1
|
Wu D, Zhu J, Yang F, Li R, Liu L, Liu D, Liu C, Qu X, Liu H, Ji M, Qin X, Hua L, Xiang Y. CTNNAL1 deficiency suppresses CFTR expression in HDM-induced asthma mouse model through ROCK1-CAL signaling pathway. Acta Biochim Biophys Sin (Shanghai) 2023; 55:1618-1629. [PMID: 37715489 PMCID: PMC10579809 DOI: 10.3724/abbs.2023152] [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: 11/18/2022] [Accepted: 04/28/2023] [Indexed: 09/17/2023] Open
Abstract
The downregulation of adhesion molecule catenin alpha-like 1 (CTNNAL1) in airway epithelial cells of asthma patients and house dust mite (HDM)-induced asthma animal models was illustrated in our previous study. It is assumed to contribute to airway inflammation and mucus hypersecretion. In this work, we further explore the underlying mechanism of CTNNAL1 in asthma. CTNNAL1-silenced female mice exhibit a decreased level of cystic fibrosis transmembrane conductance regulator (CFTR), a cAMP-activated and ATP-gated Cl - channel that correlates with mucus hypersecretion. Our previous study demonstrated that ROCK1 expression decreases but ROCK2 expression increases in the lungs of a CTNNAL1-silenced mouse model. Inhibition of ROCK1 leads to a reduction in CFTR expression in CTNNAL1-overexpressing and CTNNAL1-silenced human bronchial epithelial (HBE) cells. It has been reported that ROCK1 is a downstream target of RhoA and that activation of RhoA increases CFTR expression after CTNNAL1 deficiency in vitro and in vivo. The above results indicate that CTNNAL1 regulates CFTR expression through the ROCK1 pathway. In addition, the expression of CFTR-associated ligand (CAL) is increased after CTNNAL1 silencing, and immunoprecipitation results confirm the interaction between ROCK1 and CAL. Inhibition of CAL does not influence ROCK1 expression but increases CFTR expression in CTNNAL1-silenced HBE cells. These data suggest that CTNNAL1 deficiency decreases CFTR expression in the HDM-induced asthma mouse model through the ROCK1-CAL signaling pathway.
Collapse
Affiliation(s)
- Di Wu
- School of MedicineFoshan UniversityFoshan528000China
- Department of PhysiologySchool of Basic Medical ScienceCentral South UniversityChangsha410008China
| | - Jiahui Zhu
- Department of PhysiologySchool of Basic Medical ScienceCentral South UniversityChangsha410008China
| | - Fang Yang
- School of MedicineFoshan UniversityFoshan528000China
| | - Riwang Li
- School of MedicineFoshan UniversityFoshan528000China
| | - Lexin Liu
- Department of PhysiologySchool of Basic Medical ScienceCentral South UniversityChangsha410008China
| | - Dahai Liu
- School of MedicineFoshan UniversityFoshan528000China
| | - Chi Liu
- Department of PhysiologySchool of Basic Medical ScienceCentral South UniversityChangsha410008China
| | - Xiangping Qu
- Department of PhysiologySchool of Basic Medical ScienceCentral South UniversityChangsha410008China
| | - Huijun Liu
- Department of PhysiologySchool of Basic Medical ScienceCentral South UniversityChangsha410008China
| | - Ming Ji
- Department of PhysiologySchool of Basic Medical ScienceCentral South UniversityChangsha410008China
| | - Xiaoqun Qin
- Department of PhysiologySchool of Basic Medical ScienceCentral South UniversityChangsha410008China
| | - Lan Hua
- the Second Xiangya Hospital of Central South UniversityChangsha410011China
| | - Yang Xiang
- Department of PhysiologySchool of Basic Medical ScienceCentral South UniversityChangsha410008China
| |
Collapse
|
2
|
BK channel clustering is required for normal behavioral alcohol sensitivity in C. elegans. Sci Rep 2019; 9:10224. [PMID: 31308408 PMCID: PMC6629859 DOI: 10.1038/s41598-019-46615-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Accepted: 07/02/2019] [Indexed: 02/05/2023] Open
Abstract
The large conductance, calcium- and voltage-activated potassium channel, known as the BK channel, is one of the central proteins that mediate alcohol intoxication and tolerance across species. Although ethanol targets BK channels through direct interaction, how ethanol-mediated BK channel activation causes behavioral intoxication is poorly understood. In. C. elegans, loss of function in SLO-1, the BK channel ortholog, confers profound ethanol resistance in movement and egg-laying behaviors. Here, we show that depletion of SLO-1 channels clustered at the active zones with no change in the overall channel expression level results in locomotory resistance to the intoxicating effect of ethanol, equivalent to that of slo-1 loss-of-function mutants. Likewise, depletion of clustered SLO-1 channels in the sarcolemma and neurons leads to ethanol-resistant egg-laying behavior. By contrast, reduction in the overall SLO-1 channel level by over 70% causes only moderate ethanol resistance in movement, and minimal, if any, resistance in egg laying. Our findings strongly suggest that behavioral ethanol sensitivity is conferred by local, but not global, depression of excitability via clustered BK channels. Given that clustered BK channels are functionally coupled to, and localize near, calcium channels, ethanol may mediate its behavioral effects by targeting BK channels and their coupled calcium channels.
Collapse
|
3
|
Tan M, Liu C, Huang W, Deng L, Qin X, Xiang Y. CTNNAL1 inhibits ozone-induced epithelial-mesenchymal transition in human bronchial epithelial cells. Exp Physiol 2018; 103:1157-1169. [PMID: 29791759 DOI: 10.1113/ep086839] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2017] [Accepted: 05/14/2018] [Indexed: 12/16/2022]
Abstract
NEW FINDINGS What is the central question of this study? What is the effect of catenin alpha-like 1 (CTNNAL1), an asthma-related epithelial adhesion molecule that plays a vital role in airway epithelial wound repair, on airway epithelial-mesenchymal transition? What is the main finding and its importance? CTNNAL1 inhibits ozone-induced airway epithelial-mesenchymal transition features, mediated by repressing the expression of Twist1 mRNA and reducing TGF-β1 levels. These findings contribute to our understanding of the pathology of airway EMT and may indicate a possible therapeutic target for airway remodelling in bronchial asthma. ABSTRACT Epithelial-mesenchymal transition (EMT), a crucial event occurring during epithelial and mesenchymal repair, was reported to be a possible mechanism for airway remodelling. Our previous work showed that the expression of catenin alpha-like 1 (CTNNAL1) was down-regulated in the bronchial epithelial cells of asthmatic models and played a vital role in airway epithelial wound repair. The aim of this study was to investigate the effect of CTNNAL1 on airway EMT. Overexpression or silencing of CTNNAL1 in human bronchial epithelial cells was induced by stable transfection. CTNNAL1 was silenced in primary mouse airway epithelial cells with an effective siRNA vector. Cells were stressed by ozone for 4 days at 30 min day-1 to induce EMT. EMT features, changes in the function of co-cultured lung fibroblasts, changes in the expression of the transcriptional repressors Snail/Slug and Twist1/Twist2 and changes in the secretion of transforming growth factor β1 (TGF-β1) were assayed in different cell lines with or without ozone exposure. Both ozone exposure and silencing of CTNNAL1 induced EMT features in airway epithelial cells. Functional changes in lung fibroblasts increased after co-culture with (ozone-stressed) CTNNAL1-silenced cells. Snail and Twist1 expression increased, and the level of TGF-β1 was enhanced. Conversely, CTNNAL1 overexpression reversed EMT features, repressed mRNA levels of Twist1 and reduced the secretion of TGF-β1, both alone and in combination with ozone exposure. Our results indicate that ozone exposure induces airway EMT and that CTNNAL1 inhibits ozone-induced airway EMT. CTNNAL1 may play a role in airway EMT by repressing the expression of Twist1 mRNA and reducing the level of TGF-β1.
Collapse
Affiliation(s)
- Meiling Tan
- School of Basic Medicine, Central South University, Changsha, Hunan, 410078, China
| | - Caixia Liu
- School of Basic Medicine, Central South University, Changsha, Hunan, 410078, China
| | - Wenjie Huang
- School of Basic Medicine, Central South University, Changsha, Hunan, 410078, China
| | - Linhong Deng
- Changzhou Key Laboratory of Respiratory Medical Engineering, Institute of Biomedical Engineering and Health Sciences, Changzhou, Jiangsu, 213164, China
| | - Xiaoqun Qin
- School of Basic Medicine, Central South University, Changsha, Hunan, 410078, China
| | - Yang Xiang
- School of Basic Medicine, Central South University, Changsha, Hunan, 410078, China
| |
Collapse
|
4
|
Deep imaging of bone marrow shows non-dividing stem cells are mainly perisinusoidal. Nature 2015; 526:126-30. [PMID: 26416744 PMCID: PMC4850557 DOI: 10.1038/nature15250] [Citation(s) in RCA: 522] [Impact Index Per Article: 52.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Accepted: 07/27/2015] [Indexed: 12/11/2022]
Abstract
Haematopoietic stem cells (HSCs) reside in a perivascular niche but the specific location of this niche remains controversial. HSCs are rare and few can be found in thin tissue sections or upon live imaging, making it difficult to comprehensively localize dividing and non-dividing HSCs. Here, using a green fluorescent protein (GFP) knock-in for the gene Ctnnal1 in mice (hereafter denoted as α-catulin(GFP)), we discover that α-catulin(GFP) is expressed by only 0.02% of bone marrow haematopoietic cells, including almost all HSCs. We find that approximately 30% of α-catulin-GFP(+)c-kit(+) cells give long-term multilineage reconstitution of irradiated mice, indicating that α-catulin-GFP(+)c-kit(+) cells are comparable in HSC purity to cells obtained using the best markers currently available. We optically cleared the bone marrow to perform deep confocal imaging, allowing us to image thousands of α-catulin-GFP(+)c-kit(+) cells and to digitally reconstruct large segments of bone marrow. The distribution of α-catulin-GFP(+)c-kit(+) cells indicated that HSCs were more common in central marrow than near bone surfaces, and in the diaphysis relative to the metaphysis. Nearly all HSCs contacted leptin receptor positive (Lepr(+)) and Cxcl12(high) niche cells, and approximately 85% of HSCs were within 10 μm of a sinusoidal blood vessel. Most HSCs, both dividing (Ki-67(+)) and non-dividing (Ki-67(-)), were distant from arterioles, transition zone vessels, and bone surfaces. Dividing and non-dividing HSCs thus reside mainly in perisinusoidal niches with Lepr(+)Cxcl12(high) cells throughout the bone marrow.
Collapse
|
5
|
Bear MD, Liu T, Abualkhair S, Ghamloush MA, Hill NS, Preston I, Fanburg BL, Kayyali US, Toksoz D. Alpha-Catulin Co-Localizes With Vimentin Intermediate Filaments and Functions in Pulmonary Vascular Endothelial Cell Migration via ROCK. J Cell Physiol 2015; 231:934-43. [PMID: 26377600 DOI: 10.1002/jcp.25185] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Accepted: 09/03/2015] [Indexed: 01/01/2023]
Abstract
The ubiquitous α-catulin acts as a scaffold for distinct signalosomes including RhoA/ROCK; however, its function is not well understood. While α-catulin has homology to the cytoskeletal linkers α-catenin and vinculin, it appears to be functionally divergent. Here we further investigated α-catulin function in pulmonary vascular endothelial cells (VEC) on the premise that α-catulin has a unique cytoskeletal role. Examination of endogenous α-catulin intracellular localization by immunofluorescence revealed a highly organized cytosolic filamentous network suggestive of a cytoskeletal system in a variety of cultured VEC. Double-immunofluorescence analyses of VEC showed endogenous α-catulin co-localization with vimentin intermediate filaments. Similar to vimentin, α-catulin was found to distribute into detergent-soluble and -insoluble fractions. Treatment of VEC with withaferinA, an agent that targets vimentin filaments, disrupted the α-catulin network distribution and altered α-catulin solubility. Vimentin participates in cell migration, and withaferinA was found to inhibit VEC migration in vitro; similarly, α-catulin knock-down reduced VEC migration. Based on previous reports showing that ROCK modulates vimentin, we found that ROCK depletion attenuated VEC migration; furthermore, α-catulin depletion was shown to reduce ROCK-induced signaling. These findings indicate that α-catulin has a unique function in co-localization with vimentin filaments that contributes to VEC migration via a pathway that may involve ROCK signaling. J. Cell. Physiol. 231: 934-943, 2016. © 2015 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Michael D Bear
- Division of Pulmonary, Critical Care and Sleep, Tupper Research Institute, Tufts Medical Center, Tufts University School of Medicine, Boston, Massachusetts
| | - Tiegang Liu
- Division of Pulmonary, Critical Care and Sleep, Tupper Research Institute, Tufts Medical Center, Tufts University School of Medicine, Boston, Massachusetts
| | - Shereen Abualkhair
- Division of Pulmonary, Critical Care and Sleep, Tupper Research Institute, Tufts Medical Center, Tufts University School of Medicine, Boston, Massachusetts
| | | | - Nicholas S Hill
- Division of Pulmonary, Critical Care and Sleep, Tupper Research Institute, Tufts Medical Center, Tufts University School of Medicine, Boston, Massachusetts
| | - Ioana Preston
- Division of Pulmonary, Critical Care and Sleep, Tupper Research Institute, Tufts Medical Center, Tufts University School of Medicine, Boston, Massachusetts
| | - Barry L Fanburg
- Division of Pulmonary, Critical Care and Sleep, Tupper Research Institute, Tufts Medical Center, Tufts University School of Medicine, Boston, Massachusetts
| | - Usamah S Kayyali
- Division of Pulmonary, Critical Care and Sleep, Tupper Research Institute, Tufts Medical Center, Tufts University School of Medicine, Boston, Massachusetts
| | - Deniz Toksoz
- Division of Pulmonary, Critical Care and Sleep, Tupper Research Institute, Tufts Medical Center, Tufts University School of Medicine, Boston, Massachusetts
| |
Collapse
|
6
|
The upregulated α-catulin expression was involved in head-neck squamous cell carcinogenesis by promoting proliferation, migration, invasion, and epithelial to mesenchymal transition. Tumour Biol 2015; 37:1671-81. [DOI: 10.1007/s13277-015-3901-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Accepted: 08/05/2015] [Indexed: 10/23/2022] Open
|
7
|
The maternal-to-zygotic transition targets actin to promote robustness during morphogenesis. PLoS Genet 2013; 9:e1003901. [PMID: 24244181 PMCID: PMC3820746 DOI: 10.1371/journal.pgen.1003901] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2013] [Accepted: 09/06/2013] [Indexed: 11/26/2022] Open
Abstract
Robustness is a property built into biological systems to ensure stereotypical outcomes despite fluctuating inputs from gene dosage, biochemical noise, and the environment. During development, robustness safeguards embryos against structural and functional defects. Yet, our understanding of how robustness is achieved in embryos is limited. While much attention has been paid to the role of gene and signaling networks in promoting robust cell fate determination, little has been done to rigorously assay how mechanical processes like morphogenesis are designed to buffer against variable conditions. Here we show that the cell shape changes that drive morphogenesis can be made robust by mechanisms targeting the actin cytoskeleton. We identified two novel members of the Vinculin/α-Catenin Superfamily that work together to promote robustness during Drosophila cellularization, the dramatic tissue-building event that generates the primary epithelium of the embryo. We find that zygotically-expressed Serendipity-α (Sry-α) and maternally-loaded Spitting Image (Spt) share a redundant, actin-regulating activity during cellularization. Spt alone is sufficient for cellularization at an optimal temperature, but both Spt plus Sry-α are required at high temperature and when actin assembly is compromised by genetic perturbation. Our results offer a clear example of how the maternal and zygotic genomes interact to promote the robustness of early developmental events. Specifically, the Spt and Sry-α collaboration is informative when it comes to genes that show both a maternal and zygotic requirement during a given morphogenetic process. For the cellularization of Drosophilids, Sry-α and its expression profile may represent a genetic adaptive trait with the sole purpose of making this extreme event more reliable. Since all morphogenesis depends on cytoskeletal remodeling, both in embryos and adults, we suggest that robustness-promoting mechanisms aimed at actin could be effective at all life stages. Every embryo develops under its own unique set of circumstances, with variable inputs coming from mother, father, and the environment. To then ensure a reliable outcome, mechanisms are built into development to buffer against challenges like genetic deficiency, maternal fever, alcohol exposure, etc. This buffering, called “robustness”, can be overwhelmed, ending in miscarriage, pre-mature birth, and structural and functional birth defects. Thus, we need to understand how robustness arises in order to define an embryo's susceptibilities to genetic background and environment; and to ultimately promote healthy reproduction. In this work we provide new insight into how morphogenesis, the process of tissue building in embryos, is made more robust. First, we show that early gene expression by the embryo, or zygote, supplements the stockpile of proteins already supplied by the mother to ensure the robustness of early morphogenesis. Specifically, our data suggests that a specific gene, sry-α, and its expression by the embryo at the maternal-to-zygotic transition, is a genetic adaptation with the sole function of making the first tissue building event in the fruit fly more robust. In addition, we show that the robustness of this morphogenetic event is promoted by mechanisms regulating the actin cytoskeleton.
Collapse
|
8
|
Liang CH, Chiu SY, Hsu IL, Wu YY, Tsai YT, Ke JY, Pan SH, Hsu YC, Li KC, Yang PC, Chen YL, Hong TM. α-Catulin drives metastasis by activating ILK and driving an αvβ3 integrin signaling axis. Cancer Res 2012; 73:428-38. [PMID: 23047866 DOI: 10.1158/0008-5472.can-12-2095] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
α-Catulin is an oncoprotein that helps sustain proliferation by preventing cellular senescence. Here, we report that α-catulin also drives malignant invasion and metastasis. α-Catulin was upregulated in highly invasive non-small cell lung cancer (NSCLC) cell lines, where its ectopic expression or short-hairpin RNA-mediated attenuation enhanced or limited invasion or metastasis, respectively. α-Catulin interacted with integrin-linked kinase (ILK), a serine/threonine protein kinase implicated in cancer cell proliferation, antiapoptosis, invasion, and angiogenesis. Attenuation of ILK or α-catulin reciprocally blocked cell migration and invasion induced by the other protein. Mechanistic investigations revealed that α-catulin activated Akt-NF-κB signaling downstream of ILK, which in turn led to increased expression of fibronectin and integrin αvβ3. Pharmacologic or antibody-mediated blockade of NF-κB or αvβ3 was sufficient to inhibit α-catulin-induced cell migration and invasion. Clinically, high levels of expression of α-catulin and ILK were associated with poor overall survival in patients with NSCLC. Taken together, our study shows that α-catulin plays a critical role in cancer metastasis by activating the ILK-mediated Akt-NF-κB-αvβ3 signaling axis.
Collapse
Affiliation(s)
- Chen-Hsien Liang
- Institute of Basic Medical Sciences, Institute of Oral Medicine, and Graduate Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
9
|
Oh HJ, Abraham LS, van Hengel J, Stove C, Proszynski TJ, Gevaert K, DiMario JX, Sanes JR, van Roy F, Kim H. Interaction of α-catulin with dystrobrevin contributes to integrity of dystrophin complex in muscle. J Biol Chem 2012; 287:21717-28. [PMID: 22577143 DOI: 10.1074/jbc.m112.369496] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The dystrophin complex is a multimolecular membrane-associated protein complex whose defects underlie many forms of muscular dystrophy. The dystrophin complex is postulated to function as a structural element that stabilizes the cell membrane by linking the contractile apparatus to the extracellular matrix. A better understanding of how this complex is organized and localized will improve our knowledge of the pathogenic mechanisms of diseases that involve the dystrophin complex. In a Caenorhabditis elegans genetic study, we demonstrate that CTN-1/α-catulin, a cytoskeletal protein, physically interacts with DYB-1/α-dystrobrevin (a component of the dystrophin complex) and that this interaction is critical for the localization of the dystrophin complex near dense bodies, structures analogous to mammalian costameres. We further show that in mouse α-catulin is localized at the sarcolemma and neuromuscular junctions and interacts with α-dystrobrevin and that the level of α-catulin is reduced in α-dystrobrevin-deficient mouse muscle. Intriguingly, in the skeletal muscle of mdx mice lacking dystrophin, we discover that the expression of α-catulin is increased, suggesting a compensatory role of α-catulin in dystrophic muscle. Together, our study demonstrates that the interaction between α-catulin and α-dystrobrevin is evolutionarily conserved in C. elegans and mammalian muscles and strongly suggests that this interaction contributes to the integrity of the dystrophin complex.
Collapse
Affiliation(s)
- Hyun J Oh
- Department of Cell Biology and Anatomy, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois 60064, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Xiang Y, Qin XQ, Liu HJ, Tan YR, Liu C, Liu CX. Identification of transcription factors regulating CTNNAL1 expression in human bronchial epithelial cells. PLoS One 2012; 7:e31158. [PMID: 22359570 PMCID: PMC3281047 DOI: 10.1371/journal.pone.0031158] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2011] [Accepted: 01/03/2012] [Indexed: 11/18/2022] Open
Abstract
Adhesion molecules play important roles in airway hyperresponsiveness or airway inflammation. Our previous study indicated catenin alpha-like 1 (CTNNAL1), an alpha-catenin-related protein, was downregulated in asthma patients and animal model. In this study, we observed that the expression of CTNNAL1 was increased in lung tissue of the ozone-stressed Balb/c mice model and in acute ozone stressed human bronchial epithelial cells (HBEC). In order to identify the possible DNA-binding proteins regulating the transcription of CTNNAL1 gene in HBEC, we designed 8 oligo- nucleotide probes corresponding to various regions of the CTNNAL1 promoter in electrophoretic mobility shift assays (EMSA). We detected 5 putative transcription factors binding sites within CTNNAL1 promoter region that can recruit LEF-1, AP-2α and CREB respectively by EMSA and antibody supershift assay. Chromatin immunoprecipitation (ChIP) assay verified that AP-2 α and LEF-1 could be recruited to the CTNNAL1 promoter. Therefore we further analyzed the functions of putative AP-2 and LEF-1 sites within CTNNAL1 promoter by site-directed mutagenesis of those sites within pGL3/FR/luc. We observed a reduction in human CTNNAL1 promoter activity of mutants of both AP-2α and LEF-1 sites. Pre-treatment with ASOs targeting LEF-1and AP-2α yielded significant reduction of ozone-stress-induced CTNNAL1 expression. The activation of AP-2α and LEF-1, followed by CTNNAL1 expression, showed a correlation during a 16-hour time course. Our data suggest that a robust transcriptional CTNNAL1 up-regulation occurs during acute ozone-induced stress and is mediated at least in part by ozone-induced recruitments of LEF-1 and AP-2α to the human CTNNAL1 promoter.
Collapse
Affiliation(s)
- Yang Xiang
- Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Xiao-Qun Qin
- Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- * E-mail:
| | - Hui-Jun Liu
- Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Yu-Rong Tan
- Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Chi Liu
- Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Cai-Xia Liu
- Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| |
Collapse
|
11
|
Nakamori M, Takahashi MP. The role of α-dystrobrevin in striated muscle. Int J Mol Sci 2011; 12:1660-71. [PMID: 21673914 PMCID: PMC3111625 DOI: 10.3390/ijms12031660] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2010] [Revised: 01/29/2011] [Accepted: 02/23/2011] [Indexed: 12/29/2022] Open
Abstract
Muscular dystrophies are a group of diseases that primarily affect striated muscle and are characterized by the progressive loss of muscle strength and integrity. Major forms of muscular dystrophies are caused by the abnormalities of the dystrophin glycoprotein complex (DGC) that plays crucial roles as a structural unit and scaffolds for signaling molecules at the sarcolemma. α-Dystrobrevin is a component of the DGC and directly associates with dystrophin. α-Dystrobrevin also binds to intermediate filaments as well as syntrophin, a modular adaptor protein thought to be involved in signaling. Although no muscular dystrophy has been associated within mutations of the α-dystrobrevin gene, emerging findings suggest potential significance of α-dystrobrevin in striated muscle. This review addresses the functional role of α-dystrobrevin in muscle as well as its possible implication for muscular dystrophy.
Collapse
Affiliation(s)
- Masayuki Nakamori
- Department of Neurology, Osaka University Graduate School of Medicine, 2-2, D-4, Yamadaoka, Suita, Osaka 565-0871, Japan; E-Mail:
- Department of Neurology, University of Rochester Medical Center, 601 Elmwood Avenue, Box 645 URMC, Rochester, NY 14642, USA
| | - Masanori P. Takahashi
- Department of Neurology, Osaka University Graduate School of Medicine, 2-2, D-4, Yamadaoka, Suita, Osaka 565-0871, Japan; E-Mail:
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +81-6-6879-3571; Fax: +81-6-6879-3579
| |
Collapse
|
12
|
Fan LC, Chiang WF, Liang CH, Tsai YT, Wong TY, Chen KC, Hong TM, Chen YL. α-Catulin knockdown induces senescence in cancer cells. Oncogene 2011; 30:2610-21. [PMID: 21278790 DOI: 10.1038/onc.2010.637] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Cellular senescence functions as a tumor suppressor that protects against cancer progression. α-Catulin, an α-catenin-related protein, is reported to have tumorigenic potential because it regulates the nuclear factor-κB (NF-κB) pathway, but little is known about its clinical relevance and the mechanism through which it regulates cancer progression. Here, we found that α-catulin mRNA levels were significantly upregulated in cancer cell lines and clinical oral squamous cell carcinomas, which positively correlated with tumor size (P=0.001) and American Joint Committee on Cancer (AJCC) stage (P=0.004). α-Catulin knockdown in the OC2 and A549 cancer cell lines dramatically decreased cell proliferation and contributed to cellular senescence, and inhibited OC2 xenograft growth. Mechanistic dissection showed that α-catulin depletion strongly induced the DNA-damage response (DDR) in both cell lines, via a p53/p21-dependent pathway in A549 cells, but a p53/p21-independent pathway in OC2 cells carrying mutant p53. Global gene expression analysis revealed that α-catulin knockdown altered cell-cycle regulation and DDR pathways at the presenescent stage as well as significantly downregulate several crucial genes related to mitotic chromosome condensation, DDR and DNA repair systems, which suggests that its depletion-induced cellular senescence might be caused by chromosome condensation failures, severe DNA damage and impaired DNA repair ability. Our study provides evidence that α-catulin promotes tumor growth by preventing cellular senescence and suggests that downregulating α-catulin may be a promising therapeutic approach for cancer treatment.
Collapse
Affiliation(s)
- L-C Fan
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan, ROC
| | | | | | | | | | | | | | | |
Collapse
|
13
|
Abraham LS, Oh HJ, Sancar F, Richmond JE, Kim H. An alpha-catulin homologue controls neuromuscular function through localization of the dystrophin complex and BK channels in Caenorhabditis elegans. PLoS Genet 2010; 6. [PMID: 20865173 PMCID: PMC2928805 DOI: 10.1371/journal.pgen.1001077] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2010] [Accepted: 07/21/2010] [Indexed: 11/25/2022] Open
Abstract
The large conductance, voltage- and calcium-dependent potassium (BK) channel serves as a major negative feedback regulator of calcium-mediated physiological processes and has been implicated in muscle dysfunction and neurological disorders. In addition to membrane depolarization, activation of the BK channel requires a rise in cytosolic calcium. Localization of the BK channel near calcium channels is therefore critical for its function. In a genetic screen designed to isolate novel regulators of the Caenorhabditis elegans BK channel, SLO-1, we identified ctn-1, which encodes an α-catulin homologue with homology to the cytoskeletal proteins α-catenin and vinculin. ctn-1 mutants resemble slo-1 loss-of-function mutants, as well as mutants with a compromised dystrophin complex. We determined that CTN-1 uses two distinct mechanisms to localize SLO-1 in muscles and neurons. In muscles, CTN-1 utilizes the dystrophin complex to localize SLO-1 channels near L-type calcium channels. In neurons, CTN-1 is involved in localizing SLO-1 to a specific domain independent of the dystrophin complex. Our results demonstrate that CTN-1 ensures the localization of SLO-1 within calcium nanodomains, thereby playing a crucial role in muscles and neurons. Calcium ions are essential for many physiological processes, including neurosecretion and neuronal and muscle excitation. Paradoxically, abnormal accumulation of calcium ions is associated with cell death and has been documented as an early event in muscle and neural degenerative diseases. One mechanism to avoid detrimental calcium accumulation is to link the calcium increase with activation of calcium-dependent potassium ion channels, thereby reducing cell excitability and preventing further calcium influx. This negative feedback requires these potassium channels to be localized in close proximity to sites of calcium entry. In a Caenorhabditis elegans genetic screen, we identified α-catulin, known as a cytoskeletal regulatory protein in mammals, important for the localization of calcium-dependent potassium channels in both muscles and neurons. In muscle, α-catulin controls the localization of the dystrophin complex, a multimeric protein complex implicated in muscular dystrophy. The dystrophin complex in turn tethers the calcium-dependent potassium channels near calcium channels. In neurons, the α-catulin-mediated localization of the potassium channels is independent of the dystrophin complex. Lack of α-catulin results in mislocalization of the potassium channels, and in turn causes defects in neuromuscular function. Our results support the idea that cytoskeletal proteins function as anchor molecules that localize ion channels to specific cellular domains.
Collapse
Affiliation(s)
- Linu S. Abraham
- Department of Cell Biology and Anatomy, The Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois, United States of America
| | - Hyun J. Oh
- Department of Cell Biology and Anatomy, The Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois, United States of America
| | - Feyza Sancar
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Janet E. Richmond
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Hongkyun Kim
- Department of Cell Biology and Anatomy, The Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois, United States of America
- * E-mail:
| |
Collapse
|
14
|
α-Catulin CTN-1 is required for BK channel subcellular localization in C. elegans body-wall muscle cells. EMBO J 2010; 29:3184-95. [PMID: 20700105 DOI: 10.1038/emboj.2010.194] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2010] [Accepted: 07/20/2010] [Indexed: 11/09/2022] Open
Abstract
The BK channel, a voltage- and Ca(2+)-gated large-conductance potassium channel with many important functions, is often localized at specific subcellular domains. Although proper subcellular localization is likely a prerequisite for the channel to perform its physiological functions, little is known about the molecular basis of localization. Here, we show that CTN-1, a homologue of mammalian α-catulin, is required for subcellular localization of SLO-1, the Caenorhabditis elegans BK channel α-subunit, in body-wall muscle cells. CTN-1 was identified in a genetic screen for mutants that suppressed a lethargic phenotype caused by expressing a gain-of-function (gf) isoform of SLO-1. In body-wall muscle cells, CTN-1 coclusters with SLO-1 at regions of dense bodies, which are Z-disk analogs of mammalian skeletal muscle. In ctn-1 loss-of-function (lf) mutants, SLO-1 was mislocalized in body-wall muscle but its transcription and protein level were unchanged. Targeted rescue of ctn-1(lf) in muscle was sufficient to reinstate the lethargic phenotype in slo-1(gf);ctn-1(lf). These results suggest that CTN-1 plays an important role in BK channel function by mediating channel subcellular localization.
Collapse
|
15
|
Bear MD, Li M, Liu Y, Giel-Moloney MA, Fanburg BL, Toksoz D. The Lbc Rho guanine nucleotide exchange factor α-catulin axis functions in serotonin-induced vascular smooth muscle cell mitogenesis and RhoA/ROCK activation. J Biol Chem 2010; 285:32919-32926. [PMID: 20696764 DOI: 10.1074/jbc.m109.062513] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Serotonin (5-hydroxytryptamine, 5-HT) is mitogenic for several cell types including pulmonary arterial smooth muscle cells (PASMC), and is associated with the abnormal vascular smooth muscle remodeling that occurs in pulmonary arterial hypertension. RhoA/Rho kinase (ROCK) function is required for 5-HT-induced PASMC mitogenesis, and 5-HT activates RhoA; however, the signaling steps are poorly defined. Rho guanine nucleotide exchange factors (Rho GEFs) transduce extracellular signals to Rho, and we found that 5-HT treatment of PASMC led to increased membrane-associated Lbc Rho GEF, suggesting modulation by 5-HT. Lbc knockdown by siRNA attenuated 5-HT-induced thymidine uptake in PASMC, indicating a role in PASMC mitogenesis. 5-HT triggered Rho-dependent serum response factor-mediated reporter activation in PASMC, and this was reduced by Lbc depletion. Lbc knockdown reduced 5-HT-induced RhoA/ROCK activation, but not p42/44 ERK MAP kinase activation, suggesting that Lbc is an intermediary between 5-HT and RhoA/ROCK, but not ERK. 5-HT stimulation of PASMC led to increased association between Lbc, RhoA, and the α-catulin scaffold. Furthermore, α-catulin knockdown attenuated 5-HT-induced PASMC thymidine uptake. 5-HT-induced PASMC mitogenesis was reduced by dominant-negative G(q) protein, suggesting cooperation with Lbc/α-catulin. These results for the first time define a Rho GEF involved in vascular smooth muscle cell growth and serotonin signaling, and suggest that Lbc Rho GEF family members play distinct roles. Thus, the Lbc/α-catulin axis participates in 5-HT-induced PASMC mitogenesis and RhoA/ROCK signaling, and may be an interventional target in diseases involving vascular smooth muscle remodeling.
Collapse
Affiliation(s)
- Michael D Bear
- From the Division of Pulmonary and Critical Care, Tupper Research Institute, Tufts Medical Center, Boston, Massachusetts 02111
| | - Min Li
- From the Division of Pulmonary and Critical Care, Tupper Research Institute, Tufts Medical Center, Boston, Massachusetts 02111
| | - Yinglin Liu
- From the Division of Pulmonary and Critical Care, Tupper Research Institute, Tufts Medical Center, Boston, Massachusetts 02111
| | - Maryann A Giel-Moloney
- Department of Physiology, Tufts University School of Medicine, Boston, Massachusetts 02111
| | - Barry L Fanburg
- From the Division of Pulmonary and Critical Care, Tupper Research Institute, Tufts Medical Center, Boston, Massachusetts 02111
| | - Deniz Toksoz
- From the Division of Pulmonary and Critical Care, Tupper Research Institute, Tufts Medical Center, Boston, Massachusetts 02111.
| |
Collapse
|
16
|
Xiang Y, Tan YR, Zhang JS, Qin XQ, Hu BB, Wang Y, Qu F, Liu HJ. Wound repair and proliferation of bronchial epithelial cells regulated by CTNNAL1. J Cell Biochem 2008; 103:920-30. [PMID: 17647259 DOI: 10.1002/jcb.21461] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Adhesion molecules play vital roles in airway hyperresponsiveness (AHR) or airway inflammation. Our previous study indicated that adhesion molecule catenin alpha-like 1 (CTNNAL1) is relevant closely to asthma susceptibility, but its biological function or significance is still unclear. In the present study, we observed the temporal and spatial distribution of CTNNAL1 expression in mouse lung tissue with the OVA-sensitized asthma model and found that the level of CTNNAL1 mRNA showed a prominent negative correlation with pulmonary resistance (R(L)). To study the function of CTNNAL1 in airway, effects of CTNNAL1 on proliferation and wound repair activity of human bronchial epithelial cells (HBEC) was investigated with antisense oligonucleotide (ASO) technique. The results showed that: (1) CTNNAL1 ASO could decelerate the repairing velocity and proliferation of HBEC; (2) CTNNAL1 expression was increased on the edge cells of mechanic wounded area in culture; (3) extracellular matrix component fibronectin (Fn) obviously promoted wound repair activity and proliferation of HBEC, which could be blocked by CTNNAL1 ASO; (4) Western blot showed that Fn could promote FAK phosphorylation, which also be inhibited by CTNNAL1 ASO. In conclusion, the level of CTNNAL1 mRNA expression is highly correlated to airway resistance; CTNNAL1 may contribute to the wound repair and proliferation of HBEC. Furthermore, it may serve to Fn mediated cell-extracellular adhesion and its signal transduction.
Collapse
Affiliation(s)
- Yang Xiang
- Xiangya School of Medicine, Central South University, Changsha 410078, Hunan, PR China
| | | | | | | | | | | | | | | |
Collapse
|
17
|
Wiesner C, Winsauer G, Resch U, Hoeth M, Schmid JA, van Hengel J, van Roy F, Binder BR, de Martin R. Alpha-catulin, a Rho signalling component, can regulate NF-kappaB through binding to IKK-beta, and confers resistance to apoptosis. Oncogene 2007; 27:2159-69. [PMID: 17952117 DOI: 10.1038/sj.onc.1210863] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Rho GTPases regulate diverse cellular functions including adhesion, cytokinesis and motility, as well as the activity of the transcription factors NF-kappaB, serum response factor and C/EBP. alpha-Catulin, an alpha-catenin-related protein that shares structural similarities with cytoskeletal linker proteins, facilitates Rho signalling by serving as a scaffold for the Rho-specific guanine nucleotide exchange factor Lbc. We report here that alpha-catulin also interacts with a key component of the NF-kappaB signalling pathway, namely the IkappaB kinase (IKK)-beta. In co-immunoprecipitations, alpha-catulin can bind IKK-beta and Lbc. Ectopic expression of alpha-catulin augmented NF-kappaB activity, promoted cell migration and increased resistance to apoptosis, whereas knockdown experiments showed the opposite effects. Together, these features suggest that alpha-catulin has tumorigenic potential.
Collapse
Affiliation(s)
- C Wiesner
- Department of Vascular Biology and Thrombosis Research, Medical University of Vienna, Vienna, Austria
| | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Goossens S, Janssens B, Bonné S, De Rycke R, Braet F, van Hengel J, van Roy F. A unique and specific interaction between alphaT-catenin and plakophilin-2 in the area composita, the mixed-type junctional structure of cardiac intercalated discs. J Cell Sci 2007; 120:2126-36. [PMID: 17535849 DOI: 10.1242/jcs.004713] [Citation(s) in RCA: 95] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Alpha-catenins play key functional roles in cadherin-catenin cell-cell adhesion complexes. We previously reported on alphaT-catenin, a novel member of the alpha-catenin protein family. alphaT-catenin is expressed predominantly in cardiomyocytes, where it colocalizes with alphaE-catenin at the intercalated discs. Whether alphaT- and alphaE-catenin have specific or synergistic functions remains unknown. In this study we used the yeast two-hybrid approach to identify specific functions of alphaT-catenin. An interaction between alphaT-catenin and plakophilins was observed and subsequently confirmed by co-immunoprecipitation and colocalization. Interaction with the amino-terminal part of plakophilins appeared to be specific for the central ;adhesion-modulation' domain of alphaT-catenin. In addition, we showed, by immuno-electron microscopy, that desmosomal proteins in the heart localize not only to the desmosomes in the intercalated discs but also at adhering junctions with hybrid composition. We found that in the latter junctions, endogenous plakophilin-2 colocalizes with alphaT-catenin. By providing an extra link between the cadherin-catenin complex and intermediate filaments, the binding of alphaT-catenin to plakophilin-2 is proposed to be a means of modulating and strengthening cell-cell adhesion between cardiac muscle cells. This could explain the devastating effect of plakophilin-2 mutations on cell junction stability in intercalated discs, which lead to cardiac muscle malfunction.
Collapse
Affiliation(s)
- Steven Goossens
- Department for Molecular Biomedical Research, VIB, Ghent University, B-9052 Ghent, Belgium
| | | | | | | | | | | | | |
Collapse
|
19
|
Merdek KD, Nguyen NT, Toksoz D. Distinct activities of the alpha-catenin family, alpha-catulin and alpha-catenin, on beta-catenin-mediated signaling. Mol Cell Biol 2004; 24:2410-22. [PMID: 14993280 PMCID: PMC355851 DOI: 10.1128/mcb.24.6.2410-2422.2004] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Alpha-catenin, an integral part of cadherin-catenin adhesion complexes, is a major binding partner of beta-catenin, a key component of the Wnt pathway, which activates T-cell factor (TCF)/lymphoid enhancer factor (LEF) transcription and is often upregulated in cancers. Recently, we identified an alpha-catenin-related protein, alpha-catulin, whose function is poorly understood, as part of a Rho GTPase signaling complex. Here, based on evidence suggesting that alpha-catulin may associate with a beta-catenin fraction, we investigated the role of alpha-catenin family members in beta-catenin-mediated signals. Expression of the full length or a 103-residue region of alpha-catenin strongly inhibits the induction of the TCF/LEF-responsive TOPFLASH reporter in HEK293T cells expressing activated beta-catenin or in cancer cells with constitutively upregulated Wnt signaling, whereas alpha-catulin expression had no effect. Interestingly, alpha-catulin expression attenuates the activation of the cyclin D1 promoter, a target of Wnt pathway signals. Alpha-catulin appears to inhibit Ras-mediated signals to the cyclin D1 promoter, rather than beta-catenin signals, and the synergy between Ras and beta-catenin required to fully activate this promoter. Data suggesting the involvement of Rho in this response are presented and discussed. These results suggest a novel function for alpha-catulin and imply that alpha-catenin and alpha-catulin have distinct activities that downregulate, respectively, beta-catenin and Ras signals converging on the cyclin D1 promoter.
Collapse
Affiliation(s)
- Keith D Merdek
- Department of Physiology, Tufts University School of Medicine, Boston, Massachusetts 02111, USA.
| | | | | |
Collapse
|
20
|
Bourdeau I, Antonini SR, Lacroix A, Kirschner LS, Matyakhina L, Lorang D, Libutti SK, Stratakis CA. Gene array analysis of macronodular adrenal hyperplasia confirms clinical heterogeneity and identifies several candidate genes as molecular mediators. Oncogene 2004; 23:1575-85. [PMID: 14767469 DOI: 10.1038/sj.onc.1207277] [Citation(s) in RCA: 92] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Corticotropin (ACTH)-independent macronodular adrenal hyperplasia (AIMAH) is a heterogeneous condition in which cortisol secretion may be mediated by gastrointestinal peptide (GIP), vasopressin, catecholamines and other hormones. We studied the expression profile of AIMAH by genomic cDNA microarray analysis. Total RNA was extracted from eight tissues (three GIP-dependent) and compared to total RNA obtained from adrenal glands from 62 normal subjects. Genes had to be altered in 75% of the patients, and be up- or downregulated at a cutoff ratio of at least 2.0; 82 and 31 genes were found to be consistently up- and downregulated, respectively. Among the former were regulators of transcription, chromatin remodeling, and cell cycle and adhesion. Downregulated sequences included genes involved in immune responses and insulin signaling. Hierarchical clustering correlated with the two main AIMAH diagnostic groups: GIP-dependent and non-GIP-dependent. The genes encoding the 7B2 protein (SGNE1) and WNT1-inducible signaling pathway protein 2 (WISP2) were specifically overexpressed in the GIP-dependent AIMAH. For these, and six more genes, the data were validated by semiquantitative amplification in samples from a total of 32 patients (the original eight, six more cases of AIMAH, and 18 other adrenocortical hyperplasias and tumors) and the H295R adrenocortical cancer cell line. In conclusion, our data confirmed AIMAH's clinical heterogeneity by identifying molecularly distinct diagnostic subgroups. Several candidate genes that may be responsible for AIMAH formation and/or progression were also identified, suggesting pathways that affect the cell cycle, adhesion and transcription as possible mediators of adrenocortical hyperplasia.
Collapse
Affiliation(s)
- Isabelle Bourdeau
- Section on Endocrinology & Genetics, Developmental Endocrinology Branch, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | | | | | | | | | |
Collapse
|
21
|
Park B, Nguyen NT, Dutt P, Merdek KD, Bashar M, Sterpetti P, Tosolini A, Testa JR, Toksoz D. Association of Lbc Rho guanine nucleotide exchange factor with alpha-catenin-related protein, alpha-catulin/CTNNAL1, supports serum response factor activation. J Biol Chem 2002; 277:45361-70. [PMID: 12270917 DOI: 10.1074/jbc.m202447200] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The Rho GTPase signaling pathway is required for actin cytoskeletal organization and serum response factor-dependent gene transcription. Lbc is a Rho-specific guanine nucleotide exchange factor that contains a modulatory C-terminal region. To elucidate Lbc regulatory mechanism(s), a yeast two-hybrid screen for proteins that interact with the Lbc C-terminal region was carried out, resulting in multiple isolation of cDNAs encoding the same 734-amino acid Lbc interacting protein. The Lbc interacting protein has homology with the alpha-catenin cell adhesion component and is identical to the alpha-catenin-like alpha-catulin protein of unknown function. The human alpha-catulin gene (CTNNAL1) maps to 9q31-32. Here we identify the predicted endogenous alpha-catulin product, document alpha-catulin and Lbc co-expression in multiple human cell lines, and show alpha-catulin and Lbc subcellular co-fractionation and intracellular localization. The required regions for Lbc and alpha-catulin interaction were mapped, and complex formation between Lbc and alpha-catulin in mammalian cells was detected. Functionally, alpha-catulin co-expression leads to increased Lbc-induced serum response factor activation in vivo as measured by a transcriptional reporter assay. Furthermore, alpha-catulin co-expression enhances Lbc-induced GTP-Rho formation in vivo. These results support the concept that the recently identified alpha-catulin protein may modulate Rho pathway signaling in vivo by providing a scaffold for the Lbc Rho guanine nucleotide exchange factor.
Collapse
Affiliation(s)
- Brian Park
- Department of Physiology, Tufts University School of Medicine, Boston, Massachusetts 02111, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Vanpoucke G, Nollet F, Tejpar S, Cassiman JJ, van Roy F. The human alphaE-catenin gene CTNNA1: mutational analysis and rare occurrence of a truncated splice variant. BIOCHIMICA ET BIOPHYSICA ACTA 2002; 1574:262-8. [PMID: 11997091 DOI: 10.1016/s0167-4781(01)00378-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Abnormal expression of the alphaE-catenin protein, a component of the E-cadherin/catenin cell adhesion complex, is frequently observed in human cancer cells. An inverse correlation between alphaE-catenin expression and tumor malignancy can be of prognostic value. Mutations of the alphaE-catenin gene, CTNNA1, were described in several human cancer cell lines and were found to result in aberrant cell adhesion. We have developed a polymerase chain reaction/single-strand conformation polymorphism-based method for mutation analysis of this gene in human tumor DNA. This approach enabled us to identify several polymorphisms in a set of desmoid tumors, demonstrating that this method is suitable for alphaE-catenin mutational analysis. On the basis of our genomic characterization data, we found that the previously reported alternative splicing of the alphaE-catenin gene actually generates a frame-shift, resulting in a truncated alphaE-catenin protein. This finding is unlike the other alpha-catenin family members alphaN-catenin and vinculin, which show in-frame alternative inserts. Furthermore, real-time quantitative reverse transcriptase-PCR analysis did not reveal relevant expression levels of this alternatively spliced alphaE-catenin variant neither in any human tissue or cell line tested, nor at any mouse developmental stage tested. Thus, contrary to previous notions, alternative splicing with in-frame insertion nearby the C-terminal end of the protein is not a general feature for all members of the alpha-catenin/vinculin family.
Collapse
Affiliation(s)
- Griet Vanpoucke
- Molecular Cell Biology Unit, Department for Molecular Biomedical Research, Flanders Interuniversity Institute for Biotechnology (V.I.B.), Ghent University, Ledeganckstraat 35, B-9000, Ghent, Belgium
| | | | | | | | | |
Collapse
|
23
|
Abstract
Familial dysautonomia is a severe autosomal-recessive neurodegenerative disease that primarily affects the Ashkenazi Jewish population. We present the mapping of alpha-catulin and show that it maps precisely to the familial dysautonomia candidate region on 9q31. Patient sequence analysis identified two new sequence variants, which show linkage disequilibrium with this disease. A G to A transition at nucleotide 423 in exon 3 is a silent base change that does not alter the Val residue at position 141. A G to C transversion at nucleotide 1579 changes the Glu at postion 527 to Gln. These base changes were analyzed in several patients, unaffected Ashkenazi Jewish controls, and non-Jewish controls. Because of the presence of these sequence variants in several unaffected individuals, alpha-catulin is unlikely to be the causative gene in this disease.
Collapse
Affiliation(s)
- P C Demacio
- Department of Genetics, Hospital for Sick Children, University of Toronto, ON, Canada
| | | |
Collapse
|
24
|
Janssens B, Goossens S, Staes K, Gilbert B, van Hengel J, Colpaert C, Bruyneel E, Mareel M, van Roy F. αT-Catenin: a novel tissue-specific β-catenin-binding protein mediating strong cell-cell adhesion. J Cell Sci 2001; 114:3177-88. [PMID: 11590244 DOI: 10.1242/jcs.114.17.3177] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Cadherins are major cell-cell adhesion proteins whose cytoplasmic domains bind to catenin proteins. Strong intercellular adhesion depends on linkage of the cadherin/catenin complex to the actin cytoskeleton via α-catenin. To date, it is not clear how different cell types achieve the variable strength of cell-cell adhesion clearly needed in a multicellular organism. Here, we report the cloning and molecular characterization of αT(testis)-catenin, a novel human cDNA encoding a protein with homology to both human αE(epithelial)-catenin and αN(neural)-catenin. Although originally discovered in testis, αT-catenin is expressed in other tissues, the highest levels being observed in heart. Immunohistochemical analysis showed human αT-catenin localization at intercalated discs of cardiomyocytes and in peritubular myoid cells of testis. In cells transfected with αT-catenin cDNA, interaction with β-catenin was demonstrated by co-immunoprecipitation. Transfection of α-catenin-deficient colon carcinoma cells recruited E-cadherin and β-catenin to cell-cell contacts and functional cadherin-mediated cell-cell adhesion was restored in this way. Moreover, compaction of these cells was at least as prominent as in the case of cells expressing endogenous αE-catenin. We propose that αT-catenin is necessary for the formation of stretch-resistant cell-cell adhesion complexes, in particular, muscle cells.
Collapse
Affiliation(s)
- B Janssens
- Molecular Cell Biology Unit, Department of Molecular Biology, Flanders Interuniversity Institute for Biotechnology (VIB)-Ghent University, B-9000 Ghent, Belgium
| | | | | | | | | | | | | | | | | |
Collapse
|