and Associated Diseases. Biomedicines 2023; 11:2075. [PMID: 37509714 PMCID: PMC10377355 DOI: 10.3390/biomedicines11072075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Revised: 06/17/2023] [Accepted: 07/19/2023] [Indexed: 07/30/2023] Open
Abstract
Aging-related molecular and cellular alterations in the lung contribute to an increased susceptibility of the elderly to devastating diseases. Although the study of the aging process in the lung may benefit from the use of genetically modified mouse models and omics techniques, these approaches are still not available to most researchers and produce complex results. In this article, we review works that used naturally aged mouse models, together with immunohistochemistry (IHC) and quantitative morphologic (QM) methods in the study of the mechanisms of the aging process in the lung and its most commonly associated disorders: cancer, chronic obstructive pulmonary disease (COPD), and infectious diseases. The advantage of using naturally aged mice is that they present characteristics similar to those observed in human aging. The advantage of using IHC and QM methods lies in their simplicity, economic accessibility, and easy interpretation, in addition to the fact that they provide extremely important information. The study of the aging process in the lung and its associated diseases could allow the design of appropriate therapeutic strategies, which is extremely important considering that life expectancy and the number of elderly people continue to increase considerably worldwide.
Collapse
Affiliation(s)
- Gilberto Jaramillo-Rangel
- Department of Pathology, School of Medicine, Autonomous University of Nuevo León, Monterrey 64460, Mexico
| | | | - Adriana Ancer-Arellano
- Department of Pathology, School of Medicine, Autonomous University of Nuevo León, Monterrey 64460, Mexico
| | - Ivett Miranda-Maldonado
- Department of Pathology, School of Medicine, Autonomous University of Nuevo León, Monterrey 64460, Mexico
| | - Marta Ortega-Martínez
- Department of Pathology, School of Medicine, Autonomous University of Nuevo León, Monterrey 64460, Mexico
| |
Collapse
|
5
|
Hafdaoui S, Ciaccio C, Castellotti B, Sciacca FL, Pantaleoni C, D'Arrigo S. Case report: A novel pathogenic FRMD7 variant in a Turner syndrome patient with familial idiopathic infantile nystagmus. Front Neurol 2023; 14:1199095. [PMID: 37545716 PMCID: PMC10398378 DOI: 10.3389/fneur.2023.1199095] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Accepted: 06/28/2023] [Indexed: 08/08/2023] Open
Abstract
Infantile idiopathic nystagmus (IIN) is an oculomotor disorder characterized by involuntary bilateral, periodic ocular oscillations, predominantly on the horizontal axis. X-linked IIN (XLIIN) is the most common form of congenital nystagmus, and the FERM domain-containing gene (FRMD7) is the most common cause of pathogenesis, followed by mutations in GPR143. To date, more than 60 pathogenic FRMD7 variants have been identified, and the physiopathological pathways leading to the disease are not yet completely understood. FRMD7-associated nystagmus usually affects male patients, while it shows incomplete penetrance in female patients, who are mostly asymptomatic but sometimes present with mild ocular oscillations or, occasionally, with clear nystagmus. Here we report the first case of a patient with Turner syndrome and INN in an XLIIN pedigree, in which we identified a novel frameshift mutation (c.1492dupT) in the FRMD7 gene: the absence of one X chromosome in the patient unmasked the presence of the familial genetic nystagmus.
Collapse
Affiliation(s)
- Sara Hafdaoui
- Department of Pediatric Neurosciences, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Claudia Ciaccio
- Department of Pediatric Neurosciences, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Barbara Castellotti
- Department of Medical Genetics and Neurogenetics, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Francesca L. Sciacca
- Laboratory of Cytogenetic, Neurological Biochemistry and Neuropharmacology Unit, Department of Diagnostic and Technology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Chiara Pantaleoni
- Department of Pediatric Neurosciences, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Stefano D'Arrigo
- Department of Pediatric Neurosciences, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| |
Collapse
|
6
|
Hyaluronan-Induced CD44-iASPP Interaction Affects Fibroblast Migration and Survival. Cancers (Basel) 2023; 15:cancers15041082. [PMID: 36831425 PMCID: PMC9954134 DOI: 10.3390/cancers15041082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 02/01/2023] [Accepted: 02/02/2023] [Indexed: 02/10/2023] Open
Abstract
In the present study, we show that the inhibitor of the apoptosis-stimulating protein of p53 (iASPP) physically interacts with the hyaluronan receptor CD44 in normal and transformed cells. We noticed that the CD44 standard isoform (CD44s), but not the variant isoform (CD44v), bound to iASPP via the ankyrin-binding domain in CD44s. The formation of iASPP-CD44s complexes was promoted by hyaluronan stimulation in fibroblasts but not in epithelial cells. The cellular level of p53 affected the amount of the iASPP-CD44 complex. iASPP was required for hyaluronan-induced CD44-dependent migration and adhesion of fibroblasts. Of note, CD44 altered the sub-cellular localization of the iASPP-p53 complex; thus, ablation of CD44 promoted translocation of iASPP from the nucleus to the cytoplasm, resulting in increased formation of a cytoplasmic iASPP-p53 complex in fibroblasts. Overexpression of iASPP decreased, but CD44 increased the level of intracellular reactive oxygen species (ROS). Knock-down of CD44s, in the presence of p53, led to increased cell growth and cell density of fibroblasts by suppression of p27 and p53. Our observations suggest that the balance of iASPP-CD44 and iASPP-p53 complexes affect the survival and migration of fibroblasts.
Collapse
|
7
|
Functions of CNKSR2 and Its Association with Neurodevelopmental Disorders. Cells 2022; 11:cells11020303. [PMID: 35053419 PMCID: PMC8774548 DOI: 10.3390/cells11020303] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 01/05/2022] [Accepted: 01/13/2022] [Indexed: 02/04/2023] Open
Abstract
The Connector Enhancer of Kinase Suppressor of Ras-2 (CNKSR2), also known as CNK2 or MAGUIN, is a scaffolding molecule that contains functional protein binding domains: Sterile Alpha Motif (SAM) domain, Conserved Region in CNK (CRIC) domain, PSD-95/Dlg-A/ZO-1 (PDZ) domain, Pleckstrin Homology (PH) domain, and C-terminal PDZ binding motif. CNKSR2 interacts with different molecules, including RAF1, ARHGAP39, and CYTH2, and regulates the Mitogen-Activated Protein Kinase (MAPK) cascade and small GTPase signaling. CNKSR2 has been reported to control the development of dendrite and dendritic spines in primary neurons. CNKSR2 is encoded by the CNKSR2 gene located in the X chromosome. CNKSR2 is now considered as a causative gene of the Houge type of X-linked syndromic mental retardation (MRXHG), an X-linked Intellectual Disability (XLID) that exhibits delayed development, intellectual disability, early-onset seizures, language delay, attention deficit, and hyperactivity. In this review, we summarized molecular features, neuronal function, and neurodevelopmental disorder-related variations of CNKSR2.
Collapse
|
8
|
Ning S, Hua L, Ji Z, Fan D, Meng X, Li Z, Wang Q, Guo Z. Protein 4.1 family and ion channel proteins interact to regulate the process of heart failure in rats. Acta Histochem 2021; 123:151748. [PMID: 34271280 DOI: 10.1016/j.acthis.2021.151748] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 06/24/2021] [Accepted: 06/25/2021] [Indexed: 02/06/2023]
Abstract
Heart failure (HF) is a major cause of death in cardiovascular diseases worldwide, and its molecular mechanisms and effective prevention strategies remain to be further studied. The myocardial cytoskeleton plays a pivotal role in many heart diseases. However, little is known about the function of the membrane cytoskeleton 4.1 protein family and related regulatory mechanisms in the pathogenesis of HF. In this study, we detected the localization and expression of the protein 4.1 family and ion channel proteins in a rat HF model induced by doxorubicin (DOX), and studied the interactions between them. Our results showed that compared with the control group, the HF group displayed an increased expression level of protein 4.1R and decreased levels of protein 4.1 G and 4.1 N. The Nav1.5 protein levels were significantly increased, while the SERCA2a and Cav1.2 protein levels were significantly decreased in the HF group. Furthermore, there is co-localization and interaction between protein 4.1R and Nav1.5, protein 4.1 G and SERCA2a, protein 4.1 N and Cav1.2, respectively. Taken together, the results indicated that the protein 4.1 family might be involved in the occurrence and development of HF through its interaction with ion channel proteins, suggesting that 4.1 proteins may serve as a novel therapeutic target for HF.
Collapse
Affiliation(s)
- Shuwei Ning
- Zhengzhou Key Laboratory, Zhengzhou No. 7 People's Hospital, Zhengzhou, 450016, China
| | - Lei Hua
- Zhengzhou Key Laboratory, Zhengzhou No. 7 People's Hospital, Zhengzhou, 450016, China
| | - Zhenyu Ji
- Henan Academy of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450052, China
| | - Dandan Fan
- Henan Academy of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450052, China
| | - Xiangguang Meng
- Zhengzhou Key Laboratory, Zhengzhou No. 7 People's Hospital, Zhengzhou, 450016, China
| | - Zhiying Li
- Zhengzhou Key Laboratory, Zhengzhou No. 7 People's Hospital, Zhengzhou, 450016, China
| | - Qian Wang
- Zhengzhou Key Laboratory, Zhengzhou No. 7 People's Hospital, Zhengzhou, 450016, China
| | - Zhikun Guo
- Zhengzhou Key Laboratory, Zhengzhou No. 7 People's Hospital, Zhengzhou, 450016, China; Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, 453003, China.
| |
Collapse
|
9
|
Ikeuchi M, Yuki R, Saito Y, Nakayama Y. The tumor suppressor LATS2 reduces v-Src-induced membrane blebs in a kinase activity-independent manner. FASEB J 2021; 35:e21242. [PMID: 33368671 DOI: 10.1096/fj.202001909r] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 11/05/2020] [Accepted: 11/19/2020] [Indexed: 12/24/2022]
Abstract
When cells with excess DNA, such as tetraploid cells, undergo cell division, it can contribute to cellular transformation via asymmetrical chromosome segregation-generated genetic diversity. Cell cycle progression of tetraploid cells is suppressed by large tumor suppressor 2 (LATS2) kinase-induced inhibitory phosphorylation of the transcriptional coactivator Yes-associated protein (YAP). We recently reported that the oncogene v-Src induces tetraploidy and promotes cell cycle progression of tetraploid cells by suppressing LATS2 activity. We explore here the mechanism by which v-Src suppresses LATS2 activity and the role of LATS2 in v-Src-expressing cells. LATS2 was directly phosphorylated by v-Src and the proto-oncogene c-Src, resulting in decreased LATS2 kinase activity. This kinase-deficient LATS2 accumulated in a YAP transcriptional activity-dependent manner, and knockdown of either LATS2 or the LATS2-binding partner moesin-ezrin-radixin-like protein (Merlin) accelerated v-Src-induced membrane bleb formation. Upon v-Src expression, the interaction of Merlin with LATS2 was increased possibly due to a decrease in Merlin phosphorylation at Ser518, the dephosphorylation of which is required for the open conformation of Merlin and interaction with LATS2. LATS2 was colocalized with Merlin at the plasma membrane in a manner that depends on the Merlin-binding region of LATS2. The bleb formation in v-Src-expressing and LATS2-knockdown cells was rescued by the reexpression of wild-type or kinase-dead LATS2 but not the LATS2 mutant lacking the Merlin-binding region. These results suggest that the kinase-deficient LATS2 plays a role with Merlin at the plasma membrane in the maintenance of cortical rigidity in v-Src-expressing cells, which may cause tumor suppression.
Collapse
Affiliation(s)
- Masayoshi Ikeuchi
- Department of Biochemistry & Molecular Biology, Kyoto Pharmaceutical University, Kyoto, Japan.,DC1, Japan Society for the Promotion of Science, Tokyo, Japan
| | - Ryuzaburo Yuki
- Department of Biochemistry & Molecular Biology, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Youhei Saito
- Department of Biochemistry & Molecular Biology, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Yuji Nakayama
- Department of Biochemistry & Molecular Biology, Kyoto Pharmaceutical University, Kyoto, Japan
| |
Collapse
|
10
|
Li B, Zhang X, Lu Y, Zhao L, Guo Y, Guo S, Kang Q, Liu J, Dai L, Zhang L, Fan D, Ji Z. Protein 4.1R affects photodynamic therapy for B16 melanoma by regulating the transport of 5-aminolevulinic acid. Exp Cell Res 2021; 399:112465. [PMID: 33385415 DOI: 10.1016/j.yexcr.2020.112465] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 12/21/2020] [Accepted: 12/24/2020] [Indexed: 12/12/2022]
Abstract
Melanoma is the most aggressive malignant tumor of skin cancer as it can grow rapidly and metastasize. Photodynamic therapy (PDT) is a promising cancer ablation method for skin tumors, although it lacks efficiency owing to factors such as tumor characteristics, delivery of photosensitizers, immune response in vivo etc. Extensive investigation of molecules that can potentially modulate treatment efficacy is required. Protein 4.1R is a cytoskeletal protein molecule. Previous studies have shown that protein 4.1R knockdown reduces PDT sensitivity in mouse embryonic fibroblast cells. However, the functional role of protein 4.1R in melanoma is unclear. In this study, we aimed to elucidate the effect of protein 4.1R on PDT for melanoma in mice and the mechanism of anti-tumor immunity. Our results indicated that CRISPR/Cas9-mediated protein 4.1R knockout promotes the proliferation, migration, and invasion of B16 cells. We further investigated the potential mechanism of protein 4.1R on tumor cell PDT sensitivity. Our results showed that protein 4.1R knockout reduced the expression of membrane transporters γ-aminobutyric acid transporter (GAT)-1 and (GAT)-2 in B16 cells, which affected 5-ALA transmembrane transport and reduced the efficiency of PDT on B16 cells. Protein 4.1R knockout downregulated the anti-tumor immune response triggered by PDT in vivo. In conclusion, our data suggest that protein 4.1R is an important regulator in PDT for tumors and may promote the progress and efficacy of melanoma treatment.
Collapse
Affiliation(s)
- Bowen Li
- Henan Institute of Medical and Pharmaceutical Sciences, Academy of Medical Science, Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Xiaolin Zhang
- People's Hospital of Zhengzhou, 33 Huanghe Road, Zhengzhou, 450000, Henan, China
| | - Yu Lu
- Henan Institute of Medical and Pharmaceutical Sciences, Academy of Medical Science, Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Luyang Zhao
- BGI College & Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Yaxin Guo
- Henan Institute of Medical and Pharmaceutical Sciences, Academy of Medical Science, Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Shuangshuang Guo
- BGI College & Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Qiaozhen Kang
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Jingjing Liu
- Henan Institute of Medical and Pharmaceutical Sciences, Academy of Medical Science, Zhengzhou University, Zhengzhou, 450052, Henan, China; BGI College & Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Liping Dai
- Henan Institute of Medical and Pharmaceutical Sciences, Academy of Medical Science, Zhengzhou University, Zhengzhou, 450052, Henan, China; BGI College & Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Liguo Zhang
- Henan Institute of Medical and Pharmaceutical Sciences, Academy of Medical Science, Zhengzhou University, Zhengzhou, 450052, Henan, China; BGI College & Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Dandan Fan
- Henan Institute of Medical and Pharmaceutical Sciences, Academy of Medical Science, Zhengzhou University, Zhengzhou, 450052, Henan, China; BGI College & Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450052, Henan, China; Henan Key Laboratory for Pharmacology of Liver Diseases, Zhengzhou, 450052, Henan, China.
| | - Zhenyu Ji
- Henan Institute of Medical and Pharmaceutical Sciences, Academy of Medical Science, Zhengzhou University, Zhengzhou, 450052, Henan, China; BGI College & Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450052, Henan, China; Henan Key Laboratory for Pharmacology of Liver Diseases, Zhengzhou, 450052, Henan, China.
| |
Collapse
|
11
|
Rouka E, Gourgoulianni N, Lüpold S, Hatzoglou C, Gourgoulianis K, Blanckenhorn WU, Zarogiannis SG. The Drosophila septate junctions beyond barrier function: Review of the literature, prediction of human orthologs of the SJ-related proteins and identification of protein domain families. Acta Physiol (Oxf) 2021; 231:e13527. [PMID: 32603029 DOI: 10.1111/apha.13527] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 06/20/2020] [Accepted: 06/22/2020] [Indexed: 12/20/2022]
Abstract
The involvement of Septate Junctions (SJs) in critical cellular functions that extend beyond their role as diffusion barriers in the epithelia and the nervous system has made the fruit fly an ideal model for the study of human diseases associated with impaired Tight Junction (TJ) function. In this study, we summarized current knowledge of the Drosophila melanogaster SJ-related proteins, focusing on their unconventional functions. Additionally, we sought to identify human orthologs of the corresponding genes as well as protein domain families. The systematic literature search was performed in PubMed and Scopus databases using relevant key terms. Orthologs were predicted using the DIOPT tool and aligned protein regions were determined from the Pfam database. 3-D models of the smooth SJ proteins were built on the Phyre2 and DMPFold protein structure prediction servers. A total of 30 proteins were identified as relatives to the SJ cellular structure. Key roles of these proteins, mainly in the regulation of morphogenetic events and cellular signalling, were highlighted. The investigation of protein domain families revealed that the SJ-related proteins contain conserved domains that are required not only for cell-cell interactions and cell polarity but also for cellular signalling and immunity. DIOPT analysis of orthologs identified novel human genes as putative functional homologs of the fruit fly SJ genes. A gap in our knowledge was identified regarding the domains that occur in the proteins encoded by eight SJ-associated genes. Future investigation of these domains is needed to provide functional information.
Collapse
Affiliation(s)
- Erasmia Rouka
- Department of Physiology Faculty of Medicine School of Health Sciences University of ThessalyBIOPOLIS Larissa Greece
| | - Natalia Gourgoulianni
- Department of Evolutionary Biology and Environmental Studies University of Zurich Zurich Switzerland
| | - Stefan Lüpold
- Department of Evolutionary Biology and Environmental Studies University of Zurich Zurich Switzerland
| | - Chrissi Hatzoglou
- Department of Physiology Faculty of Medicine School of Health Sciences University of ThessalyBIOPOLIS Larissa Greece
- Department of Respiratory Medicine Faculty of Medicine School of Health Sciences University of ThessalyBIOPOLIS Larissa Greece
| | - Konstantinos Gourgoulianis
- Department of Respiratory Medicine Faculty of Medicine School of Health Sciences University of ThessalyBIOPOLIS Larissa Greece
| | - Wolf U. Blanckenhorn
- Department of Evolutionary Biology and Environmental Studies University of Zurich Zurich Switzerland
| | - Sotirios G. Zarogiannis
- Department of Physiology Faculty of Medicine School of Health Sciences University of ThessalyBIOPOLIS Larissa Greece
- Department of Respiratory Medicine Faculty of Medicine School of Health Sciences University of ThessalyBIOPOLIS Larissa Greece
| |
Collapse
|
12
|
Ni H, Ji D, Huang Z, Li J. SMAGP knockdown inhibits the malignant phenotypes of glioblastoma cells by inactivating the PI3K/Akt pathway. Arch Biochem Biophys 2020; 695:108628. [PMID: 33049294 DOI: 10.1016/j.abb.2020.108628] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 10/04/2020] [Accepted: 10/09/2020] [Indexed: 12/31/2022]
Abstract
Small trans-membrane and glycosylated protein (SMAGP), a novel small trans-membrane glycoprotein, is reported to be upregulated in multiple cancers and involved in tumor development. However, little is known about its role in the development of glioblastoma (GBM). GEPIA database was used to analyze SMAGP expression and evaluate the prognostic value of SMAGP in GBM. GO and KEGG pathway enrichment analyses were used to predict the biological functions and pathways of SMAGP and 948 SMAGP-correlated genes using DAVID database. Cell viability, colony formation ability, apoptosis, and invasion were evaluated by MTT, colony formation assay, flow cytometry analysis, and Transwell invasion assay, respectively. Western blot was applied to detect the expression of SMAGP, matrix metalloproteinase (MMP)-2, and MMP-9 and analyze the changes of phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt) signaling. Results showed that SMAGP was upregulated and correlated with poor prognosis in GBM. Functional annotation analysis revealed that SMAGP and 948 SMAGP-correlated genes were primarily associated with cell adhesion and PI3K/Akt pathway. SMAGP interference inhibited cell viability and colony formation ability and promoted apoptosis in GBM cells. Moreover, SMAGP interference inhibited GBM cell invasion and suppressed MMP-2 and MMP-9 expression. Additionally, SMAGP silencing inhibited the PI3K/Akt pathway in GBM cells. Overexpression of Akt abolished the effects of SMAGP knockdown on the malignant phenotypes of GBM cells. In conclusion, SMAGP silencing inhibited the malignant phenotypes of GBM cells by inactivating the PI3K/Akt pathway.
Collapse
Affiliation(s)
- Hongzao Ni
- Department of Neurosurgery, The Affiliated Huai'an Hospital of Xuzhou Medical University, The Second People's Hospital of Huai'an, Huai'an, 223002, China
| | - Daofei Ji
- Department of Neurosurgery, The Second Hospital of Xuzhou Medical University, Xuzhou, 221006, China
| | - Zhixiong Huang
- Department of Neurology, Nanshi Hospital, Nanyang, 473065, China
| | - Jing Li
- Department of Neurosurgery, The Affiliated Huai'an Hospital of Xuzhou Medical University, The Second People's Hospital of Huai'an, Huai'an, 223002, China.
| |
Collapse
|
13
|
Lv T, Miao Y, Xu T, Sun W, Sang Y, Jia F, Zhang X. Circ-EPB41L5 regulates the host gene EPB41L5 via sponging miR-19a to repress glioblastoma tumorigenesis. Aging (Albany NY) 2020; 12:318-339. [PMID: 31905344 PMCID: PMC6977680 DOI: 10.18632/aging.102617] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Accepted: 12/05/2019] [Indexed: 12/14/2022]
Abstract
Background: Circular RNAs (circRNAs) are widely expressed non-coding RNAs in eukaryotic cells, involved in regulating tumorigenesis of several types of cancers. However, the expression profiles and the precise functional role in glioblastoma remain unclear. Results: Circ-EPB41L5 was downregulated in glioblastoma tissues and cell lines compared to the normal brain tissues and cell lines. Low circ-EPB41L5 expression was correlated to the poor prognosis of glioblastoma patients, while the overexpression inhibited proliferation, clone formation, migration, and invasion abilities of glioma cells, and the suppression had counter effects. Furthermore, RNA-seq results determined that the host gene was the target gene of circ-EPB41L5, which served as a sponge against miR-19a and inhibited miR-19a activity from upregulating the expression of EPB41L5. Finally, we found that circ-EPB41L5 regulated the RhoC expression and phosphorylation of AKT through EPB41L5. Conclusion: The current study highlights a novel suppressive function of circ-EPB41L5 and reveals that circ-EPB41L5/miR-19a/EPB41L5/p-AKT regulatory axis plays a striking role in the progression of glioblastoma, which provides a novel insight into the mechanisms underlying glioblastoma. Methods: The expression profiles of circRNAs in glioblastoma were determined by Illumina HiSeq from six glioblastoma tissues and six normal brain tissues. Then, the correlation between circ-EPB41L5 expression and clinical features and the survival time of 45 glioblastoma patients was detected. The interaction between circ-EPB41L5, miR-19a, and EPB41L5 was assessed by luciferase reporter and RNA pull-down assays. The effects of expression of the ectopic intervention of circ-EPB41L5 or EPB41L5 on proliferation, clone formation, migration, and invasion in vitro and tumorigenesis in vivo were observed to evaluate the function of circ-EPB41L5 or EPB41L5.
Collapse
Affiliation(s)
- Tao Lv
- Department of Neurosurgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Yifeng Miao
- Department of Neurosurgery, Ren Ji Hospital South Campus, School of Medicine, Shanghai Jiao Tong University, Shanghai 201112, China
| | - Tianqi Xu
- Department of Neurosurgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Wenhua Sun
- Department of Neurosurgery, Ren Ji Hospital South Campus, School of Medicine, Shanghai Jiao Tong University, Shanghai 201112, China
| | - Youzhou Sang
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Feng Jia
- Department of Neurosurgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Xiaohua Zhang
- Department of Neurosurgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| |
Collapse
|
14
|
Chen L, Wang T, Ji X, Ding C, Liang T, Liu X, Lu J, Guo X, Kang Q, Ji Z. Cytoskeleton protein 4.1R suppresses murine keratinocyte cell hyperproliferation via activating the Akt/ERK pathway in an EGFR-dependent manner. Exp Cell Res 2019; 384:111648. [DOI: 10.1016/j.yexcr.2019.111648] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 08/30/2019] [Accepted: 09/24/2019] [Indexed: 01/01/2023]
|
15
|
Heldin P, Kolliopoulos C, Lin CY, Heldin CH. Involvement of hyaluronan and CD44 in cancer and viral infections. Cell Signal 2019; 65:109427. [PMID: 31654718 DOI: 10.1016/j.cellsig.2019.109427] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 09/19/2019] [Accepted: 09/19/2019] [Indexed: 02/07/2023]
Abstract
Hyaluronan and its major receptor CD44 are ubiquitously distributed. They have important structural as well as signaling roles, regulating tissue homeostasis, and their expression levels are tightly regulated. In addition to signaling initiated by the interaction of the intracellular domain of CD44 with cytoplasmic signaling molecules, CD44 has important roles as a co-receptor for different types of receptors of growth factors and cytokines. Dysregulation of hyaluronan-CD44 interactions is seen in diseases, such as inflammation and cancer. In the present communication, we discuss the mechanism of hyaluronan-induced signaling via CD44, as well as the involvement of hyaluronan-engaged CD44 in malignancies and in viral infections.
Collapse
Affiliation(s)
- Paraskevi Heldin
- Department of Medical Biochemistry and Microbiology, Box 582, Uppsala University, SE-751 23 Uppsala, Sweden.
| | - Constantinos Kolliopoulos
- Department of Medical Biochemistry and Microbiology, Box 582, Uppsala University, SE-751 23 Uppsala, Sweden
| | - Chun-Yu Lin
- Department of Medical Biochemistry and Microbiology, Box 582, Uppsala University, SE-751 23 Uppsala, Sweden; Division of Infectious Diseases, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University Department of Surgery, Uppsala University, Sweden; Department of Surgical Sciences, Uppsala University, Akademiska Hospital, 751 85 Uppsala, Sweden
| | - Carl-Henrik Heldin
- Department of Medical Biochemistry and Microbiology, Box 582, Uppsala University, SE-751 23 Uppsala, Sweden.
| |
Collapse
|
16
|
Yuan X, Piao L, Wang L, Han X, Zhuang M, Liu Z. Pivotal roles of protein 4.1B/DAL‑1, a FERM‑domain containing protein, in tumor progression (Review). Int J Oncol 2019; 55:979-987. [PMID: 31545421 DOI: 10.3892/ijo.2019.4877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Accepted: 09/04/2019] [Indexed: 11/06/2022] Open
Abstract
Protein 4.1B/DAL‑1, encoded by erythrocyte membrane protein band 4.1‑like 3 (EPB41L3), belongs to the protein 4.1 superfamily, a group of proteins that share a conserved four.one‑ezrin‑radixin‑moesin (FERM) domain. Protein 4.1B/DAL‑1 serves a crucial role in cytoskeletal organization and a number of processes through multiple interactions with membrane proteins via its FERM, spectrin‑actin‑binding and C‑terminal domains. A number of studies have indicated that a loss of EPB41L3 expression is commonly observed in lung cancer, breast cancer, esophageal squamous cell carcinoma and meningiomas. DNA methylation and a loss of heterozygosity have been reported to contribute to the downregulation of EPB41L3. To date, the biological functions of protein 4.1B/DAL‑1 in carcinogenesis remain unknown. The present review summarizes the current understanding of the role of protein 4.1B/DAL‑1 in cancer and highlights its potential as a cancer diagnostic and prognostic biomarker in cancer therapeutics.
Collapse
Affiliation(s)
- Xiaofeng Yuan
- Department of Orthopaedics, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213000, P.R. China
| | - Lianhua Piao
- Institute of Bioinformatics and Medical Engineering, Jiangsu University of Technology, Changzhou, Jiangsu 213001, P.R. China
| | - Luhui Wang
- Department of Orthopaedics, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213000, P.R. China
| | - Xu Han
- Department of Urology, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213000, P.R. China
| | - Ming Zhuang
- Department of Orthopaedics, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213000, P.R. China
| | - Zhiwei Liu
- Department of Orthopaedics, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213000, P.R. China
| |
Collapse
|
17
|
Feng G, Guo K, Yan Q, Ye Y, Shen M, Ruan S, Qiu S. Expression of Protein 4.1 Family in Breast Cancer: Database Mining for 4.1 Family Members in Malignancies. Med Sci Monit 2019; 25:3374-3389. [PMID: 31063460 PMCID: PMC6524556 DOI: 10.12659/msm.914085] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2018] [Accepted: 01/25/2019] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND The protein 4.1 family is a family of cytoskeletal proteins that play an important role in maintaining normal cell morphology and cell adhesion, migration, division, and intercellular signaling. The main aim of this study was to explore the prognostic significance of the protein 4.1 family in breast cancer (BC) patients and to provide new biomarkers and therapeutic targets for the diagnosis and treatment of BC. MATERIAL AND METHODS The expression of 4.1 family members in various tumor types was compared to normal controls using the ONCOMINE and GOBO databases. The prognostic significance of the 4.1 family in BC patients was determined by Kaplan-Meier Plotter. RESULTS EPB41L2 (4.1G) was expressed at higher levels in normal tissues compared with BC patients for all 4.1 family members. In survival analysis, 4.1G and EPB41 (4.1R) mRNA high expressions were associated with better survival in BC patients. Moreover, 4.1G high expression was significantly associated with longer overall survival (OS) in luminal A and protracted relapse-free survival (RFS) in luminal B subtype BC patients who received Tamoxifen treatment. In addition, high expression of each 4.1 family member also showed better prognostic value in different molecular subtypes of BC. CONCLUSIONS These results indicate that the protein 4.1 family can be regarded as novel biomarkers and potential therapeutic targets for BC. Further research is needed to explore the detailed biological functions.
Collapse
Affiliation(s)
- Guan Feng
- The First Clinical College of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, P.R. China
| | - Kaibo Guo
- The First Clinical College of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, P.R. China
| | - Qingying Yan
- The First Clinical College of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, P.R. China
| | - Ye Ye
- Hangzhou Vocational and Technical College, Hangzhou, Zhejiang, P.R. China
| | - Minhe Shen
- Department of Medical Oncology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, P.R. China
| | - Shanming Ruan
- Department of Medical Oncology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, P.R. China
| | - Shengliang Qiu
- The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, P.R. China
| |
Collapse
|
18
|
Hartsough EJ, Weiss MB, Heilman SA, Purwin TJ, Kugel CH, Rosenbaum SR, Erkes DA, Tiago M, HooKim K, Chervoneva I, Aplin AE. CADM1 is a TWIST1-regulated suppressor of invasion and survival. Cell Death Dis 2019; 10:281. [PMID: 30911007 PMCID: PMC6433918 DOI: 10.1038/s41419-019-1515-3] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 03/05/2019] [Accepted: 03/07/2019] [Indexed: 12/11/2022]
Abstract
Metastatic cancer remains a clinical challenge; however, patients diagnosed prior to metastatic dissemination have a good prognosis. The transcription factor, TWIST1 has been implicated in enhancing the migration and invasion steps within the metastatic cascade, but the range of TWIST1-regulated targets is poorly described. In this study, we performed expression profiling to identify the TWIST1-regulated transcriptome of melanoma cells. Gene ontology pathway analysis revealed that TWIST1 and epithelial to mesenchymal transition (EMT) were inversely correlated with levels of cell adhesion molecule 1 (CADM1). Chromatin immunoprecipitation (ChIP) studies and promoter assays demonstrated that TWIST1 physically interacts with the CADM1 promoter, suggesting TWIST1 directly represses CADM1 levels. Increased expression of CADM1 resulted in significant inhibition of motility and invasiveness of melanoma cells. In addition, elevated CADM1 elicited caspase-independent cell death in non-adherent conditions. Expression array analysis suggests that CADM1 directed non-adherent cell death is associated with loss of mitochondrial membrane potential and subsequent failure of oxidative phosphorylation pathways. Importantly, tissue microarray analysis and clinical data from TCGA indicate that CADM1 expression is inversely associated with melanoma progression and positively correlated with better overall survival in patients. Together, these data suggest that CADM1 exerts tumor suppressive functions in melanoma by reducing invasive potential and may be considered a biomarker for favorable prognosis.
Collapse
Affiliation(s)
- Edward J Hartsough
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA, 19107, USA.,Sidney Kimmel Cancer Center at Jefferson, Philadelphia, PA, 19107, USA.,Department of Pharmacology and Physiology at Drexel University College of Medicine, Philadelphia, PA, 19102, USA
| | - Michele B Weiss
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA, 19107, USA
| | - Shea A Heilman
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA, 19107, USA
| | - Timothy J Purwin
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA, 19107, USA
| | - Curtis H Kugel
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA, 19107, USA
| | - Sheera R Rosenbaum
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA, 19107, USA
| | - Dan A Erkes
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA, 19107, USA
| | - Manoela Tiago
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA, 19107, USA
| | - Kim HooKim
- Departments of Pathology, Anatomy, and Cell Biology, Thomas Jefferson University, Philadelphia, PA, 19107, USA
| | - Inna Chervoneva
- Sidney Kimmel Cancer Center at Jefferson, Philadelphia, PA, 19107, USA.,Division of Biostatistics in Pharmacology and Experimental Therapeutics, Thomas Jefferson University, Philadelphia, PA, 19107, USA
| | - Andrew E Aplin
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA, 19107, USA. .,Sidney Kimmel Cancer Center at Jefferson, Philadelphia, PA, 19107, USA.
| |
Collapse
|
19
|
Jeong MH, Park SY, Lee SH, Seo J, Yoo JY, Park SH, Kim MJ, Lee S, Jang S, Choi HK, Lee JE, Shin SJ, Choi KC, Cheong JH, Yoon HG. EPB41L5 Mediates TGFβ-Induced Metastasis of Gastric Cancer. Clin Cancer Res 2019; 25:3617-3629. [PMID: 30814110 DOI: 10.1158/1078-0432.ccr-18-2959] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Revised: 11/27/2018] [Accepted: 02/21/2019] [Indexed: 11/16/2022]
Abstract
PURPOSE Because of disease heterogeneity, limited studies on effective chemotherapies and therapeutic agents for advanced gastric cancer are available. Erythrocyte membrane protein band 4.1-like 5 (EPB41L5) has critical roles in renal and breast cancer metastasis. However, its role in metastatic gastric cancer remains unknown. EXPERIMENTAL DESIGN The specimens of 78 gastric cancer patients were analyzed by oligonucleotide microarray and survival analysis. In vitro experiments and metastatic mice models were used to assess the effects of EPB41L5 on gastric cancer metastasis. RESULTS Gastric cancer patients with high EPB41L5 levels had poor prognosis and low survival rate. Further, TGFβ1-induced EPB41L5 expression promoted gastric cancer cell migration and invasion by Smad-dependent TGFβ signaling. Phospho-Smad3 recruitment to the EPB41L5 promoter was significantly inhibited by a TGFβ inhibitor. EPB41L5 overexpression increased lung metastasis of gastric cancer cells in nude mice, which was completely reversed by anti-EPB41L5 monoclonal antibody treatment. Importantly, p120-catenin knockdown abolished EPB41L5-enhanced gastric cancer cell metastasis. Anti-EPB41L5 monoclonal antibody treatment blocked the association of EPB41L5 with p120-catenin. CONCLUSIONS TGFβ/EPB41L5/p120-catenin axis regulates gastric cancer cell metastasis, and EPB41L5 is a promising therapeutic target for advanced gastric cancer.
Collapse
Affiliation(s)
- Mi-Hyeon Jeong
- Department of Biochemistry and Molecular Biology, Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, Seoul, Korea
| | - Soo-Yeon Park
- Department of Biochemistry and Molecular Biology, Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, Seoul, Korea
| | - Seung-Hyun Lee
- Department of Biochemistry and Molecular Biology, Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, Seoul, Korea
| | - JaeSung Seo
- Department of Biochemistry and Molecular Biology, Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, Seoul, Korea
| | - Jung-Yoon Yoo
- Department of Biochemistry and Molecular Biology, Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, Seoul, Korea
| | - Seung-Ho Park
- Department of Biomedical Sciences, Asan Medical Center, AMIST, University of Ulsan College of Medicine, Seoul, Korea
| | - Mi Jeong Kim
- Department of Biochemistry and Molecular Biology, Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, Seoul, Korea
| | - SooYeon Lee
- Department of Biochemistry and Molecular Biology, Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, Seoul, Korea
| | - Subhin Jang
- Department of Biochemistry and Molecular Biology, Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, Seoul, Korea
| | - Hyo-Kyoung Choi
- Division of Nutrition and Metabolism Research Group, Korea Food Research Institute, Gyeonggi-do, Korea
| | - Jae Eun Lee
- Department of Surgery, Yonsei University College of Medicine, Seoul, Korea
| | - Su-Jin Shin
- Department of Pathology, Hanyang University College of Medicine, Seoul, Korea
| | - Kyung-Chul Choi
- Department of Biomedical Sciences, Asan Medical Center, AMIST, University of Ulsan College of Medicine, Seoul, Korea.
| | - Jae-Ho Cheong
- Department of Biochemistry and Molecular Biology, Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, Seoul, Korea. .,Department of Surgery, Yonsei University College of Medicine, Seoul, Korea
| | - Ho-Geun Yoon
- Department of Biochemistry and Molecular Biology, Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, Seoul, Korea.
| |
Collapse
|
20
|
Saifetiarova J, Bhat MA. Ablation of cytoskeletal scaffolding proteins, Band 4.1B and Whirlin, leads to cerebellar purkinje axon pathology and motor dysfunction. J Neurosci Res 2018; 97:313-331. [PMID: 30447021 DOI: 10.1002/jnr.24352] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Revised: 10/12/2018] [Accepted: 10/12/2018] [Indexed: 12/23/2022]
Abstract
The cerebellar cortex receives neural information from other brain regions to allow fine motor coordination and motor learning. The primary output neurons from the cerebellum are the Purkinje neurons that transmit inhibitory responses to deep cerebellar nuclei through their myelinated axons. Altered morphological organization and electrical properties of the Purkinje axons lead to detrimental changes in locomotor activity often leading to cerebellar ataxias. Two cytoskeletal scaffolding proteins Band 4.1B (4.1B) and Whirlin (Whrn) have been previously shown to play independent roles in axonal domain organization and maintenance in myelinated axons in the spinal cord and sciatic nerves. Immunoblot analysis had indicated cerebellar expression for both 4.1B and Whrn; however, their subcellular localization and cerebellum-specific functions have not been characterized. Using 4.1B and Whrn single and double mutant animals, we show that both proteins are expressed in common cellular compartments of the cerebellum and play cooperative roles in preservation of the integrity of Purkinje neuron myelinated axons. We demonstrate that both 4.1B and Whrn are required for the maintenance of axonal ultrastructure and health. Loss of 4.1B and Whrn leads to axonal transport defects manifested by formation of swellings containing cytoskeletal components, membranous organelles, and vesicles. Moreover, ablation of both proteins progressively affects cerebellar function with impairment in locomotor performance detected by altered gait parameters. Together, our data indicate that 4.1B and Whrn are required for maintaining proper axonal cytoskeletal organization and axonal domains, which is necessary for cerebellum-controlled fine motor coordination.
Collapse
Affiliation(s)
- Julia Saifetiarova
- Department of Cellular and Integrative Physiology, Center for Biomedical Neuroscience, Long School of Medicine, University of Texas Health Science Center, San Antonio, Texas
| | - Manzoor A Bhat
- Department of Cellular and Integrative Physiology, Center for Biomedical Neuroscience, Long School of Medicine, University of Texas Health Science Center, San Antonio, Texas
| |
Collapse
|
21
|
Fierro J, Haynes DR, Washbourne P. 4.1Ba is necessary for glutamatergic synapse formation in the sensorimotor circuit of developing zebrafish. PLoS One 2018; 13:e0205255. [PMID: 30286167 PMCID: PMC6171929 DOI: 10.1371/journal.pone.0205255] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Accepted: 09/23/2018] [Indexed: 01/04/2023] Open
Abstract
During the process of synapse formation, thousands of proteins assemble at prospective sites of cell-cell communication. Although many of these proteins have been identified, the roles they play in generating functional connections during development remain unknown. 4.1 scaffolding proteins have been implicated in synapse formation and maturation in vitro, but in vivo studies for some family members have suggested these proteins are not important for this role. We examined the role of family member 4.1B because it has been implicated in glutamatergic synaptogenesis, but has not been described in vivo. We identified two 4.1B genes in zebrafish, 4.1Ba and 4.1Bb, by sequence comparisons and synteny analysis. In situ hybridization shows these genes are differentially expressed, with 4.1Ba expressed primarily in the nervous system and 4.1Bb expressed in the nervous system and muscle, but not the spinal cord. We focused our studies on 4.1Ba in the spinal cord. 4.1Ba knockdown reduced the number of glutamatergic synapses at caudal primary motor neurons and caused an increase in the duration of touch-evoked coiling. These results suggest 4.1Ba is important for the formation of functional glutamatergic synapses in the developing zebrafish spinal cord.
Collapse
Affiliation(s)
- Javier Fierro
- Institute of Neuroscience, University of Oregon, Eugene, Oregon, United States of America
| | - Dylan R. Haynes
- Institute of Neuroscience, University of Oregon, Eugene, Oregon, United States of America
| | - Philip Washbourne
- Institute of Neuroscience, University of Oregon, Eugene, Oregon, United States of America
| |
Collapse
|
22
|
Chang RM, Pei L, Fang F, Xu JF, Yang H, Zuo CH, Zhou JH, Luo GQ, Yang LY. YMO1 suppresses invasion and metastasis by inhibiting RhoC signaling and predicts favorable prognosis in hepatocellular carcinoma. Oncotarget 2018; 7:55585-55600. [PMID: 27487132 PMCID: PMC5342438 DOI: 10.18632/oncotarget.10866] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2015] [Accepted: 06/29/2016] [Indexed: 12/22/2022] Open
Abstract
Previous studies have shown that 4.1 proteins, which are deregulated in many cancers, contribute to cell adhesion and motility. Yurt/Mosaic eyes-like 1 (YMO1) is a member of 4.1 protein family but it is unclear whether YMO1 plays a role in tumor invasion. This study aimed to investigate the effects of YMO1 on hepatocellular carcinoma (HCC) and attempted to elucidate the underlying molecular mechanisms. YMO1 expression in HCC tissues and its correlation with clinicopathological features and postoperative prognosis was analyzed. The results showed that YMO1 was down-regulated in the highly metastatic HCC cell line and in human tumor tissues. Underexpression of YMO1 indicated poor prognosis of HCC patients. Restoration of YMO1 expression caused a significant decrease in cell migration and invasiveness in vitro. In vivo study showed that YMO1 reduced liver tumor invasion and metastasis in xenograft mice. YMO1 directly inhibited RhoC activation. YMO1 expression in HCC was regulated by PAX5. Analysis of YMO1 expression levels in human HCC patients revealed a significant correlation of YMO1 expression with PAX5 and RhoC. Our findings revealed that YMO1 predicts favorable prognosis and the data suggest that YMO1 suppresses tumor invasion and metastasis by inhibiting RhoC activity.
Collapse
Affiliation(s)
- Rui-Min Chang
- Liver Cancer Laboratory, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Lei Pei
- Liver Cancer Laboratory, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Feng Fang
- Liver Cancer Laboratory, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Jiang-Feng Xu
- Liver Cancer Laboratory, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Hao Yang
- Liver Cancer Laboratory, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Chao-Hui Zuo
- Department of Abdominal Surgical Oncology, Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Jian-Hua Zhou
- Department of Pathology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Geng-Qiu Luo
- Department of Pathology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Lian-Yue Yang
- Liver Cancer Laboratory, Xiangya Hospital, Central South University, Changsha 410008, China.,Department of Surgery, Xiangya Hospital, Central South University, Changsha 410008, China
| |
Collapse
|
23
|
Duran CL, Howell DW, Dave JM, Smith RL, Torrie ME, Essner JJ, Bayless KJ. Molecular Regulation of Sprouting Angiogenesis. Compr Physiol 2017; 8:153-235. [PMID: 29357127 DOI: 10.1002/cphy.c160048] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The term angiogenesis arose in the 18th century. Several studies over the next 100 years laid the groundwork for initial studies performed by the Folkman laboratory, which were at first met with some opposition. Once overcome, the angiogenesis field has flourished due to studies on tumor angiogenesis and various developmental models that can be genetically manipulated, including mice and zebrafish. In addition, new discoveries have been aided by the ability to isolate primary endothelial cells, which has allowed dissection of various steps within angiogenesis. This review will summarize the molecular events that control angiogenesis downstream of biochemical factors such as growth factors, cytokines, chemokines, hypoxia-inducible factors (HIFs), and lipids. These and other stimuli have been linked to regulation of junctional molecules and cell surface receptors. In addition, the contribution of cytoskeletal elements and regulatory proteins has revealed an intricate role for mobilization of actin, microtubules, and intermediate filaments in response to cues that activate the endothelium. Activating stimuli also affect various focal adhesion proteins, scaffold proteins, intracellular kinases, and second messengers. Finally, metalloproteinases, which facilitate matrix degradation and the formation of new blood vessels, are discussed, along with our knowledge of crosstalk between the various subclasses of these molecules throughout the text. Compr Physiol 8:153-235, 2018.
Collapse
Affiliation(s)
- Camille L Duran
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, Texas, USA
| | - David W Howell
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, Texas, USA
| | - Jui M Dave
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, Texas, USA
| | - Rebecca L Smith
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, Texas, USA
| | - Melanie E Torrie
- Department of Genetics, Development and Cell Biology, Iowa State University, Ames, Iowa, USA
| | - Jeffrey J Essner
- Department of Genetics, Development and Cell Biology, Iowa State University, Ames, Iowa, USA
| | - Kayla J Bayless
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, Texas, USA
| |
Collapse
|
24
|
Rangel L, Lospitao E, Ruiz-Sáenz A, Alonso MA, Correas I. Alternative polyadenylation in a family of paralogous EPB41 genes generates protein 4.1 diversity. RNA Biol 2016; 14:236-244. [PMID: 27981895 DOI: 10.1080/15476286.2016.1270003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Alternative polyadenylation (APA) is a step in mRNA 3'-end processing that contributes to the complexity of the transcriptome by generating isoforms that differ in either their coding sequence or their 3'-untranslated regions (UTRs). The EPB41 genes, EPB41, EPB41L2, EPB41L3 and EPB41L1, encode an impressively complex array of structural adaptor proteins (designated 4.1R, 4.1G, 4.1B and 4.1N, respectively) by using alternative transcriptional promoters and tissue-specific alternative pre-mRNA splicing. The great variety of 4.1 proteins mainly results from 5'-end and internal processing of the EPB41 pre-mRNAs. Thus, 4.1 proteins can vary in their N-terminal extensions but all contain a highly homologous C-terminal domain (CTD). Here we study a new group of EPB41-related mRNAs that originate by APA and lack the exons encoding the CTD characteristic of prototypical 4.1 proteins, thereby encoding a new type of 4.1 protein. For the EPB41 gene, this type of processing was observed in all 11 human tissues analyzed. Comparative genomic analysis of EPB41 indicates that APA is conserved in various mammals. In addition, we show that APA also functions for the EPB41L2, EPB41L3 and EPB41L1 genes, but in a more restricted manner in the case of the latter 2 than it does for the EPB41 and EPB41L2 genes. Our study shows alternative polyadenylation to be an additional mechanism for the generation of 4.1 protein diversity in the already complex EPB41-related genes. Understanding the diversity of EPB41 RNA processing is essential for a full appreciation of the many 4.1 proteins expressed in normal and pathological tissues.
Collapse
Affiliation(s)
- Laura Rangel
- a Departamento de Biología Molecular , Universidad Autónoma de Madrid (UAM), Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas (CSIC), Nicolás Cabrera , Cantoblanco, Madrid , Spain
| | - Eva Lospitao
- a Departamento de Biología Molecular , Universidad Autónoma de Madrid (UAM), Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas (CSIC), Nicolás Cabrera , Cantoblanco, Madrid , Spain
| | - Ana Ruiz-Sáenz
- a Departamento de Biología Molecular , Universidad Autónoma de Madrid (UAM), Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas (CSIC), Nicolás Cabrera , Cantoblanco, Madrid , Spain
| | - Miguel A Alonso
- a Departamento de Biología Molecular , Universidad Autónoma de Madrid (UAM), Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas (CSIC), Nicolás Cabrera , Cantoblanco, Madrid , Spain
| | - Isabel Correas
- a Departamento de Biología Molecular , Universidad Autónoma de Madrid (UAM), Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas (CSIC), Nicolás Cabrera , Cantoblanco, Madrid , Spain
| |
Collapse
|
25
|
4.1N is involved in a flotillin-1/β-catenin/Wnt pathway and suppresses cell proliferation and migration in non-small cell lung cancer cell lines. Tumour Biol 2016; 37:12713-12723. [DOI: 10.1007/s13277-016-5146-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Accepted: 07/11/2016] [Indexed: 01/14/2023] Open
|
26
|
Mudadu MA, Porto-Neto LR, Mokry FB, Tizioto PC, Oliveira PSN, Tullio RR, Nassu RT, Niciura SCM, Tholon P, Alencar MM, Higa RH, Rosa AN, Feijó GLD, Ferraz ALJ, Silva LOC, Medeiros SR, Lanna DP, Nascimento ML, Chaves AS, Souza ARDL, Packer IU, Torres RAA, Siqueira F, Mourão GB, Coutinho LL, Reverter A, Regitano LCA. Genomic structure and marker-derived gene networks for growth and meat quality traits of Brazilian Nelore beef cattle. BMC Genomics 2016; 17:235. [PMID: 26979536 PMCID: PMC4791965 DOI: 10.1186/s12864-016-2535-3] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Accepted: 02/25/2016] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Nelore is the major beef cattle breed in Brazil with more than 130 million heads. Genome-wide association studies (GWAS) are often used to associate markers and genomic regions to growth and meat quality traits that can be used to assist selection programs. An alternative methodology to traditional GWAS that involves the construction of gene network interactions, derived from results of several GWAS is the AWM (Association Weight Matrices)/PCIT (Partial Correlation and Information Theory). With the aim of evaluating the genetic architecture of Brazilian Nelore cattle, we used high-density SNP genotyping data (~770,000 SNP) from 780 Nelore animals comprising 34 half-sibling families derived from highly disseminated and unrelated sires from across Brazil. The AWM/PCIT methodology was employed to evaluate the genes that participate in a series of eight phenotypes related to growth and meat quality obtained from this Nelore sample. RESULTS Our results indicate a lack of structuring between the individuals studied since principal component analyses were not able to differentiate families by its sires or by its ancestral lineages. The application of the AWM/PCIT methodology revealed a trio of transcription factors (comprising VDR, LHX9 and ZEB1) which in combination connected 66 genes through 359 edges and whose biological functions were inspected, some revealing to participate in biological growth processes in literature searches. CONCLUSIONS The diversity of the Nelore sample studied is not high enough to differentiate among families neither by sires nor by using the available ancestral lineage information. The gene networks constructed from the AWM/PCIT methodology were a useful alternative in characterizing genes and gene networks that were allegedly influential in growth and meat quality traits in Nelore cattle.
Collapse
Affiliation(s)
- Maurício A Mudadu
- Embrapa Agricultural Informatics, Av. André Tosello, 209, Campinas, SP, Brazil. .,Embrapa Southeast Livestock, Rodovia Washington Luiz, Km 234, São Carlos, SP, Brazil.
| | - Laercio R Porto-Neto
- Commonwealth Scientific and Industrial Research Organization - Agriculture, 306 Carmody Road, Brisbane, QLD, Australia
| | - Fabiana B Mokry
- Department of Genetics and Evolution, Federal University of São Carlos, Rodovia Washington Luiz, Km 235, São Carlos, SP, Brazil
| | - Polyana C Tizioto
- Department of Genetics and Evolution, Federal University of São Carlos, Rodovia Washington Luiz, Km 235, São Carlos, SP, Brazil
| | - Priscila S N Oliveira
- Department of Genetics and Evolution, Federal University of São Carlos, Rodovia Washington Luiz, Km 235, São Carlos, SP, Brazil
| | - Rymer R Tullio
- Embrapa Southeast Livestock, Rodovia Washington Luiz, Km 234, São Carlos, SP, Brazil
| | - Renata T Nassu
- Embrapa Southeast Livestock, Rodovia Washington Luiz, Km 234, São Carlos, SP, Brazil
| | - Simone C M Niciura
- Embrapa Southeast Livestock, Rodovia Washington Luiz, Km 234, São Carlos, SP, Brazil
| | - Patrícia Tholon
- Embrapa Southeast Livestock, Rodovia Washington Luiz, Km 234, São Carlos, SP, Brazil
| | - Maurício M Alencar
- Embrapa Southeast Livestock, Rodovia Washington Luiz, Km 234, São Carlos, SP, Brazil
| | - Roberto H Higa
- Embrapa Agricultural Informatics, Av. André Tosello, 209, Campinas, SP, Brazil
| | - Antônio N Rosa
- Embrapa Beef Cattle, Av. Rádio Maia, 830, Campo Grande, MS, Brazil
| | - Gélson L D Feijó
- Embrapa Beef Cattle, Av. Rádio Maia, 830, Campo Grande, MS, Brazil
| | - André L J Ferraz
- State University of Mato Grosso do Sul, Rodovia Uems-Aquidauana km 12, Aquidauana, MS, Brazil
| | - Luiz O C Silva
- Embrapa Beef Cattle, Av. Rádio Maia, 830, Campo Grande, MS, Brazil
| | | | - Dante P Lanna
- Department of Animal Science, University of São Paulo, Av. Padua Dias, 11306, Piracicaba, SP, Brazil
| | - Michele L Nascimento
- Department of Animal Science, University of São Paulo, Av. Padua Dias, 11306, Piracicaba, SP, Brazil
| | - Amália S Chaves
- Department of Animal Science, University of São Paulo, Av. Padua Dias, 11306, Piracicaba, SP, Brazil
| | - Andrea R D L Souza
- Faculdade de Medicina Veterinaria e Zootecnia, Federal University of Mato Grosso do Sul, Av. Senador Filinto Müller, 2443, Campo Grande, MS, Brazil
| | - Irineu U Packer
- Department of Animal Science, University of São Paulo, Av. Padua Dias, 11306, Piracicaba, SP, Brazil
| | | | - Fabiane Siqueira
- Embrapa Beef Cattle, Av. Rádio Maia, 830, Campo Grande, MS, Brazil
| | - Gerson B Mourão
- Department of Animal Science, University of São Paulo, Av. Padua Dias, 11306, Piracicaba, SP, Brazil
| | - Luiz L Coutinho
- Department of Animal Science, University of São Paulo, Av. Padua Dias, 11306, Piracicaba, SP, Brazil
| | - Antonio Reverter
- Commonwealth Scientific and Industrial Research Organization - Agriculture, 306 Carmody Road, Brisbane, QLD, Australia
| | - Luciana C A Regitano
- Embrapa Southeast Livestock, Rodovia Washington Luiz, Km 234, São Carlos, SP, Brazil
| |
Collapse
|
27
|
Zhang L, Hu A, Li M, Zhang H, Ren C, An X, Liu C. 4.1N suppresses hypoxia-induced epithelial-mesenchymal transition in epithelial ovarian cancer cells. Mol Med Rep 2015; 13:837-44. [PMID: 26648170 DOI: 10.3892/mmr.2015.4634] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2015] [Accepted: 10/29/2015] [Indexed: 11/06/2022] Open
Abstract
Protein 4.1N (4.1N) is a member of the protein 4.1 family and is essential for the regulation of cell adhesion, motility and signaling. Previous studies have suggested that 4.1N may serve a tumor suppressor role. However, the molecular mechanisms remain unclear. In the current study, the role of 4.1N in the downregulation of hypoxia‑induced factor 1α (HIF‑1α) under hypoxic conditions and therefore the suppression of hypoxia induced epithelial‑mesenchymal transition (EMT) was investigated. The data were obtained from overexpressed and knockdown 4.1N epithelial ovarian cancer (EOC) cell lines. It was identified that 4.1N was capable of regulating the sub‑cellular localization and expression levels of HIF‑1α, by which 4.1N served a dominant role in the suppression of hypoxia‑induced EMT and associated genes. Collectively, the data of the current study identified 4.1N as an inhibitor of hypoxia‑induced tumor progression in EOC cells and highlighted its potential role in EOC therapy.
Collapse
Affiliation(s)
- Letian Zhang
- Department of Pathology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, P.R. China
| | - Ajin Hu
- Department of Pathology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, P.R. China
| | - Mengrui Li
- Department of Pathology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, P.R. China
| | - Hongquan Zhang
- Department of Histology and Embryology, Peking University Health Science Center, Beijing 100191, P.R. China
| | - Caixia Ren
- Department of Histology and Embryology, Peking University Health Science Center, Beijing 100191, P.R. China
| | - Xiuli An
- College of Life Science, Zhengzhou University, Zhengzhou, Henan 450051, P.R. China
| | - Congrong Liu
- Department of Pathology, School of Basic Medical Sciences, Third Hospital, Peking University Health Science Center, Beijing 100191, P.R. China
| |
Collapse
|
28
|
Wang Z, Zhang J, Ye M, Zhu M, Zhang B, Roy M, Liu J, An X. Tumor suppressor role of protein 4.1B/DAL-1. Cell Mol Life Sci 2014; 71:4815-30. [PMID: 25183197 PMCID: PMC11113756 DOI: 10.1007/s00018-014-1707-z] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2014] [Revised: 07/21/2014] [Accepted: 08/15/2014] [Indexed: 12/14/2022]
Abstract
Protein 4.1B/DAL-1 is a membrane skeletal protein that belongs to the protein 4.1 family. Protein 4.1B/DAL-1 is localized to sites of cell-cell contact and functions as an adapter protein, linking the plasma membrane to the cytoskeleton or associated cytoplasmic signaling effectors and facilitating their activities in various pathways. Protein 4.1B/DAL-1 is involved in various cytoskeleton-associated processes, such as cell motility and adhesion. Moreover, protein 4.1B/DAL-1 also plays a regulatory role in cell growth, differentiation, and the establishment of epithelial-like cell structures. Protein 4.1B/DAL-1 is normally expressed in multiple human tissues, but loss of its expression or prominent down-regulation of its expression is frequently observed in corresponding tumor tissues and tumor cell lines, suggesting that protein 4.1B/DAL-1 is involved in the molecular pathogenesis of these tumors and acts as a potential tumor suppressor. This review will focus on the structure of protein 4.1B/DAL-1, 4.1B/DAL-1-interacting molecules, 4.1B/DAL-1 inactivation and tumor progression, and anti-tumor activity of the 4.1B/DAL-1.
Collapse
Affiliation(s)
- Zi Wang
- Molecular Biology Research Center, School of Life Sciences, Central South University, 110 Xiangya Road, Changsha, 410078 China
| | - Ji Zhang
- Molecular Biology Research Center, School of Life Sciences, Central South University, 110 Xiangya Road, Changsha, 410078 China
- Department of Hematology, The First Affiliated Hospital, University of South China, Hengyang, 421001 China
| | - Mao Ye
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, Hunan University, Changsha, 410082 China
| | - Min Zhu
- Molecular Biology Research Center, School of Life Sciences, Central South University, 110 Xiangya Road, Changsha, 410078 China
| | - Bin Zhang
- Department of Histology and Embryology, Xiangya School Medicine, Central South University, Changsha, 410083 China
| | - Mridul Roy
- Molecular Biology Research Center, School of Life Sciences, Central South University, 110 Xiangya Road, Changsha, 410078 China
| | - Jing Liu
- Molecular Biology Research Center, School of Life Sciences, Central South University, 110 Xiangya Road, Changsha, 410078 China
- State Key Laboratory of Medical Genetics, Central South University, 110 Xiangya Road, Changsha, 410078 China
| | - Xiuli An
- Laboratory of Membrane Biology, New York Blood Center, 310 E 67th Street, New York, 10065 USA
| |
Collapse
|
29
|
Zhan Y, Chadee DN. Inhibition of Cdc42-mediated activation of mixed lineage kinase 3 by the tumor suppressor protein merlin. Small GTPases 2014; 1:183-186. [PMID: 21686274 DOI: 10.4161/sgtp.1.3.14935] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2010] [Revised: 12/28/2010] [Accepted: 01/23/2011] [Indexed: 11/19/2022] Open
Abstract
Mammalian mitogen-activated protein kinase (MAPK) signaling pathways respond to diverse extracellular signals and coordinate a range of cellular responses. Mixed lineage kinase 3 (MLK3) is a member of the mixed lineage kinase family of MAPK kinase kinases (MAP3Ks) that functions to regulate multiple MAPK signaling pathways. Activated forms of the Rho GTP ases, Rac and Cdc42, interact with MLK3 through the Cdc42/Rac-interactive binding (CRIB) motif and promote MLK3 catalytic activity. Our recent findings demonstrate that merlin, the product of the neurofibromatosis type 2 (NF2) tumor suppressor gene, is a physiological inhibitor of MLK3. Our results suggest that merlin inhibits MLK3 activity by blocking the Cdc42-MLK3 interaction. In this commentary, the effect of merlin on Cdc42-mediated activation of MLK3 and MAPK signaling will be discussed.
Collapse
Affiliation(s)
- Yu Zhan
- Department of Biological Sciences; University of Toledo; Toledo, OH USA
| | | |
Collapse
|
30
|
A functional and protein-protein interaction analysis of neuroepithelial cell transforming gene 1 in hepatocellular carcinoma. Tumour Biol 2014; 35:11219-27. [DOI: 10.1007/s13277-014-2454-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2014] [Accepted: 08/05/2014] [Indexed: 01/26/2023] Open
|
31
|
Bosanquet DC, Ye L, Harding KG, Jiang WG. FERM family proteins and their importance in cellular movements and wound healing (review). Int J Mol Med 2014; 34:3-12. [PMID: 24820650 DOI: 10.3892/ijmm.2014.1775] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2014] [Accepted: 03/10/2014] [Indexed: 11/06/2022] Open
Abstract
Motility is a requirement for a number of biological processes, including embryonic development, neuronal development, immune responses, cancer progression and wound healing. Specific to wound healing is the migration of endothelial cells, fibroblasts and other key cellular players into the wound space. Aberrations in wound healing can result in either chronic wounds or abnormally healed wounds. The protein 4.1R, ezrin, radixin, moesin (FERM) superfamily consists of over 40 proteins all containing a three lobed N-terminal FERM domain which binds a variety of cell-membrane associated proteins and lipids. The C-terminal ends of these proteins typically contain an actin-binding domain (ABD). These proteins therefore mediate the linkage between the cell membrane and the actin cytoskeleton, and are involved in cellular movements and migration. Certain FERM proteins have been shown to promote cancer metastasis via this very mechanism. Herein we review the effects of a number of FERM proteins on wound healing and cancer. We show how these proteins typically aid wound healing through their effects on increasing cellular migration and movements, but also typically promote metastasis in cancer. We conclude that FERM proteins play important roles in cellular migration, with markedly different outcomes in the context of cancer and wound healing.
Collapse
Affiliation(s)
- David C Bosanquet
- Departments of Surgery and Wound Healing, Cardiff University School of Medicine, Heath Park, Cardiff CF14 4XW, UK
| | - Lin Ye
- Departments of Surgery and Wound Healing, Cardiff University School of Medicine, Heath Park, Cardiff CF14 4XW, UK
| | - Keith G Harding
- Departments of Surgery and Wound Healing, Cardiff University School of Medicine, Heath Park, Cardiff CF14 4XW, UK
| | - Wen G Jiang
- Departments of Surgery and Wound Healing, Cardiff University School of Medicine, Heath Park, Cardiff CF14 4XW, UK
| |
Collapse
|
32
|
ICAM-2 confers a non-metastatic phenotype in neuroblastoma cells by interaction with α-actinin. Oncogene 2014; 34:1553-62. [PMID: 24704826 DOI: 10.1038/onc.2014.87] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2013] [Revised: 01/29/2014] [Accepted: 02/24/2014] [Indexed: 12/16/2022]
Abstract
Progressive metastatic disease is a major cause of mortality for patients diagnosed with multiple types of solid tumors. One of the long-term goals of our laboratory is to identify molecular interactions that regulate metastasis, as a basis for developing agents that inhibit this process. Toward this goal, we recently demonstrated that intercellular adhesion molecule-2 (ICAM-2) converted neuroblastoma (NB) cells from a metastatic to a non-metastatic phenotype, a previously unknown function for ICAM-2. Interestingly, ICAM-2 suppressed metastatic but not tumorigenic potential in preclinical models, supporting a novel mechanism of regulating metastasis. We hypothesized that the effects of ICAM-2 on NB cell phenotype depend on the interaction of ICAM-2 with the cytoskeletal linker protein α-actinin. The goal of the study presented here was to evaluate the impact of α-actinin binding to ICAM-2 on the phenotype of NB tumor cells. We used in silico approaches to examine the likelihood that the cytoplasmic domain of ICAM-2 binds directly to α-actinin. We then expressed variants of ICAM-2 with mutated α-actinin-binding domains, and compared the impact of ICAM-2 and each variant on NB cell adhesion, migration, anchorage-independent growth, co-precipitation with α-actinin and production of localized and disseminated tumors in vivo. The in vitro and in vivo characteristics of cells expressing ICAM-2 variants with modified α-actinin-binding domains differed from cells expressing ICAM-2 wild type (WT) and also from cells that expressed no detectable ICAM-2. Like the WT protein, ICAM-2 variants inhibited cell adhesion, migration and colony growth in vitro. However, unlike the WT protein, ICAM-2 variants did not completely suppress development of disseminated NB tumors in vivo. The data suggest the presence of α-actinin-dependent and α-actinin-independent mechanisms, and indicate that the interaction of ICAM-2 with α-actinin is critical to conferring an ICAM-2-mediated non-metastatic phenotype in NB cells.
Collapse
|
33
|
Baines AJ, Lu HC, Bennett PM. The Protein 4.1 family: hub proteins in animals for organizing membrane proteins. BIOCHIMICA ET BIOPHYSICA ACTA 2014; 1838:605-19. [PMID: 23747363 DOI: 10.1016/j.bbamem.2013.05.030] [Citation(s) in RCA: 101] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2013] [Revised: 05/22/2013] [Accepted: 05/28/2013] [Indexed: 01/10/2023]
Abstract
Proteins of the 4.1 family are characteristic of eumetazoan organisms. Invertebrates contain single 4.1 genes and the Drosophila model suggests that 4.1 is essential for animal life. Vertebrates have four paralogues, known as 4.1R, 4.1N, 4.1G and 4.1B, which are additionally duplicated in the ray-finned fish. Protein 4.1R was the first to be discovered: it is a major mammalian erythrocyte cytoskeletal protein, essential to the mechanochemical properties of red cell membranes because it promotes the interaction between spectrin and actin in the membrane cytoskeleton. 4.1R also binds certain phospholipids and is required for the stable cell surface accumulation of a number of erythrocyte transmembrane proteins that span multiple functional classes; these include cell adhesion molecules, transporters and a chemokine receptor. The vertebrate 4.1 proteins are expressed in most tissues, and they are required for the correct cell surface accumulation of a very wide variety of membrane proteins including G-Protein coupled receptors, voltage-gated and ligand-gated channels, as well as the classes identified in erythrocytes. Indeed, such large numbers of protein interactions have been mapped for mammalian 4.1 proteins, most especially 4.1R, that it appears that they can act as hubs for membrane protein organization. The range of critical interactions of 4.1 proteins is reflected in disease relationships that include hereditary anaemias, tumour suppression, control of heartbeat and nervous system function. The 4.1 proteins are defined by their domain structure: apart from the spectrin/actin-binding domain they have FERM and FERM-adjacent domains and a unique C-terminal domain. Both the FERM and C-terminal domains can bind transmembrane proteins, thus they have the potential to be cross-linkers for membrane proteins. The activity of the FERM domain is subject to multiple modes of regulation via binding of regulatory ligands, phosphorylation of the FERM associated domain and differential mRNA splicing. Finally, the spectrum of interactions of the 4.1 proteins overlaps with that of another membrane-cytoskeleton linker, ankyrin. Both ankyrin and 4.1 link to the actin cytoskeleton via spectrin, and we hypothesize that differential regulation of 4.1 proteins and ankyrins allows highly selective control of cell surface protein accumulation and, hence, function. This article is part of a Special Issue entitled: Reciprocal influences between cell cytoskeleton and membrane channels, receptors and transporters. Guest Editor: Jean Claude Hervé
Collapse
Affiliation(s)
| | - Hui-Chun Lu
- Randall Division of Cell and Molecular Biophysics, King's College London, UK
| | - Pauline M Bennett
- Randall Division of Cell and Molecular Biophysics, King's College London, UK.
| |
Collapse
|
34
|
Wang J, Song J, An C, Dong W, Zhang J, Yin C, Hale J, Baines AJ, Mohandas N, An X. A 130-kDa protein 4.1B regulates cell adhesion, spreading, and migration of mouse embryo fibroblasts by influencing actin cytoskeleton organization. J Biol Chem 2013; 289:5925-37. [PMID: 24381168 DOI: 10.1074/jbc.m113.516617] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Protein 4.1B is a member of protein 4.1 family, adaptor proteins at the interface of membranes and the cytoskeleton. It is expressed in most mammalian tissues and is known to be required in formation of nervous and cardiac systems; it is also a tumor suppressor with a role in metastasis. Here, we explore functions of 4.1B using primary mouse embryonic fibroblasts (MEF) derived from wild type and 4.1B knock-out mice. MEF cells express two 4.1B isoforms: 130 and 60-kDa. 130-kDa 4.1B was absent from 4.1B knock-out MEF cells, but 60-kDa 4.1B remained, suggesting incomplete knock-out. Although the 130-kDa isoform was predominantly located at the plasma membrane, the 60-kDa isoform was enriched in nuclei. 130-kDa-deficient 4.1B MEF cells exhibited impaired cell adhesion, spreading, and migration; they also failed to form actin stress fibers. Impaired cell spreading and stress fiber formation were rescued by re-expression of the 130-kDa 4.1B but not the 60-kDa 4.1B. Our findings document novel, isoform-selective roles for 130-kDa 4.1B in adhesion, spreading, and migration of MEF cells by affecting actin organization, giving new insight into 4.1B functions in normal tissues as well as its role in cancer.
Collapse
Affiliation(s)
- Jie Wang
- From the Department of Biophysics, Peking University Health Science Center, Xueyuan Road, Haidian District, Beijing 100191, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Xi C, Ren C, Hu A, Lin J, Yao Q, Wang Y, Gao Z, An X, Liu C. Defective expression of Protein 4.1N is correlated to tumor progression, aggressive behaviors and chemotherapy resistance in epithelial ovarian cancer. Gynecol Oncol 2013; 131:764-71. [PMID: 23994105 DOI: 10.1016/j.ygyno.2013.08.015] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2013] [Revised: 07/16/2013] [Accepted: 08/08/2013] [Indexed: 12/17/2022]
Abstract
OBJECTIVE Protein 4.1N (4.1N) is a member of the Protein 4.1 family that is involved in cellular processes such as cell adhesion, migration and signaling. In this study, we evaluated the expression of 4.1N protein and its potential roles in epithelial ovarian cancer (EOC) tumorigenesis and progression. METHODS 4.1N protein expression was investigated in a total of 280 samples including 74 normal tissues, 35 benign, 30 borderline and 141 malignant epithelial ovarian tumors by immunohistochemistry. Correlation between 4.1N expression levels and clinicopathologic features was statistically analyzed. The expression of 4.1N in EOC cell lines was examined by western blotting. RESULTS Immunohistochemistry analysis revealed that, although there was no loss of 4.1N expression in normal tissues and benign tumors, absence of Protein 4.1N was significantly more common in EOCs (44.0%) than in borderline tumors (3.3%) (p<0.001). Furthermore, loss or decreased expression of 4.1N protein expression was correlated with malignant potential of the tumors (14.3% in benign tumors, 56.7% in borderline tumors and 92.9% in malignancy) (p<0.001). In EOC samples, loss of 4.1N protein was significantly associated with advanced-stage (p=0.004), ascites (p=0.009), omental metastasis (p=0.018), suboptimal debulking (p=0.024), poorly histological differentiation (p=0.009), high-grade serous carcinoma (p=0.001), short progression-free-survival (p=0.018) and poor chemosensitivity to first-line chemotherapy (p=0.029). Moreover, western blotting analysis revealed that expression of 4.1N protein was lost in 4/8 (50%) EOC cell lines. CONCLUSIONS 4.1N protein expression level was significantly decreased during malignant transformation of epithelial ovarian tumors and that loss of 4.1N expression was closely correlated to poorly differentiated and biologically aggressive EOCs.
Collapse
Affiliation(s)
- Chenguang Xi
- Department of Pathology, Peking University Health Science Center, Beijing 100191, China
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Abstract
Our understanding of the FERM (4.1/ezrin/radixin/moesin) protein family has been rapidly expanding in the last few years, with the result that many new physiological functions have been ascribed to these biochemically unique proteins. In the present review, we will discuss a number of new FRMD (FERM domain)-containing proteins that were initially discovered from genome sequencing but are now being established through biochemical and genetic studies to be involved both in normal cellular processes, but are also associated with a variety of human diseases.
Collapse
|
37
|
Wang Z, He ML, Zhao JM, Qing HH, Wu Y. Meta-analysis of Associations of the Ezrin Gene with Human Osteosarcoma Response to Chemotherapy and Prognosis. Asian Pac J Cancer Prev 2013; 14:2753-8. [DOI: 10.7314/apjcp.2013.14.5.2753] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
|
38
|
Sugiyama H, Mizutani K, Kurita S, Okimoto N, Shimono Y, Takai Y. Interaction of Necl-4/CADM4 with ErbB3 and integrin α6 β4 and inhibition of ErbB2/ErbB3 signaling and hemidesmosome disassembly. Genes Cells 2013; 18:519-28. [PMID: 23611113 DOI: 10.1111/gtc.12056] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2012] [Accepted: 03/05/2012] [Indexed: 12/14/2022]
Abstract
Nectin-like molecule 4 (Necl-4)/CADM4, a transmembrane cell-cell adhesion molecule with three Ig-like domains, was shown to serve as a tumor suppressor, but its mode of action has not been elucidated. In this study, we showed that Necl-4 interacted in cis with ErbB3 through their extracellular regions, recruited PTPN13 and inhibited the heregulin-induced activation of the ErbB2/ErbB3 signaling. In addition, we extended our previous finding that Necl-4 interacts in cis with integrin α6 β4 through their extracellular regions and found that Necl-4 inhibited the phorbol ester-induced disassembly of hemidesmosomes. These results indicate that Necl-4 serves as a tumor suppressor by inhibiting the ErbB2/ErbB3 signaling and hemidesmosome disassembly.
Collapse
Affiliation(s)
- Hirokazu Sugiyama
- Division of Molecular and Cellular Biology, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan
| | | | | | | | | | | |
Collapse
|
39
|
Bosanquet DC, Ye L, Harding KG, Jiang WG. Expressed in high metastatic cells (Ehm2) is a positive regulator of keratinocyte adhesion and motility: The implication for wound healing. J Dermatol Sci 2013; 71:115-21. [PMID: 23664528 DOI: 10.1016/j.jdermsci.2013.04.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2012] [Revised: 03/17/2013] [Accepted: 04/04/2013] [Indexed: 12/18/2022]
Abstract
BACKGROUND Multiple factors have been shown to delay dermal wound healing. These resultant wounds pose a significant problem in terms of morbidity and healthcare spend. Recently, an increasing volume of research has focused on the molecular perturbations underlying non-healing wounds. OBJECTIVES This study investigates the effect of a novel cancer promoter, Ehm2, in wound healing. Ehm2 belongs to the FERM family of proteins, known to be involved in membrane-cytoskeletal interactions, and has been shown to promote cancer metastasis in melanoma, prostate cancer and breast cancer. METHODS Ehm2 mRNA levels were analysed using qRT-PCR, standardised to GAPDH, from either acute or chronic wounds, and normal skin. IHC analysis was also undertaken from wound edge biopsies. An anti-Ehm2 transgene was created and transfected into the HaCaT cell line. The effect of Ehm2 knockdown on migration, adhesion, growth, cell cycle progression and apoptosis was analysed using standard laboratory methods. Western Blot analysis was used to investigate potential downstream protein interactions. RESULTS Ehm2 is expressed nearly three times higher in acute wound tissues, compared to chronic wound tissues. Increased Ehm2 expression is found in wounds undergoing healing, especially at the leading wound edge. In vitro, Ehm2 knockdown reduces cellular adhesion, migration and motility, without affecting growth, cell cycle and apoptosis. Finally, Ehm2 knockdown results in reduced NWasp protein expression. CONCLUSION These results suggest Ehm2 may be an important player in the wound healing process, and show that Ehm2 knockdown downregulates the expression of NWasp, through which it may have its effect on cellular migration.
Collapse
Affiliation(s)
- David C Bosanquet
- Departments of Surgery and Wound Healing, Cardiff University School of Medicine, Heath Park, Cardiff CF14 4XW, UK.
| | | | | | | |
Collapse
|
40
|
Einheber S, Meng X, Rubin M, Lam I, Mohandas N, An X, Shrager P, Kissil J, Maurel P, Salzer JL. The 4.1B cytoskeletal protein regulates the domain organization and sheath thickness of myelinated axons. Glia 2012; 61:240-53. [PMID: 23109359 DOI: 10.1002/glia.22430] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2012] [Accepted: 09/10/2012] [Indexed: 12/13/2022]
Abstract
Myelinated axons are organized into specialized domains critical to their function in saltatory conduction, i.e., nodes, paranodes, juxtaparanodes, and internodes. Here, we describe the distribution and role of the 4.1B protein in this organization. 4.1B is expressed by neurons, and at lower levels by Schwann cells, which also robustly express 4.1G. Immunofluorescence and immuno-EM demonstrates 4.1B is expressed subjacent to the axon membrane in all domains except the nodes. Mice deficient in 4.1B have preserved paranodes, based on marker staining and EM in contrast to the juxtaparanodes, which are substantially affected in both the PNS and CNS. The juxtaparanodal defect is evident in developing and adult nerves and is neuron-autonomous based on myelinating cocultures in which wt Schwann cells were grown with 4.1B-deficient neurons. Despite the juxtaparanodal defect, nerve conduction velocity is unaffected. Preservation of paranodal markers in 4.1B deficient mice is associated with, but not dependent on an increase of 4.1R at the axonal paranodes. Loss of 4.1B in the axon is also associated with reduced levels of the internodal proteins, Necl-1 and Necl-2, and of alpha-2 spectrin. Mutant nerves are modestly hypermyelinated and have increased numbers of Schmidt-Lanterman incisures, increased expression of 4.1G, and express a residual, truncated isoform of 4.1B. These results demonstrate that 4.1B is a key cytoskeletal scaffold for axonal adhesion molecules expressed in the juxtaparanodal and internodal domains that unexpectedly regulates myelin sheath thickness.
Collapse
Affiliation(s)
- Steven Einheber
- School of Health Sciences, Hunter College, City University of New York, New York, New York, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Jung Y, McCarty JH. Band 4.1 proteins regulate integrin-dependent cell spreading. Biochem Biophys Res Commun 2012; 426:578-84. [PMID: 22982319 DOI: 10.1016/j.bbrc.2012.08.129] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2012] [Accepted: 08/25/2012] [Indexed: 11/30/2022]
Abstract
Integrins link the extracellular matrix (ECM) to the cytoskeleton to control cell behaviors including adhesion, spreading and migration. Band 4.1 proteins contain 4.1, ezrin, radixin, moesin (FERM) domains that likely mediate signaling events and cytoskeletal reorganization via integrins. However, the mechanisms by which Band 4.1 proteins and integrins are functionally interconnected remain enigmatic. Here we have investigated roles for Band 4.1 proteins in integrin-mediated cell spreading using primary astrocytes as a model system. We demonstrate that Proteins 4.1B and 4.1G show dynamic patterns of sub-cellular localization in astrocytes spreading on fibronectin. During early stages of cell spreading Proteins 4.1B and 4.1G are enriched in ECM adhesion sites but become more diffusely localized at later stages of spreading. Combinatorial inactivation of Protein 4.1B and 4.1G expression leads to impaired astrocyte spreading. Furthermore, in exogenous expression systems we show that the isolated Protein 4.1 FERM domain significantly enhances integrin-mediated cell spreading. Protein 4.1B is dispensable for reactive astrogliosis in experimental models of cortical injury, likely due to functional compensation by related Protein 4.1 family members. Collectively, these findings reveal that Band 4.1 proteins are important intracellular components for integrin-mediated cell spreading.
Collapse
Affiliation(s)
- Youngsin Jung
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston TX 77030, United States
| | | |
Collapse
|
42
|
Shingleton AW. The regulation of organ size in Drosophila: physiology, plasticity, patterning and physical force. Organogenesis 2012; 6:76-87. [PMID: 20885854 DOI: 10.4161/org.6.2.10375] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2009] [Revised: 10/16/2009] [Accepted: 10/20/2009] [Indexed: 12/19/2022] Open
Abstract
The correct regulation of organ size is a fundamental developmental process, the failure of which can compromise organ function and organismal integrity. Consequently, the mechanisms that regulate organ size have been subject to intense research. This research has highlighted four classes of mechanism that are involved in organ size regulation: physiology, plasticity, patterning and physical force. Nevertheless, how these mechanisms are integrated and converge on the cellular process that regulate organ growth is unknown. One group of animals where this integration is beginning to be achieved is in the insects. Here, I review the different mechanisms that regulate organ size in insects, and describe our current understanding of how these mechanisms interact. The genes and hormones involved are remarkably conserved in all animals, so these studies in insects provide a precedent for future research on organ size regulation in mammals.
Collapse
|
43
|
Ji Z, Shi X, Liu X, Shi Y, Zhou Q, Liu X, Li L, Ji X, Gao Y, Qi Y, Kang Q. The membrane-cytoskeletal protein 4.1N is involved in the process of cell adhesion, migration and invasion of breast cancer cells. Exp Ther Med 2012; 4:736-740. [PMID: 23170136 PMCID: PMC3501401 DOI: 10.3892/etm.2012.653] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2012] [Accepted: 07/11/2012] [Indexed: 11/06/2022] Open
Abstract
Protein 4.1N belongs to the protein 4.1 superfamily that links transmembrane proteins to the actin cytoskeleton. Recent evidence has shown that protein 4.1 is important in tumor suppression. However, the functions of 4.1N in the metastasis of breast cancer are largely unknown. In the present study, MCF-7, T-47D and MDA-MB-231 breast cancer cell lines with various metastatic abilities were employed. Protein 4.1N was found to be expressed in poorly metastatic MCF-7 and middle metastatic T-47D cell lines, and was predominantly associated with cell-cell junctions. However, no 4.1N expression was detected in the highly metastatic MDA-MB-231 cells. Moreover, re-expression of 4.1N in MDA-MB-231 cells inhibited cell adhesion, migration and invasion. The results suggest that protein 4.1N is a negative regulator of cell metastasis in breast cancer.
Collapse
Affiliation(s)
- Zhenyu Ji
- Department of Bioengineering, Zhengzhou University, Zhengzhou 450001; ; Henan Academy of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450052
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Wang T, Pei X, Zhan J, Hu J, Yu Y, Zhang H. FERM-containing protein FRMD5 is a p120-catenin interacting protein that regulates tumor progression. FEBS Lett 2012; 586:3044-50. [PMID: 22846708 DOI: 10.1016/j.febslet.2012.07.019] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2012] [Revised: 07/03/2012] [Accepted: 07/06/2012] [Indexed: 11/26/2022]
Abstract
FERM family proteins have been known to play an important role in tumor progression. FERM-domain containing protein 5 (FRMD5), a novel putative cytoskeletal protein, is an unknown function protein. Here, we reported that FRMD5 localized at the cell adherens junction and formed a molecular complex with p120-catenin through its C-terminal region. Functionally, we found that knockdown of endogenous FRMD5 promotes lung cancer cell migration and invasion in vitro as well as tumor growth in vivo, suggesting a tumor suppressive effect. These findings indicated that FRMD5 may play a role in p120-catenin-based cell-cell contact and is involved in the regulation of tumor progression.
Collapse
Affiliation(s)
- Tao Wang
- Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Peking University Health Science Center, Beijing, China
| | | | | | | | | | | |
Collapse
|
45
|
Loss of expression of the differentially expressed in adenocarcinoma of the lung (DAL-1) protein is associated with metastasis of non-small cell lung carcinoma cells. Tumour Biol 2012; 33:1915-25. [PMID: 22782504 DOI: 10.1007/s13277-012-0452-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2012] [Accepted: 06/26/2012] [Indexed: 12/31/2022] Open
Abstract
The differentially expressed in adenocarcinoma of the lung-1 (DAL-1) protein is a member of the membrane-associated cytoskeleton protein 4.1 family. This protein was previously found to be downregulated or lost in more than half of primary non-small cell lung cancers (NSCLC). In this study, the relationship between DAL-1 expression and NSCLC metastasis was examined. DAL-1 mRNA and protein levels were measured in NSCLC cell lines and in tumor cells isolated from the pleural fluid of NSCLC patients clinically diagnosed with distant metastases to the bone or brain. The results revealed that DAL-1 expression was observed in two (GLC-82 and NCI-H460) out of seven metastatic NSCLC cell lines examined. DAL-1 expression was not observed in the cells isolated from the pleural fluid in nine out of ten patients. Overexpression of DAL-1 in A549 cells, a cell line lacking endogenous DAL-1, inhibited cell migration and invasion by approximately 38 and 48 %, respectively. In contrast, DAL-1 knockdown in NCI-H460 cells enhanced the migration and invasion potential of this cell line 4.6- and 3-fold, respectively. Furthermore, DAL-1 promoter methylation was observed in six of nine pleural fluid NSCLC cell isolates and in two cell lines (A549 and H1299), as evidenced by a lack of endogenous DAL-1. Demethylation in A549 cells successfully restored DAL-1 mRNA and protein expression levels, resulting in a parallel remarkable inhibition of migration and invasion. These results indicated that DAL-1 was pivotal in triggering NSCLC migration and invasion and that loss of DAL-1 expression was due to the epigenetic methylation.
Collapse
|
46
|
The Role of Hyaluronic Acid in Atherosclerosis and Intimal Hyperplasia. J Surg Res 2012; 173:e63-72. [DOI: 10.1016/j.jss.2011.09.025] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2011] [Revised: 08/19/2011] [Accepted: 09/14/2011] [Indexed: 01/27/2023]
|
47
|
Jost S, Gutmann DH. Neurofibromatosis and other genetic syndromes. HANDBOOK OF CLINICAL NEUROLOGY 2012; 105:569-82. [PMID: 22230519 DOI: 10.1016/b978-0-444-53502-3.00009-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- Sarah Jost
- Department of Nedurology, Washington University School of Medicine, St. Louis, MO, USA
| | | |
Collapse
|
48
|
Sluchanko NN, Gusev NB. 14-3-3 proteins and regulation of cytoskeleton. BIOCHEMISTRY (MOSCOW) 2011; 75:1528-46. [PMID: 21417993 DOI: 10.1134/s0006297910130031] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The proteins of the 14-3-3 family are universal adapters participating in multiple processes running in the cell. We describe the structure, isoform composition, and distribution of 14-3-3 proteins in different tissues. Different elements of 14-3-3 structure important for dimer formation and recognition of protein targets are analyzed in detail. Special attention is paid to analysis of posttranslational modifications playing important roles in regulation of 14-3-3 function. The data of the literature concerning participation of 14-3-3 in regulation of intercellular contacts and different elements of cytoskeleton formed by microfilaments are analyzed. We also describe participation of 14-3-3 in regulation of small G-proteins and protein kinases important for proper functioning of cytoskeleton. The data on the interaction of 14-3-3 with different components of microtubules are presented, and the probable role of 14-3-3 in developing of certain neurodegenerative diseases is discussed. The data of the literature concerning the role of 14-3-3 in formation and normal functioning of intermediate filaments are also reviewed. It is concluded that due to its adapter properties 14-3-3 plays an important role in cytoskeleton regulation. The cytoskeletal proteins that are abundant in the cell might compete with the other protein targets of 14-3-3 and therefore can indirectly regulate many intracellular processes that are dependent on 14-3-3.
Collapse
Affiliation(s)
- N N Sluchanko
- Department of Biochemistry, Faculty of Biology, Lomonosov Moscow State University, Russia
| | | |
Collapse
|
49
|
The cytoskeletal adaptor protein band 4.1B is required for the maintenance of paranodal axoglial septate junctions in myelinated axons. J Neurosci 2011; 31:8013-24. [PMID: 21632923 DOI: 10.1523/jneurosci.1015-11.2011] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Precise targeting and maintenance of axonal domains in myelinated axons is essential for saltatory conduction. Caspr and Caspr2, which localize at paranodal and juxtaparanodal domains, contain binding sites for the cytoskeletal adaptor protein 4.1B. The exact role of 4.1B in the organization and maintenance of axonal domains is still not clear. Here, we report the generation and characterization of 4.1B-null mice. We show that loss of 4.1B in the PNS results in mislocalization of Caspr at paranodes and destabilization of paranodal axoglial septate junctions (AGSJs) as early as postnatal day 30. In the CNS, Caspr localization is progressively disrupted and ultrastructural analysis showed paranodal regions that were completely devoid of AGSJs, with axolemma separated from the myelin loops, and loops coming off the axolemma. Most importantly, our phenotypic analysis of previously generated 4.1B mutants, used in the study by Horresh et al. (2010), showed that Caspr localization was not affected in the PNS, even after 1 year; and 4.1R was neither expressed, nor enriched at the paranodes. Furthermore, ultrastructural analysis of these 4.1B mutants showed destabilization of CNS AGSJs at ∼ 1 year. We also discovered that the 4.1B locus is differentially expressed in the PNS and CNS, and generates multiple splice isoforms in the PNS, suggesting 4.1B may function differently in the PNS versus CNS. Together, our studies provide direct evidence that 4.1B plays a pivotal role in interactions between the paranodal AGSJs and axonal cytoskeleton, and that 4.1B is critically required for long-term maintenance of axonal domains in myelinated axons.
Collapse
|
50
|
Nagata M, Sakurai-Yageta M, Yamada D, Goto A, Ito A, Fukuhara H, Kume H, Morikawa T, Fukayama M, Homma Y, Murakami Y. Aberrations of a cell adhesion molecule CADM4 in renal clear cell carcinoma. Int J Cancer 2011; 130:1329-37. [DOI: 10.1002/ijc.26160] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2010] [Accepted: 01/27/2011] [Indexed: 11/07/2022]
|