Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Development ; 151(7)2024 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-38456551

RESUMO

Adhesion between stem cells and their niche provides stable anchorage and signaling cues to sustain properties such as quiescence. Skeletal muscle stem cells (MuSCs) adhere to an adjacent myofiber via cadherin-catenin complexes. Previous studies on N- and M-cadherin in MuSCs revealed that although N-cadherin is required for quiescence, they are collectively dispensable for MuSC niche localization and regenerative activity. Although additional cadherins are expressed at low levels, these findings raise the possibility that cadherins are unnecessary for MuSC anchorage to the niche. To address this question, we conditionally removed from MuSCs ß- and γ-catenin, and, separately, αE- and αT-catenin, factors that are essential for cadherin-dependent adhesion. Catenin-deficient MuSCs break quiescence similarly to N-/M-cadherin-deficient MuSCs, but exit the niche and are depleted. Combined in vivo, ex vivo and single cell RNA-sequencing approaches reveal that MuSC attrition occurs via precocious differentiation, re-entry to the niche and fusion to myofibers. These findings indicate that cadherin-catenin-dependent adhesion is required for anchorage of MuSCs to their niche and for preservation of the stem cell compartment. Furthermore, separable cadherin-regulated functions govern niche localization, quiescence and MuSC maintenance.


Assuntos
Caderinas , Nicho de Células-Tronco , Nicho de Células-Tronco/genética , Caderinas/genética , Caderinas/metabolismo , Fibras Musculares Esqueléticas/metabolismo , Transdução de Sinais , Cateninas/genética , Cateninas/metabolismo , Músculo Esquelético/metabolismo , Adesão Celular/genética
2.
Development ; 150(2)2023 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-36621002

RESUMO

The cardiomyocyte phenotypic switch from a proliferative to terminally differentiated state results in the loss of regenerative potential of the mammalian heart shortly after birth. Nonmuscle myosin IIB (NM IIB)-mediated actomyosin contractility regulates cardiomyocyte cytokinesis in the embryonic heart, and NM IIB levels decline after birth, suggesting a role for cellular tension in the regulation of cardiomyocyte cell cycle activity in the postnatal heart. To investigate the role of actomyosin contractility in cardiomyocyte cell cycle arrest, we conditionally activated ROCK2 kinase domain (ROCK2:ER) in the murine postnatal heart. Here, we show that α5/ß1 integrin and fibronectin matrix increase in response to actomyosin-mediated tension. Moreover, activation of ROCK2:ER promotes nuclear translocation of Yap, a mechanosensitive transcriptional co-activator, and enhances cardiomyocyte proliferation. Finally, we show that reduction of myocardial α5 integrin rescues the myocardial proliferation phenotype in ROCK2:ER hearts. These data demonstrate that cardiomyocytes respond to increased intracellular tension by altering their intercellular contacts in favor of cell-matrix interactions, leading to Yap nuclear translocation, thus uncovering a function for nonmuscle myosin contractility in promoting cardiomyocyte proliferation in the postnatal heart.


Assuntos
Actomiosina , Integrina alfa5 , Animais , Camundongos , Citoesqueleto de Actina/metabolismo , Actomiosina/metabolismo , Proliferação de Células , Integrina alfa5/metabolismo , Mamíferos/metabolismo , Miócitos Cardíacos/metabolismo , Fatores de Transcrição/metabolismo
3.
Proc Natl Acad Sci U S A ; 116(31): 15560-15569, 2019 07 30.
Artigo em Inglês | MEDLINE | ID: mdl-31300538

RESUMO

The roles of cellular orientation during trabecular and ventricular wall morphogenesis are unknown, and so are the underlying mechanisms that regulate cellular orientation. Myocardial-specific Numb and Numblike double-knockout (MDKO) hearts display a variety of defects, including in cellular orientation, patterns of mitotic spindle orientation, trabeculation, and ventricular compaction. Furthermore, Numb- and Numblike-null cardiomyocytes exhibit cellular behaviors distinct from those of control cells during trabecular morphogenesis based on single-cell lineage tracing. We investigated how Numb regulates cellular orientation and behaviors and determined that N-cadherin levels and membrane localization are reduced in MDKO hearts. To determine how Numb regulates N-cadherin membrane localization, we generated an mCherry:Numb knockin line and found that Numb localized to diverse endocytic organelles but mainly to the recycling endosome. Consistent with this localization, cardiomyocytes in MDKO did not display defects in N-cadherin internalization but rather in postendocytic recycling to the plasma membrane. Furthermore, N-cadherin overexpression via a mosaic model partially rescued the defects in cellular orientation and trabeculation of MDKO hearts. Our study unravels a phenomenon that cardiomyocytes display spatiotemporal cellular orientation during ventricular wall morphogenesis, and its disruption leads to abnormal trabecular and ventricular wall morphogenesis. Furthermore, we established a mechanism by which Numb modulates cellular orientation and consequently trabecular and ventricular wall morphogenesis by regulating N-cadherin recycling to the plasma membrane.


Assuntos
Caderinas/metabolismo , Ventrículos do Coração/embriologia , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas de Membrana/metabolismo , Miócitos Cardíacos/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Organogênese , Animais , Caderinas/genética , Peptídeos e Proteínas de Sinalização Intracelular/genética , Proteínas de Membrana/genética , Camundongos , Camundongos Knockout , Miócitos Cardíacos/citologia , Proteínas do Tecido Nervoso/genética
4.
Development ; 145(5)2018 03 08.
Artigo em Inglês | MEDLINE | ID: mdl-29467248

RESUMO

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.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Citoesqueleto/metabolismo , Miocárdio/metabolismo , Miócitos Cardíacos/metabolismo , Fosfoproteínas/metabolismo , alfa Catenina/fisiologia , Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Animais , Animais Recém-Nascidos , Comunicação Celular/genética , Proteínas de Ciclo Celular , Células Cultivadas , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Miócitos Cardíacos/fisiologia , Fosfoproteínas/fisiologia , Proteínas de Sinalização YAP , alfa Catenina/genética
5.
Cell Rep ; 21(8): 2236-2250, 2017 Nov 21.
Artigo em Inglês | MEDLINE | ID: mdl-29166613

RESUMO

Many adult stem cells display prolonged quiescence, promoted by cues from their niche. Upon tissue damage, a coordinated transition to the activated state is required because non-physiological breaks in quiescence often lead to stem cell depletion and impaired regeneration. Here, we identify cadherin-mediated adhesion and signaling between muscle stem cells (satellite cells [SCs]) and their myofiber niche as a mechanism that orchestrates the quiescence-to-activation transition. Conditional removal of N-cadherin and M-cadherin in mice leads to a break in SC quiescence, with long-term expansion of a regeneration-proficient SC pool. These SCs have an incomplete disruption of the myofiber-SC adhesive junction and maintain niche residence and cell polarity, yet show properties of SCs in a state of transition from quiescence toward full activation. Among these is nuclear localization of ß-catenin, which is necessary for this phenotype. Injury-induced perturbation of niche adhesive junctions is therefore a likely first step in the quiescence-to-activation transition.


Assuntos
Caderinas/metabolismo , Músculo Esquelético/citologia , Células Satélites de Músculo Esquelético/citologia , Células-Tronco/citologia , Animais , Divisão Celular/fisiologia , Polaridade Celular/fisiologia , Proliferação de Células/fisiologia , Camundongos , Regeneração/fisiologia , Transdução de Sinais/fisiologia
6.
Dev Biol ; 428(1): 118-134, 2017 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-28552735

RESUMO

Tissue development and regeneration involve high-ordered morphogenetic processes that are governed by elements of the cytoskeleton in conjunction with cell adhesion molecules. Such processes are particularly important in the lens whose structure dictates its function. Studies of our lens-specific N-cadherin conditional knockout mouse (N-cadcKO) revealed an essential role for N-cadherin in the migration of the apical tips of differentiating lens fiber cells along the apical surfaces of the epithelium, a region termed the Epithelial Fiber Interface (EFI), that is necessary for normal fiber cell elongation and the morphogenesis. Studies of the N-cadcKO lens suggest that N-cadherin function in fiber cell morphogenesis is linked to the activation of Rac1 and myosin II, both signaling pathways central to the regulation of cell motility including determining the directionality of cellular movement. The absence of N-cadherin did not disrupt lateral contacts between fiber cells during development, and the maintenance of Aquaporin-0 and increased expression of EphA2 at cell-cell interfaces suggests that these molecules may function in this role. E-cadherin was maintained in newly differentiating fiber cells without interfering with expression of lens-specific differentiation proteins but was not able to replace N-cadherin function in these cells. The dependence of migration of the fiber cell apical domains along the EFI for lens morphogenesis on N-cadherin provides new insight into the process of tissue development.


Assuntos
Caderinas/metabolismo , Diferenciação Celular/fisiologia , Células Epiteliais/citologia , Cristalino/embriologia , Morfogênese/fisiologia , Animais , Aquaporinas/metabolismo , Caderinas/genética , Movimento Celular/genética , Ativação Enzimática , Epitélio/fisiologia , Proteínas do Olho/metabolismo , Cristalino/citologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Miosina Tipo II/metabolismo , Neuropeptídeos/metabolismo , Receptor EphA2/biossíntese , Proteínas rac1 de Ligação ao GTP/metabolismo
7.
Cell Rep ; 10(9): 1477-1486, 2015 Mar 10.
Artigo em Inglês | MEDLINE | ID: mdl-25753414

RESUMO

In contrast to the accepted pro-proliferative effect of cell-matrix adhesion, the proliferative effect of cadherin-mediated cell-cell adhesion remains unresolved. Here, we studied the effect of N-cadherin on cell proliferation in the vasculature. We show that N-cadherin is induced in smooth muscle cells (SMCs) in response to vascular injury, an in vivo model of tissue stiffening and proliferation. Complementary experiments performed with deformable substrata demonstrated that stiffness-mediated activation of a focal adhesion kinase (FAK)-p130Cas-Rac signaling pathway induces N-cadherin. Additionally, by culturing paired and unpaired SMCs on microfabricated adhesive islands of different areas, we found that N-cadherin relaxes the spreading requirement for SMC proliferation. In vivo SMC deletion of N-cadherin strongly reduced injury-induced cycling. Finally, SMC-specific deletion of FAK inhibited proliferation after vascular injury, and this was accompanied by reduced induction of N-cadherin. Thus, a stiffness- and FAK-dependent induction of N-cadherin connects cell-matrix to cell-cell adhesion and regulates the degree of cell spreading needed for cycling.

8.
Cell Tissue Res ; 360(3): 773-83, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25673211

RESUMO

Strong cell-cell adhesion mediated by adherens junctions is dependent on anchoring the transmembrane cadherin molecule to the underlying actin cytoskeleton. To do this, the cadherin cytoplasmic domain interacts with catenin proteins, which include α-catenin that binds directly to filamentous actin. Originally thought to be a static structure, the connection between the cadherin/catenin adhesion complex and the actin cytoskeleton is now considered to be dynamic and responsive to both intercellular and intracellular signals. Alpha-catenins are mechanosensing proteins that undergo conformational change in response to cytoskeletal tension thus modifying the linkage between the cadherin and the actin cytoskeleton. There are three α-catenin isoforms expressed in mouse and human: αE-catenin (CTNNA1), αN-catenin (CTNNA2) and αT-catenin (CTNNA3). This review summarizes recent progress in understanding the in vivo function(s) of α-catenins in tissue morphogenesis, homeostasis and disease. The role of α-catenin in the regulation of cellular proliferation will be discussed in the context of cancer and regeneration.


Assuntos
Saúde , Coração/fisiologia , Neoplasias/metabolismo , Regeneração , alfa Catenina/metabolismo , Animais , Humanos , Modelos Biológicos
9.
Circ Res ; 116(1): 70-9, 2015 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-25305307

RESUMO

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.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proliferação de Células/fisiologia , Miócitos Cardíacos/metabolismo , Fosfoproteínas/metabolismo , alfa Catenina/fisiologia , Animais , Animais Recém-Nascidos , Proteínas de Ciclo Celular , Células Cultivadas , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Ratos , Proteínas de Sinalização YAP
10.
Cell Commun Adhes ; 21(3): 169-79, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24766605

RESUMO

Intercellular adhesive junctions are essential for maintaining the physical integrity of tissues; this is particularly true for the heart that is under constant mechanical load. The correct functionality of the heart is dependent on the electrical and mechanical coordination of its constituent cardiomyocytes. The intercalated disc (ID) structure located at the termini of the rod-shaped adult cardiomyocyte contains various junctional proteins responsible for the integration of structural information and cell-cell communication. According to the classical description, the ID consists of three distinct junctional complexes: adherens junction (AJ), desmosome (Des), and gap junction (GJ) that work together to mediate mechanical and electrical coupling of cardiomyocytes. However, recent morphological and molecular studies indicate that AJ and Des components are capable of mixing together resulting in a "hybrid adhering junction" or "area composita." This review summarizes recent progress in understanding the in vivo function(s) of AJ components in cardiac homeostasis and disease.


Assuntos
Caderinas/metabolismo , Cateninas/metabolismo , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Junções Aderentes/metabolismo , Animais , Humanos
11.
Prog Mol Biol Transl Sci ; 116: 263-89, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23481199

RESUMO

Of the 20 classical cadherin subtypes identified in mammals, the functions of the two initially identified family members E- (epithelial) and N- (neural) cadherin have been most extensively studied. E- and N-Cadherin have mostly mutually exclusive expression patterns, with E-cadherin expressed primarily in epithelial cells, whereas N-cadherin is found in a variety of cells, including neural, muscle, and mesenchymal cells. N-Cadherin function, in particular, appears to be cell context-dependent, as it can mediate strong cell-cell adhesion in the heart but induces changes in cell behavior in favor of a migratory phenotype in the context of epithelial-mesenchymal transition (EMT). The ability of tumor cells to alter their cadherin expression profile, for example, E- to N-cadherin, is critical for malignant progression. Recent advances in mouse molecular genetics, and specifically tissue-specific knockout and knockin alleles of N-cadherin, have provided some unexpected results. This chapter highlights some of the genetic studies that explored the complex role of N-cadherin in embryonic development and disease.


Assuntos
Caderinas/metabolismo , Adesão Celular/fisiologia , Desenvolvimento Embrionário/genética , Regulação da Expressão Gênica no Desenvolvimento , Transdução de Sinais , Animais , Progressão da Doença , Camundongos
12.
Cell Signal ; 25(1): 93-100, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23022961

RESUMO

Plakoglobin (PG, γ-Catenin, JUP), a member of the armadillo protein family and close homolog of ß-catenin, functions to link cell surface cadherin molecules with the cytoskeleton. PG is the only junctional component found in both desmosomes and adherens junctions and thus plays a critical role in the regulation of cell-cell adhesion. Similar to ß-catenin, PG is able to interact with components of the Wnt signaling pathway and directly affect gene expression by binding with LEF/TCF transcription factors. In addition, it has been proposed that PG functions primarily as a competitive inhibitor of ß-catenin transcriptional activity by sequestering LEF/TCF. Compared to ß-catenin, the contribution of PG as a transcriptional regulator in either physiological or pathological conditions is poorly understood. There is increasing clinical interest in PG as both a structural protein as well as a signaling molecule as mutations have been identified in the human PG gene that cause Arrhythmogenic Right Ventricular Cardiomyopathy (ARVC) and cutaneous syndromes. This review will discuss the connection between altered cell adhesion and gene expression and its contribution to disease pathogenesis.


Assuntos
Proteínas do Domínio Armadillo/metabolismo , Adesão Celular , Miocárdio/metabolismo , Animais , Proteínas do Domínio Armadillo/química , Proteínas do Domínio Armadillo/genética , Displasia Arritmogênica Ventricular Direita/metabolismo , Displasia Arritmogênica Ventricular Direita/patologia , Regulação da Expressão Gênica , Humanos , Transdução de Sinais , beta Catenina/química , beta Catenina/metabolismo , gama Catenina/química , gama Catenina/genética , gama Catenina/metabolismo
13.
Genesis ; 50(10): 717-27, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22522917

RESUMO

Mutations in the human Jup gene cause arrhythmogenic right ventricular cardiomyopathy (ARVC), a heart muscle disease that often leads to sudden cardiac death. Inactivation of the murine Jup gene (also known as plakoglobin) results in embryonic lethality due to cardiac rupture. In an effort to generate a conditional knockout allele, a neomycin cassette was introduced into the murine plakoglobin (PG) gene. This allele (PG F(N)) functions as a hypomorph when combined with a null allele (PG Δ). About half of the PG F(N)/Δ animals were smaller than their littermates and died before weaning age, whereas the remaining PG F(N)/Δ animals survived. Despite the reduced levels of PG in the heart, there were no signs of cardiomyopathy or cardiac dysfunction as determined by echocardiography. Importantly, the PG homolog, ß-catenin (CTNNB1), was increased in the PG F(N)/Δ hearts. In addition to its structural role as part of the N-cadherin/catenin adhesion complex, ß-catenin is a downstream effector of Wnt signaling. However, no change in ß-catenin/TCF reporter activity was observed in PG F(N)/Δ embryos suggesting that excess ß-catenin was not likely causing increased transcription of Wnt/ß-catenin target genes. These data suggest novel function(s) for PG beyond the heart and define a critical threshold of PG expression that is necessary for postnatal survival.


Assuntos
Alelos , Miocárdio/metabolismo , gama Catenina/genética , Animais , Displasia Arritmogênica Ventricular Direita/genética , Caderinas/genética , Caderinas/metabolismo , Deleção de Genes , Regulação da Expressão Gênica no Desenvolvimento , Genes Letais , Coração/anatomia & histologia , Coração/crescimento & desenvolvimento , Coração/fisiopatologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Transcrição Gênica , Via de Sinalização Wnt , beta Catenina/genética , beta Catenina/metabolismo , gama Catenina/metabolismo
14.
J Cell Sci ; 125(Pt 4): 1058-67, 2012 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-22421363

RESUMO

It is generally accepted that the intercalated disc (ICD) required for mechano-electrical coupling in the heart consists of three distinct junctional complexes: adherens junctions, desmosomes and gap junctions. However, recent morphological and molecular data indicate a mixing of adherens junctional and desmosomal components, resulting in a 'hybrid adhering junction' or 'area composita'. The α-catenin family member αT-catenin, part of the N-cadherin-catenin adhesion complex in the heart, is the only α-catenin that interacts with the desmosomal protein plakophilin-2 (PKP2). Thus, it has been postulated that αT-catenin might serve as a molecular integrator of the two adhesion complexes in the area composita. To investigate the role of αT-catenin in the heart, gene targeting technology was used to delete the Ctnna3 gene, encoding αT-catenin, in the mouse. The αT-catenin-null mice are viable and fertile; however, the animals exhibit progressive cardiomyopathy. Adherens junctional and desmosomal proteins were unaffected by loss of αT-catenin, with the exception of the desmosomal protein PKP2. Immunogold labeling at the ICD demonstrated in the αT-catenin-null heart a preferential reduction of PKP2 at the area composita compared with the desmosome. Furthermore, gap junction protein Cx43 was reduced at the ICD, including its colocalization with N-cadherin. Gap junction remodeling in αT-catenin-knockout hearts was associated with an increased incidence of ventricular arrhythmias after acute ischemia. This novel animal model demonstrates for the first time how perturbation in αT-catenin can affect both PKP2 and Cx43 and thereby highlights the importance of understanding the crosstalk between the junctional proteins of the ICD and its implications for arrhythmogenic cardiomyopathy.


Assuntos
Arritmias Cardíacas/fisiopatologia , Cardiomiopatia Dilatada/patologia , Junções Comunicantes/metabolismo , Ventrículos do Coração/fisiopatologia , Isquemia Miocárdica/complicações , Miócitos Cardíacos/metabolismo , alfa Catenina/deficiência , Junções Aderentes/metabolismo , Animais , Arritmias Cardíacas/etiologia , Arritmias Cardíacas/patologia , Caderinas/metabolismo , Cardiomiopatia Dilatada/etiologia , Cardiomiopatia Dilatada/fisiopatologia , Conexina 43/deficiência , Conexina 43/metabolismo , Desmossomos/metabolismo , Modelos Animais de Doenças , Eletrocardiografia , Junções Comunicantes/patologia , Ventrículos do Coração/patologia , Camundongos , Camundongos Knockout , Mutação , Traumatismo por Reperfusão Miocárdica , Miocárdio/metabolismo , Miocárdio/patologia , Miócitos Cardíacos/patologia , Placofilinas/deficiência , Placofilinas/metabolismo , alfa Catenina/genética , alfa Catenina/metabolismo
15.
Dev Biol ; 364(2): 178-91, 2012 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-22342243

RESUMO

Cell adhesion controls various embryonic morphogenetic processes, including the development of the enteric nervous system (ENS). Ablation of ß1-integrin (ß1-/-) expression in enteric neural crest cells (ENCC) in mice leads to major alterations in the ENS structure caused by reduced migration and increased aggregation properties of ENCC during gut colonization, which gives rise to a Hirschsprung's disease-like phenotype. In the present study, we examined the role of N-cadherin in ENS development and the interplay with ß1 integrins during this process. The Ht-PA-Cre mouse model was used to target gene disruption of N-cadherin and ß1 integrin in migratory NCC and to produce single- and double-conditional mutants for these two types of adhesion receptors. Double mutation of N-cadherin and ß1 integrin led to embryonic lethality with severe defects in ENS development. N-cadherin-null (Ncad-/-) ENCC exhibited a delayed colonization in the developing gut at E12.5, although this was to a lesser extent than in ß1-/- mutants. This delay of Ncad-/- ENCC migration was recovered at later stages of development. The double Ncad-/-; ß1-/- mutant ENCC failed to colonize the distal part of the gut and there was more severe aganglionosis in the proximal hindgut than in the single mutants for N-cadherin or ß1-integrin. This was due to an altered speed of locomotion and directionality in the gut wall. The abnormal aggregation defect of ENCC and the disorganized ganglia network in the ß1-/- mutant was not observed in the double mutant. This indicates that N-cadherin enhances the effect of the ß1-integrin mutation and demonstrates cooperation between these two adhesion receptors during ENS ontogenesis. In conclusion, our data reveal that N-cadherin is not essential for ENS development but it does modulate the modes of ENCC migration and acts in concert with ß1-integrin to control the proper development of the ENS.


Assuntos
Caderinas/metabolismo , Sistema Nervoso Entérico/crescimento & desenvolvimento , Integrina beta1/metabolismo , Animais , Caderinas/genética , Caderinas/fisiologia , Adesão Celular/fisiologia , Diferenciação Celular/fisiologia , Movimento Celular/fisiologia , Sistema Nervoso Entérico/metabolismo , Sistema Nervoso Entérico/fisiologia , Feminino , Integrina beta1/genética , Integrina beta1/fisiologia , Masculino , Camundongos , Crista Neural/embriologia , Crista Neural/fisiologia , Transdução de Sinais/fisiologia
16.
Mol Cell Biol ; 32(6): 1056-67, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22252313

RESUMO

Arrhythmic right ventricular cardiomyopathy (ARVC) is a hereditary heart muscle disease that causes sudden cardiac death (SCD) in young people. Almost half of ARVC patients have a mutation in genes encoding cell adhesion proteins of the desmosome, including plakoglobin (JUP). We previously reported that cardiac tissue-specific plakoglobin (PG) knockout (PG CKO) mice have no apparent conduction abnormality and survive longer than expected. Importantly, the PG homolog, ß-catenin (CTNNB1), showed increased association with the gap junction protein connexin43 (Cx43) in PG CKO hearts. To determine whether ß-catenin is required to maintain cardiac conduction in the absence of PG, we generated mice lacking both PG and ß-catenin specifically in the heart (i.e., double knockout [DKO]). The DKO mice exhibited cardiomyopathy, fibrous tissue replacement, and conduction abnormalities resulting in SCD. Loss of the cadherin linker proteins resulted in dissolution of the intercalated disc (ICD) structure. Moreover, Cx43-containing gap junction plaques were reduced at the ICD, consistent with the arrhythmogenicity of the DKO hearts. Finally, ambulatory electrocardiogram monitoring captured the abrupt onset of spontaneous lethal ventricular arrhythmia in the DKO mice. In conclusion, these studies demonstrate that the N-cadherin-binding partners, PG and ß-catenin, are indispensable for maintaining mechanoelectrical coupling in the heart.


Assuntos
Arritmias Cardíacas/fisiopatologia , Caderinas/metabolismo , Junções Comunicantes/patologia , Coração/fisiopatologia , beta Catenina/genética , gama Catenina/genética , Animais , Arritmias Cardíacas/genética , Arritmias Cardíacas/metabolismo , Arritmias Cardíacas/patologia , Conexina 43/metabolismo , Junções Comunicantes/genética , Junções Comunicantes/metabolismo , Deleção de Genes , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Miocárdio/metabolismo , Miocárdio/patologia , Ligação Proteica , beta Catenina/metabolismo , gama Catenina/metabolismo
17.
Exp Biol Med (Maywood) ; 236(7): 816-22, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21680756

RESUMO

Cell adhesion, mediated by N-cadherin, is critical for embryogenesis since N-cadherin-null embryos die during mid-gestation with multiple developmental defects. To investigate the role of N-cadherin in heart muscle development, N-cadherin was specifically deleted from myocardial cells in mice. The structural integrity of the myocardial cell wall was compromised in the N-cadherin mutant embryos, leading to a malformed heart and a delay in embryonic development. In contrast, cardiac-specific deletion of αE-catenin, found in adherens junctions, or ß-catenin, did not cause any morphological defects in the embryonic heart, presumably due to compensation by αT-catenin that is normally found in intercalated disks and γ-catenin (plakoglobin), respectively. Embryos lacking ß-catenin in the heart also exhibited a cardiac defect, but only later in development resulting in partial lethality. These genetic studies underscore the importance of the N-cadherin/catenin complex in cardiogenesis.


Assuntos
Caderinas/metabolismo , Coração/embriologia , Organogênese/efeitos dos fármacos , beta Catenina/metabolismo , Animais , Caderinas/deficiência , Deleção de Genes , Camundongos , beta Catenina/deficiência
18.
J Biol Chem ; 286(23): 20478-89, 2011 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-21507952

RESUMO

The intercalated disc serves as an organizing center for various cell surface components at the termini of the cardiomyocyte, thus ensuring proper mechanoelectrical coupling throughout the myocardium. The cell adhesion molecule, N-cadherin, is an essential component of the intercalated disc. Cardiac-specific deletion of N-cadherin leads to abnormal electrical conduction and sudden arrhythmic death in mice. The mechanisms linking the loss of N-cadherin in the heart and spontaneous malignant ventricular arrhythmias are poorly understood. To investigate whether ion channel remodeling contributes to arrhythmogenesis in N-cadherin conditional knock-out (N-cad CKO) mice, cardiac myocyte excitability and voltage-gated potassium channel (Kv), as well as inwardly rectifying K(+) channel remodeling, were investigated in N-cad CKO cardiomyocytes by whole cell patch clamp recordings. Action potential duration was prolonged in N-cad CKO ventricle myocytes compared with wild type. Relative to wild type, I(K,slow) density was significantly reduced consistent with decreased expression of Kv1.5 and Kv accessory protein, Kcne2, in the N-cad CKO myocytes. The decreased Kv1.5/Kcne2 expression correlated with disruption of the actin cytoskeleton and reduced cortactin at the sarcolemma. Biochemical experiments revealed that cortactin co-immunoprecipitates with Kv1.5. Finally, cortactin was required for N-cadherin-mediated enhancement of Kv1.5 channel activity in a heterologous expression system. Our results demonstrate a novel mechanistic link among the cell adhesion molecule, N-cadherin, the actin-binding scaffold protein, cortactin, and Kv channel remodeling in the heart. These data suggest that in addition to gap junction remodeling, aberrant Kv1.5 channel function contributes to the arrhythmogenic phenotype in N-cad CKO mice.


Assuntos
Potenciais de Ação/fisiologia , Caderinas/metabolismo , Cortactina/metabolismo , Junções Comunicantes/metabolismo , Canal de Potássio Kv1.5/metabolismo , Miócitos Cardíacos/metabolismo , Animais , Arritmias Cardíacas/genética , Arritmias Cardíacas/metabolismo , Caderinas/genética , Cortactina/genética , Junções Comunicantes/genética , Canal de Potássio Kv1.5/genética , Camundongos , Camundongos Knockout , Especificidade de Órgãos/genética , Canais de Potássio de Abertura Dependente da Tensão da Membrana/genética , Canais de Potássio de Abertura Dependente da Tensão da Membrana/metabolismo
19.
Mol Cell Biol ; 31(6): 1134-44, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21245375

RESUMO

Mutations in the plakoglobin (JUP) gene have been identified in arrhythmogenic right ventricular cardiomyopathy (ARVC) patients. However, the mechanisms underlying plakoglobin dysfunction involved in the pathogenesis of ARVC remain poorly understood. Plakoglobin is a component of both desmosomes and adherens junctions located at the intercalated disc (ICD) of cardiomyocytes, where it functions to link cadherins to the cytoskeleton. In addition, plakoglobin functions as a signaling protein via its ability to modulate the Wnt/ß-catenin signaling pathway. To investigate the role of plakoglobin in ARVC, we generated an inducible cardiorestricted knockout (CKO) of the plakoglobin gene in mice. Plakoglobin CKO mice exhibited progressive loss of cardiac myocytes, extensive inflammatory infiltration, fibrous tissue replacement, and cardiac dysfunction similar to those of ARVC patients. Desmosomal proteins from the ICD were decreased, consistent with altered desmosome ultrastructure in plakoglobin CKO hearts. Despite gap junction remodeling, plakoglobin CKO hearts were refractory to induced arrhythmias. Ablation of plakoglobin caused increase ß-catenin stabilization associated with activated AKT and inhibition of glycogen synthase kinase 3ß. Finally, ß-catenin/TCF transcriptional activity may contribute to the cardiac hypertrophy response in plakoglobin CKO mice. This novel model of ARVC demonstrates for the first time how plakoglobin affects ß-catenin activity in the heart and its implications for disease pathogenesis.


Assuntos
Displasia Arritmogênica Ventricular Direita/genética , Displasia Arritmogênica Ventricular Direita/patologia , Técnicas de Inativação de Genes , beta Catenina/metabolismo , gama Catenina/genética , Junções Aderentes/genética , Junções Aderentes/metabolismo , Junções Aderentes/patologia , Animais , Displasia Arritmogênica Ventricular Direita/metabolismo , Citocinas/imunologia , Desmossomos/genética , Desmossomos/metabolismo , Desmossomos/patologia , Regulação da Expressão Gênica , Humanos , Camundongos , Camundongos Knockout , Miocárdio/patologia , beta Catenina/genética , gama Catenina/metabolismo
20.
J Neurosci ; 30(30): 9984-9, 2010 Jul 28.
Artigo em Inglês | MEDLINE | ID: mdl-20668183

RESUMO

Persistent changes in spine shape are coupled to long-lasting synaptic plasticity in hippocampus. The molecules that coordinate such persistent structural and functional plasticity are unknown. Here, we generated mice in which the cell adhesion molecule N-cadherin was conditionally ablated from postnatal, excitatory synapses in hippocampus. We applied to adult mice of either sex a combination of whole-cell recording, two-photon microscopy, and spine morphometric analysis to show that postnatal ablation of N-cadherin has profound effects on the stability of coordinated spine enlargement and long-term potentiation (LTP) at mature CA1 synapses, with no effects on baseline spine density or morphology, baseline properties of synaptic neurotransmission, or long-term depression. Thus, N-cadherin couples persistent spine structural modifications with long-lasting synaptic functional modifications associated selectively with LTP, revealing unexpectedly distinct roles at mature synapses in comparison with earlier, broader functions in synapse and spine development.


Assuntos
Região CA1 Hipocampal/citologia , Caderinas/metabolismo , Espinhas Dendríticas/fisiologia , Potenciação de Longa Duração/fisiologia , Células Piramidais/ultraestrutura , Sinapses/metabolismo , Potenciais de Ação/fisiologia , Animais , Biofísica/métodos , Caderinas/deficiência , Estimulação Elétrica/métodos , Potenciais Pós-Sinápticos Excitadores/genética , Técnicas In Vitro , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia Imunoeletrônica/métodos , Técnicas de Patch-Clamp/métodos , Estatísticas não Paramétricas , Sinapses/ultraestrutura
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA