Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
Add more filters










Publication year range
1.
Mol Biol Cell ; 32(4): 348-361, 2021 02 15.
Article in English | MEDLINE | ID: mdl-33378218

ABSTRACT

Sustained cell migration is essential for wound healing and cancer metastasis. The epidermal growth factor receptor (EGFR) signaling cascade is known to drive cell migration and proliferation. While the signal transduction downstream of EGFR has been extensively investigated, our knowledge of the initiation and maintenance of EGFR signaling during cell migration remains limited. The metalloprotease TACE (tumor necrosis factor alpha converting enzyme) is responsible for producing active EGFR family ligands in the via ligand shedding. Sustained TACE activity may perpetuate EGFR signaling and reduce a cell's reliance on exogenous growth factors. Using a cultured keratinocyte model system, we show that depletion of α-catenin perturbs adherens junctions, enhances cell proliferation and motility, and decreases dependence on exogenous growth factors. We show that the underlying mechanism for these observed phenotypical changes depends on enhanced autocrine/paracrine release of the EGFR ligand transforming growth factor alpha in a TACE-dependent manner. We demonstrate that proliferating keratinocyte epithelial cell clusters display waves of oscillatory extracellular signal-regulated kinase (ERK) activity, which can be eliminated by TACE knockout, suggesting that these waves of oscillatory ERK activity depend on autocrine/paracrine signals produced by TACE. These results provide new insights into the regulatory role of adherens junctions in initiating and maintaining autocrine/paracrine signaling with relevance to wound healing and cellular transformation.


Subject(s)
ADAM17 Protein/metabolism , Adherens Junctions/metabolism , alpha Catenin/metabolism , ADAM17 Protein/physiology , Adherens Junctions/physiology , Cell Line, Tumor , Cell Movement/physiology , Cell Proliferation , Epidermal Growth Factor/metabolism , Epithelial Cells/metabolism , ErbB Receptors/metabolism , HaCaT Cells , Humans , Metalloproteases/metabolism , Paracrine Communication/physiology , Phosphorylation , Signal Transduction , Transforming Growth Factor alpha/metabolism , alpha Catenin/physiology
2.
Elife ; 82019 12 13.
Article in English | MEDLINE | ID: mdl-31833472

ABSTRACT

During organogenesis, precise control of spindle orientation balances proliferation and differentiation. In the developing murine epidermis, planar and perpendicular divisions yield symmetric and asymmetric fate outcomes, respectively. Classically, division axis specification involves centrosome migration and spindle rotation, events occurring early in mitosis. Here, we identify a novel orientation mechanism which corrects erroneous anaphase orientations during telophase. The directionality of reorientation correlates with the maintenance or loss of basal contact by the apical daughter. While the scaffolding protein LGN is known to determine initial spindle positioning, we show that LGN also functions during telophase to reorient oblique divisions toward perpendicular. The fidelity of telophase correction also relies on the tension-sensitive adherens junction proteins vinculin, α-E-catenin, and afadin. Failure of this corrective mechanism impacts tissue architecture, as persistent oblique divisions induce precocious, sustained differentiation. The division orientation plasticity provided by telophase correction may enable progenitors to adapt to local tissue needs.


Subject(s)
Epidermal Cells/cytology , Epithelial Cells/cytology , Telophase/physiology , Actomyosin/physiology , Anaphase , Animals , Cell Self Renewal , Cell Shape , Cytoskeleton/ultrastructure , Epidermis/embryology , Female , Genes, Reporter , Intravital Microscopy , Male , Mice , Mice, Inbred C57BL , Microfilament Proteins/deficiency , Microfilament Proteins/genetics , Microfilament Proteins/physiology , Protein Conformation , RNA Interference , Spindle Apparatus/ultrastructure , Vinculin/genetics , Vinculin/physiology , alpha Catenin/genetics , alpha Catenin/physiology
3.
Mol Biol Cell ; 30(17): 2115-2123, 2019 08 01.
Article in English | MEDLINE | ID: mdl-31188702

ABSTRACT

Maintaining tissue integrity during epidermal morphogenesis depends on α-catenin, which connects the cadherin complex to F-actin. We show that the adhesion modulation domain (AMD) of Caenorhabditis elegans HMP-1/α-catenin regulates its F-actin-binding activity and organization of junctional-proximal actin in vivo. Deleting the AMD increases F-actin binding in vitro and leads to excess actin recruitment to adherens junctions in vivo. Reducing actin binding through a compensatory mutation in the C-terminus leads to improved function. Based on the effects of phosphomimetic and nonphosphorylatable mutations, phosphorylation of S509, within the AMD, may regulate F-actin binding. Taken together, these data establish a novel role for the AMD in regulating the actin-binding ability of an α-catenin and its proper function during epithelial morphogenesis.


Subject(s)
Actins/metabolism , Caenorhabditis elegans Proteins/metabolism , alpha Catenin/metabolism , Actin Cytoskeleton/metabolism , Adherens Junctions/metabolism , Animals , Cadherins/metabolism , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/physiology , Cell Adhesion/genetics , Morphogenesis/physiology , Mutation , Phosphorylation , Protein Binding/physiology , Protein Domains/physiology , alpha Catenin/physiology
4.
Development ; 145(5)2018 03 08.
Article in English | MEDLINE | ID: mdl-29467248

ABSTRACT

Shortly after birth, muscle cells of the mammalian heart lose their ability to divide. At the same time, the N-cadherin/catenin cell adhesion complex accumulates at the cell termini, creating a specialized type of cell-cell contact called the intercalated disc (ICD). To investigate the relationship between ICD maturation and proliferation, αE-catenin (Ctnna1) and αT-catenin (Ctnna3) genes were deleted to generate cardiac-specific α-catenin double knockout (DKO) mice. DKO mice exhibited aberrant N-cadherin expression, mislocalized actomyosin activity and increased cardiomyocyte proliferation that was dependent on Yap activity. To assess effects on tension, cardiomyocytes were cultured on deformable polyacrylamide hydrogels of varying stiffness. When grown on a stiff substrate, DKO cardiomyocytes exhibited increased cell spreading, nuclear Yap and proliferation. A low dose of either a myosin or RhoA inhibitor was sufficient to block Yap accumulation in the nucleus. Finally, activation of RhoA was sufficient to increase nuclear Yap in wild-type cardiomyocytes. These data demonstrate that α-catenins regulate ICD maturation and actomyosin contractility, which, in turn, control Yap subcellular localization, thus providing an explanation for the loss of proliferative capacity in the newborn mammalian heart.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cytoskeleton/metabolism , Myocardium/metabolism , Myocytes, Cardiac/metabolism , Phosphoproteins/metabolism , alpha Catenin/physiology , Adaptor Proteins, Signal Transducing/physiology , Animals , Animals, Newborn , Cell Communication/genetics , Cell Cycle Proteins , Cells, Cultured , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocytes, Cardiac/physiology , Phosphoproteins/physiology , YAP-Signaling Proteins , alpha Catenin/genetics
5.
Oncotarget ; 7(34): 55518-55528, 2016 08 23.
Article in English | MEDLINE | ID: mdl-27487124

ABSTRACT

Accumulating evidence indicates that deregulation of cancer-associated pseudogene is involved in the pathogenesis of cancer. In the study, we demonstrated that pseudogene CTNNAP1, for the CTNNA1 gene, was dysregulated in colorectal cancer and the degree of dysregulation was remarkably associated with tumor node metastasis (TNM) stage (P<0.05). The mechanistic experiments revealed that pseudogene CTNNAP1 played a pivotal role in the regulation of its cognate gene CTNNA1 by competition for microRNA-141. Moreover, gain-of-function approaches showed that overexpression of CTNNAP1 or CTNNA1 significantly inhibited cell proliferation and tumor growth in vitro and in vivo by inducing G0/G1 cell cycle arrest. Our findings add a new regulatory circuit via competing endogenous RNA (ceRNA) cross-talk between pseudogene CTNNAP1 and its cognate gene CTNNA1, and provide new insights into potential diagnostic biomarker for monitoring human colorectal cancer.


Subject(s)
Colorectal Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Genes, Tumor Suppressor/physiology , Pseudogenes/physiology , alpha Catenin/genetics , Adult , Aged , Cell Proliferation , Colorectal Neoplasms/genetics , Down-Regulation , Female , Humans , Male , MicroRNAs/physiology , Middle Aged , alpha Catenin/physiology
6.
Methods ; 94: 13-8, 2016 Feb 01.
Article in English | MEDLINE | ID: mdl-26318089

ABSTRACT

Mechanosensing of the micro-environments has been shown to be essential for cell survival, growth, differentiation and migration. The mechanosensing pathways are mediated by a set of mechanosensitive proteins located at focal adhesion and cell-cell adherens junctions as well as in the cytoskeleton network. Here we review the applications of magnetic tweezers on elucidating the molecular mechanisms of the mechanosensing proteins. The scope of this review includes the principles of the magnetic tweezers technology, theoretical analysis of force-dependent stability and interaction of mechanosensing proteins, and recent findings obtained using magnetic tweezers.


Subject(s)
Cell Adhesion Molecules/physiology , Mechanotransduction, Cellular , Cell Adhesion Molecules/chemistry , Electron Spin Resonance Spectroscopy , Extracellular Matrix/chemistry , Extracellular Matrix/physiology , Focal Adhesions/chemistry , Focal Adhesions/physiology , Magnetic Phenomena , Protein Stability , Talin/chemistry , Talin/physiology , Vinculin/chemistry , Vinculin/physiology , alpha Catenin/chemistry , alpha Catenin/physiology
7.
J Cell Biol ; 210(4): 647-61, 2015 Aug 17.
Article in English | MEDLINE | ID: mdl-26261181

ABSTRACT

The function of the actin-binding domain of α-catenin, αABD, including its possible role in the direct anchorage of the cadherin-catenin complex to the actin cytoskeleton, has remained uncertain. We identified two point mutations on the αABD surface that interfere with αABD binding to actin and used them to probe the role of α-catenin-actin interactions in adherens junctions. We found that the junctions directly bound to actin via αABD were more dynamic than the junctions bound to actin indirectly through vinculin and that recombinant αABD interacted with cortical actin but not with actin bundles. This interaction resulted in the formation of numerous short-lived cortex-bound αABD clusters. Our data suggest that αABD clustering drives the continuous assembly of transient, actin-associated cadherin-catenin clusters whose disassembly is maintained by actin depolymerization. It appears then that such actin-dependent αABD clustering is a unique molecular mechanism mediating both integrity and reassembly of the cell-cell adhesive interface formed through weak cis- and trans-intercadherin interactions.


Subject(s)
Actins/metabolism , Cadherins/metabolism , alpha Catenin/physiology , Adherens Junctions/metabolism , Cell Line, Tumor , Humans , Kinetics , Microscopy, Fluorescence , Protein Binding , Protein Interaction Domains and Motifs , Protein Transport , Time-Lapse Imaging , Vinculin/metabolism , alpha Catenin/chemistry
8.
J Biol Chem ; 290(31): 18890-903, 2015 Jul 31.
Article in English | MEDLINE | ID: mdl-26070562

ABSTRACT

α-Catenin plays a crucial role in cadherin-mediated adhesion by binding to ß-catenin, F-actin, and vinculin, and its dysfunction is linked to a variety of cancers and developmental disorders. As a mechanotransducer in the cadherin complex at intercellular adhesions, mechanical and force-sensing properties of α-catenin are critical to its proper function. Biochemical data suggest that α-catenin adopts an autoinhibitory conformation, in the absence of junctional tension, and biophysical studies have shown that α-catenin is activated in a tension-dependent manner that in turn results in the recruitment of vinculin to strengthen the cadherin complex/F-actin linkage. However, the molecular switch mechanism from autoinhibited to the activated state remains unknown for α-catenin. Here, based on the results of an aggregate of 3 µs of molecular dynamics simulations, we have identified a dynamic salt-bridge network within the core M region of α-catenin that may be the structural determinant of the stability of the autoinhibitory conformation. According to our constant-force steered molecular dynamics simulations, the reorientation of the MII/MIII subdomains under force may constitute an initial step along the transition pathway. The simulations also suggest that the vinculin-binding domain (subdomain MI) is intrinsically much less stable than the other two subdomains in the M region (MII and MIII). Our findings reveal several key insights toward a complete understanding of the multistaged, force-induced conformational transition of α-catenin to the activated conformation.


Subject(s)
alpha Catenin/chemistry , Amino Acid Sequence , Cell Adhesion , Humans , Molecular Dynamics Simulation , Molecular Sequence Data , Protein Stability , Protein Structure, Secondary , Protein Structure, Tertiary , alpha Catenin/physiology
9.
Cell Adh Migr ; 9(3): 167-74, 2015.
Article in English | MEDLINE | ID: mdl-25914082

ABSTRACT

The disproportional enlargement of the neocortex through evolution has been instrumental in the success of vertebrates, in particular mammals. The neocortex is a multilayered sheet of neurons generated from a simple proliferative neuroepithelium through a myriad of mechanisms with substantial evolutionary conservation. This developing neuroepithelium is populated by progenitors that can generate additional progenitors as well as post-mitotic neurons. Subtle alterations in the production of progenitors vs. differentiated cells during development can result in dramatic differences in neocortical size. This review article will examine how cadherin adhesion proteins, in particular α-catenin and N-cadherin, function in regulating the neural progenitor microenvironment, cell proliferation, and differentiation in cortical development.


Subject(s)
Adherens Junctions/physiology , Cadherins/physiology , Neocortex/growth & development , alpha Catenin/physiology , beta Catenin/physiology , Animals , Cell Differentiation , Cell Proliferation , Cellular Microenvironment , Humans , Mice , Neurons/physiology , Signal Transduction
11.
Circ Res ; 116(1): 70-9, 2015 Jan 02.
Article in English | MEDLINE | ID: mdl-25305307

ABSTRACT

RATIONALE: Shortly after birth, muscle cells of the mammalian heart lose their ability to divide. Thus, they are unable to effectively replace dying cells in the injured heart. The recent discovery that the transcriptional coactivator Yes-associated protein (Yap) is necessary and sufficient for cardiomyocyte proliferation has gained considerable attention. However, the upstream regulators and signaling pathways that control Yap activity in the heart are poorly understood. OBJECTIVE: To investigate the role of α-catenins in the heart using cardiac-specific αE- and αT-catenin double knockout mice. METHODS AND RESULTS: We used 2 cardiac-specific Cre transgenes to delete both αE-catenin (Ctnna1) and αT-catenin (Ctnna3) genes either in the perinatal or in the adult heart. Perinatal depletion of α-catenins increased cardiomyocyte number in the postnatal heart. Increased nuclear Yap and the cell cycle regulator cyclin D1 accompanied cardiomyocyte proliferation in the α-catenin double knockout hearts. Fetal genes were increased in the α-catenin double knockout hearts indicating a less mature cardiac gene expression profile. Knockdown of α-catenins in neonatal rat cardiomyocytes also resulted in increased proliferation, which could be blocked by knockdown of Yap. Finally, inactivation of α-catenins in the adult heart using an inducible Cre led to increased nuclear Yap and cardiomyocyte proliferation and improved contractility after myocardial infarction. CONCLUSIONS: These studies demonstrate that α-catenins are critical regulators of Yap, a transcriptional coactivator essential for cardiomyocyte proliferation. Furthermore, we provide proof of concept that inhibiting α-catenins might be a useful strategy to promote myocardial regeneration after injury.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cell Proliferation/physiology , Myocytes, Cardiac/metabolism , Phosphoproteins/metabolism , alpha Catenin/physiology , Animals , Animals, Newborn , Cell Cycle Proteins , Cells, Cultured , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Rats , YAP-Signaling Proteins
12.
Annu Rev Cell Dev Biol ; 30: 291-315, 2014.
Article in English | MEDLINE | ID: mdl-25062360

ABSTRACT

Cadherins are the principal adhesion proteins at intercellular junctions and function as the biochemical Velcro that binds cells together. Besides this mechanical function, cadherin complexes are also mechanotransducers that sense changes in tension and trigger adaptive reinforcement of intercellular junctions. The assembly and regulation of cadherin adhesions are central to their mechanical functions, and new evidence is presented for a comprehensive model of cadherin adhesion, which is surprisingly more complex than previously appreciated. Recent findings also shed new light on mechanisms that regulate cadherin junction assembly, adhesion, and mechanotransduction. We further describe recent evidence for cadherin-based mechanotransduction, and the rudiments of the molecular mechanism, which involves α-catenin and vinculin as key elements. Potential roles of a broader cast of possible force-sensitive partners are considered, as well as known and speculative biological consequences of adhesion and force transduction at cadherin-mediated junctions.


Subject(s)
Cadherins/physiology , Cell Adhesion/physiology , Mechanotransduction, Cellular/physiology , Actins/physiology , Allosteric Regulation , Animals , Cadherins/chemistry , Cytoskeleton/physiology , Endocytosis , Glycosylation , Humans , Models, Biological , Models, Molecular , Morphogenesis , Protein Processing, Post-Translational , Protein Structure, Tertiary , Signal Transduction , Structure-Activity Relationship , Vinculin/physiology , alpha Catenin/physiology
13.
Toxicol Sci ; 141(1): 254-62, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24973089

ABSTRACT

Cisplatin is one of the most potent and widely used antitumor drugs. However, the use of cisplatin is limited by its side effect, nephrotoxicity. Evidence has shown an increased incidence and severity of acute kidney injury (AKI) in the elderly. Previous studies from our laboratory demonstrate a decrease in α(E)-catenin expression in aged kidney. In this study, we investigated whether the loss of α(E)-catenin may increase cisplatin nephrotoxicity. To study the effects of reduced α(E)-catenin, a cell line with stable knockdown of α(E)-catenin (C2 cells) was used; NT3 is nontargeted control. C2 cells exhibited a significant loss of viability as determined by MTT assay compared with NT3 cells after cisplatin challenge, but showed no difference in lactate dehydrogenase (LDH) leakage. Increased caspase 3/7 activation and PARP cleavage was observed in C2 cells after cisplatin treatment. Z-VAD, a pan-caspase inhibitor, abolished the difference in susceptibility between NT3 and C2 cells. Interestingly, the expression of α(E)-catenin was further decreased after cisplatin treatment. Furthermore, in vivo data demonstrated a significant increase in serum creatinine at 72 h after a single dose of cisplatin in 24-month-old rats, but not in 4-month-old rats. Increased expression of KIM-1 and in situ apoptosis were also detected in aged kidney after cisplatin challenge. Taken together, these data suggest that loss of α(E)-catenin increases apoptosis of tubular epithelial cells which may contribute to the increased nephrotoxicity induced by cisplatin in aged kidney.


Subject(s)
Antineoplastic Agents/toxicity , Apoptosis/drug effects , Cisplatin/toxicity , Epithelial Cells/drug effects , Kidney Diseases/chemically induced , Kidney Tubules, Proximal/drug effects , alpha Catenin/physiology , Aging/metabolism , Aging/pathology , Animals , Blotting, Western , Cell Culture Techniques , Cell Line , Cell Survival/drug effects , Epithelial Cells/metabolism , Epithelial Cells/pathology , Gene Knockdown Techniques , Kidney Diseases/genetics , Kidney Diseases/pathology , Kidney Tubules, Proximal/metabolism , Kidney Tubules, Proximal/pathology , Male , Rats, Inbred F344 , Real-Time Polymerase Chain Reaction , alpha Catenin/genetics
14.
Proc Natl Acad Sci U S A ; 111(14): 5260-5, 2014 Apr 08.
Article in English | MEDLINE | ID: mdl-24706864

ABSTRACT

α-Catenin (α-cat) is an actin-binding protein required for cell-cell cohesion. Although this adhesive function for α-cat is well appreciated, cells contain a substantial amount of nonjunctional α-cat that may be used for other functions. We show that α-cat is a nuclear protein that can interact with ß-catenin (ß-cat) and T-cell factor (TCF) and that the nuclear accumulation of α-cat depends on ß-cat. Using overexpression, knockdown, and chromatin immunoprecipitation approaches, we show that α-cat attenuates Wnt/ß-cat-responsive genes in a manner that is downstream of ß-cat/TCF loading on promoters. Both ß-cat- and actin-binding domains of α-cat are required to inhibit Wnt signaling. A nuclear-targeted form of α-cat induces the formation of nuclear filamentous actin, whereas cells lacking α-cat show altered nuclear actin properties. Formation of nuclear actin filaments correlates with reduced RNA synthesis and altered chromatin organization. Conversely, nuclear extracts made from cells lacking α-cat show enhanced general transcription in vitro, an activity that can be partially rescued by restoring the C-terminal actin-binding region of α-cat. These data demonstrate that α-cat may limit gene expression by affecting nuclear actin organization.


Subject(s)
Transcription, Genetic/physiology , alpha Catenin/physiology , Cell Line, Tumor , Humans , Signal Transduction
15.
J Cell Sci ; 127(Pt 8): 1779-91, 2014 Apr 15.
Article in English | MEDLINE | ID: mdl-24522187

ABSTRACT

The findings presented here demonstrate the role of α-catenin in cadherin-based adhesion and mechanotransduction in different mechanical contexts. Bead-twisting measurements in conjunction with imaging, and the use of different cell lines and α-catenin mutants reveal that the acute local mechanical manipulation of cadherin bonds triggers vinculin and actin recruitment to cadherin adhesions in an actin- and α-catenin-dependent manner. The modest effect of α-catenin on the two-dimensional binding affinities of cell surface cadherins further suggests that force-activated adhesion strengthening is due to enhanced cadherin-cytoskeletal interactions rather than to α-catenin-dependent affinity modulation. Complementary investigations of cadherin-based rigidity sensing also suggest that, although α-catenin alters traction force generation, it is not the sole regulator of cell contractility on compliant cadherin-coated substrata.


Subject(s)
Cadherins/blood , Cadherins/physiology , Cell Adhesion , Mechanotransduction, Cellular , alpha Catenin/physiology , Actins/metabolism , Animals , Binding Sites , Biomechanical Phenomena , Cadherins/chemistry , Cell Line, Tumor , Dogs , Erythrocytes/metabolism , Humans , Kinetics , Madin Darby Canine Kidney Cells , Protein Interaction Domains and Motifs , Protein Transport , Vinculin/metabolism
16.
Nat Cell Biol ; 16(3): 245-54, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24509793

ABSTRACT

Basal-like breast cancer is a highly aggressive tumour subtype associated with poor prognosis. Aberrant activation of NF-κB signalling is frequently found in triple-negative basal-like breast cancer cells, but the cause of this activation has remained elusive.Here we report that α-catenin functions as a tumour suppressor in E-cadherin-negative basal-like breast cancer cells by inhibiting NF-κB signalling. Mechanistically, α-catenin interacts with the IκBα protein, and stabilizes IκBα by inhibiting its ubiquitylation and its association with the proteasome. This stabilization in turn prevents nuclear localization of RelA and p50, leading to decreased expression of TNF-α, IL-8 and RelB. In human breast cancer, CTNNA1 expression is specifically downregulated in the basal-like subtype, correlates with clinical outcome and inversely correlates with TNF and RELB expression. Taken together, these results uncover a previously undescribed mechanism by which the NF-κB pathway is activated in E-cadherin-negative basal-like breast cancer.


Subject(s)
Breast Neoplasms/metabolism , Cadherins/metabolism , Neoplasms, Basal Cell/metabolism , Transcription Factor RelB/genetics , alpha Catenin/physiology , Active Transport, Cell Nucleus , Animals , Antigens, CD , Base Sequence , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Nucleus/metabolism , Cell Proliferation , Down-Regulation , Female , Humans , I-kappa B Proteins/metabolism , Mice , Mice, Nude , Molecular Sequence Data , NF-KappaB Inhibitor alpha , NF-kappa B p50 Subunit/metabolism , Neoplasm Transplantation , Neoplasms, Basal Cell/pathology , Promoter Regions, Genetic , Proteasome Endopeptidase Complex/metabolism , Protein Stability , Signal Transduction , Transcription Factor RelA/metabolism , Transcription Factor RelB/metabolism , Tumor Burden , Tumor Suppressor Proteins/physiology , Ubiquitination
17.
Anim Reprod Sci ; 140(3-4): 189-94, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23820070

ABSTRACT

In previous research, several WNT signaling pathway genes including transcription factor 12 (TCF12), catenin alpha-like protein 1 (CTNNAL1) and wingless-type MMTV integration site family, member 10B (WNT10B) were differentially expressed in PMSG-hCG stimulated preovulatory ovarian follicles of Large White and Chinese Taihu sows. In the present research, these three genes were selected as the candidate genes for litter size traits in pigs. Four mutations (TCF12 c.-201+65 G>A, TCF12 c.-200-300 G>A, CTNNAL1 c.1878 G>C and WNT10B c.*12 C>T) were detected in eleven pig populations, and results indicated CTNNAL1 c.1878 G and WNT10B c.*12 C were the major alleles in all tested pig populations, while TCF12 c.-201+65 A and TCF12 c.-200-300 A were the major alleles in several Chinese native pig breeds. Association analysis of four mutations with litter size in Large White and DIV pigs showed that both the signficant differences of total number born (TNB) and number born alive (NBA) among three genotypes and the significance of additive effects appeared at TCF12 c.-200-300 G>A and CTNNAL1 c.1878 G>C loci, suggesting these two mutations might be reliable markers for pig selection and breeding.


Subject(s)
Animals, Newborn/physiology , Basic Helix-Loop-Helix Transcription Factors/physiology , Polymorphism, Single Nucleotide/physiology , Swine/physiology , Wnt Proteins/physiology , alpha Catenin/physiology , Animals , Animals, Newborn/genetics , Basic Helix-Loop-Helix Transcription Factors/genetics , DNA/chemistry , DNA/genetics , Female , Genotype , Linear Models , Litter Size/genetics , Litter Size/physiology , Ovarian Follicle/physiology , Polymerase Chain Reaction/veterinary , Polymorphism, Restriction Fragment Length , Polymorphism, Single Nucleotide/genetics , Pregnancy , Quantitative Trait Loci/genetics , Quantitative Trait Loci/physiology , Swine/genetics , Wnt Proteins/genetics , alpha Catenin/genetics
18.
PLoS One ; 8(1): e55069, 2013.
Article in English | MEDLINE | ID: mdl-23359820

ABSTRACT

Cancer cell invasion is the critical first step of metastasis, yet, little is known about how cancer cells invade and initiate metastasis in a complex extracellular matrix. Using a cell line from bone metastasis of prostate cancer (PC3), we analyzed how prostate cancer cells migrate in a physiologically relevant 3D Matrigel. We found that PC3 cells migrated more efficiently as multi-cellular clusters than isolated single cells, suggesting that the presence of cell-cell adhesion improves 3D cell migration. Perturbation of N-cadherin function by transfection of either the N-cadherin cytoplasmic domain or shRNA specific to N-cadherin abolished collective cell migration. Interestingly, PC3 cells do not express α-catenin, an actin binding protein in the cadherin complex. When the full-length α-catenin was re-introduced, the phenotype of PC3 cells reverted back to a more epithelial phenotype with a decreased cell migration rate in 3D Matrigel. Interestingly, we found that the N-terminal half of α-catenin was sufficient to suppress invasive phenotype. Taken together, these data suggest that the formation of N-cadherin junctions promotes 3D cell migration of prostate cancer cells, and this is partly due to an aberrant regulation of the N-cadherin complex in the absence of α-catenin.


Subject(s)
Cadherins/physiology , Prostatic Neoplasms/pathology , alpha Catenin/physiology , Cell Line, Tumor , Humans , Male , Prostatic Neoplasms/metabolism , alpha Catenin/metabolism
19.
Placenta ; 33(7): 554-60, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22534068

ABSTRACT

Transcriptional activation of CTNNA3, encoding αT-catenin, by the Y153H mutated form of the human STOX1 transcription factor was proposed to be responsible for altered fetal trophoblast invasion into the maternal endometrium during placentation in pre-eclampsia. Here we have generated a mouse model to investigate the in vivo effects of ectopic αT-catenin expression on trophoblast invasion. Histological analysis was used to determine the invasive capacities of trophoblasts from transgenic embryos, as well as proliferation rates of spongiotrophoblasts in the junctional zone. Augmented expression of αT-catenin reduced the number of invading trophoblasts but did not cause embryonic mortality. The, αT-catenin positive cells could still invade into the decidual layer and migrated as deeply as wild-type trophoblasts. Furthermore, the junctional zone is enlarged in placentas of mice overexpressing αT-catenin due to hyperproliferation of the residing spongiotrophoblasts, suggesting a pivotal role of αT-catenin levels in the control of the proliferative versus invasive state of trophoblasts during placentation. Our study provides, for the first time, in vivo data on the effects of increased levels of αT-catenin in the placenta.


Subject(s)
Placentation/physiology , Trophoblasts/physiology , alpha Catenin/genetics , Animals , Cell Proliferation , Embryo, Mammalian/metabolism , Female , Gene Expression , Heterozygote , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation , Placenta/chemistry , Placenta/cytology , Placenta/metabolism , Pregnancy , Proteins/genetics , RNA, Untranslated , Trophoblasts/cytology , alpha Catenin/analysis , alpha Catenin/physiology
20.
Biol Reprod ; 86(5): 141, 1-10, 2012 May.
Article in English | MEDLINE | ID: mdl-22378759

ABSTRACT

CDH1 is a cell-cell adhesion molecule expressed in the epithelium to coordinate key morphogenetic processes, establish cell polarity, and regulate epithelial differentiation and proliferation. To determine the role of CDH1 in the mouse uterus, Cdh1 was conditionally ablated by crossing Pgr-Cre and Cdh1-flox mice, and the phenotype was characterized. We found that loss of Cdh1 results in a disorganized cellular structure of the epithelium and ablation of endometrial glands in the neonatal uterus. Cdh1(d/d) mice lost adherens junctions (CTNNB1 and CTNNA1) and tight junctions (claudin, occludin, and ZO-1 proteins) in the neonatal uterus, leading to loss of epithelial cell-cell interaction. Ablation of Cdh1 induced abnormal epithelial proliferation and massive apoptosis, and disrupted Wnt and Hox gene expression in the neonatal uterus. Although the uteri of Cdh1(d/d) mice did not show any myometrial defects, ablation of Cdh1 inhibited expression of epithelial (cytokeratin 8) and stromal (CD10) markers. Cdh1(d/d) mice were infertile because of defects during implantation and decidualization. Furthermore, we showed in the model of conditional ablation of both Cdh1 and Trp53 in the uterus that interrupting cell cycle regulation through the loss of Cdh1 leads to abnormal uterine development. The uteri of Cdh1(d/d) Trp53(d/d) mice exhibited histological features of endometrial carcinomas with myometrial invasion. Collectively, these findings suggest that CDH1 has an important role in structural and functional development of the uterus as well as adult uterine function. CDH1 has a capacity to control cell fate by altering directional cell proliferation and apoptosis.


Subject(s)
Cell Cycle Proteins/physiology , Cell Differentiation/physiology , Endometrium/growth & development , Uterus/physiology , Adherens Junctions , Animals , Apoptosis/physiology , Carcinoma/genetics , Carcinoma/pathology , Carcinoma/physiopathology , Cdh1 Proteins , Cell Cycle Proteins/genetics , Cell Proliferation , Claudins/physiology , Endometrial Neoplasms/genetics , Endometrial Neoplasms/pathology , Endometrial Neoplasms/physiopathology , Endometrium/cytology , Endometrium/physiology , Female , Gene Expression Regulation , Homeodomain Proteins/physiology , Keratin-8/biosynthesis , Membrane Proteins/physiology , Mice , Mice, Knockout , Neprilysin/biosynthesis , Occludin , Phosphoproteins/physiology , Tight Junctions , Tumor Suppressor Protein p53/biosynthesis , Wnt Signaling Pathway/physiology , Zonula Occludens-1 Protein , alpha Catenin/physiology , beta Catenin/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...