Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 110
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Biochem Biophys Res Commun ; 586: 14-19, 2022 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-34823217

RESUMO

Plakophilin3 (PKP3) loss leads to tumor progression and metastasis of colon cancer cells. The goal of this report was to determine if PKP3 loss led to increased disease progression in mice. We generated a colonocyte-specific knockout of PKP3 in APCmin mice, which led to increased adenoma formation, the formation of rectal prolapse, and a significant decrease in survival. The observed increase in rectal prolapse formation and decrease in survival correlated with an increase in the expression of Lipocalin2 (LCN2). Increased disease progression was observed even upon treatment with 5-fluorouracil (5FU). These results suggest that an increase in LCN2 expression might lead to therapy resistance and that LCN2 might serve as a potential therapeutic target in colorectal cancer.


Assuntos
Adenoma/genética , Neoplasias Colorretais/genética , Resistencia a Medicamentos Antineoplásicos/genética , Lipocalina-2/genética , Placofilinas/genética , Prolapso Retal/genética , Adenoma/tratamento farmacológico , Adenoma/mortalidade , Adenoma/patologia , Animais , Antimetabólitos Antineoplásicos/farmacologia , Colo/efeitos dos fármacos , Colo/metabolismo , Colo/patologia , Neoplasias Colorretais/tratamento farmacológico , Neoplasias Colorretais/mortalidade , Neoplasias Colorretais/patologia , Progressão da Doença , Feminino , Fluoruracila/farmacologia , Regulação Neoplásica da Expressão Gênica , Queratina-8/genética , Queratina-8/metabolismo , Lipocalina-2/metabolismo , Masculino , Metaloproteinase 7 da Matriz/genética , Metaloproteinase 7 da Matriz/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Placofilinas/deficiência , Prolapso Retal/tratamento farmacológico , Prolapso Retal/mortalidade , Prolapso Retal/patologia , Transdução de Sinais , Análise de Sobrevida
2.
Development ; 146(21)2019 11 08.
Artigo em Inglês | MEDLINE | ID: mdl-31601548

RESUMO

A switch from E- to N-cadherin regulates the transition from pluripotency to neural identity, but the mechanism by which cadherins regulate differentiation was previously unknown. Here, we show that the acquisition of N-cadherin stabilises neural identity by dampening anti-neural signals. We use quantitative image analysis to show that N-cadherin promotes neural differentiation independently of its effects on cell cohesiveness. We reveal that cadherin switching diminishes the level of nuclear ß-catenin, and that N-cadherin also dampens FGF activity and consequently stabilises neural fate. Finally, we compare the timing of cadherin switching and differentiation in vivo and in vitro, and find that this process becomes dysregulated during in vitro differentiation. We propose that N-cadherin helps to propagate a stable neural identity throughout the emerging neuroepithelium, and that dysregulation of this process contributes to asynchronous differentiation in culture.


Assuntos
Caderinas/fisiologia , Células-Tronco Embrionárias/citologia , Neurônios/citologia , beta Catenina/fisiologia , Animais , Diferenciação Celular , Linhagem da Célula , Núcleo Celular/fisiologia , Células Cultivadas , Fatores de Crescimento de Fibroblastos/fisiologia , Camadas Germinativas/fisiologia , Camundongos , Camundongos Transgênicos , Células-Tronco Pluripotentes/citologia
3.
BMC Cancer ; 22(1): 451, 2022 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-35468745

RESUMO

BACKGROUND: Nonclustered mouse protocadherin genes (Pcdh) encode proteins with a typical single ectodomain and a cytoplasmic domain with conserved motifs completely different from those of classic cadherins. Alternative splice isoforms differ in the size of these cytoplasmic domains. In view of the compelling evidence for gene silencing of protocadherins in human tumors, we started investigations on Pcdh functions in mouse cancer models. METHODS: For Pcdh10, we generated two mouse lines: one with floxed exon 1, leading to complete Pcdh10 ablation upon Cre action, and one with floxed exons 2 and 3, leading to ablation of only the long isoforms of Pcdh10. In a mouse medulloblastoma model, we used GFAP-Cre action to locally ablate Pcdh10 in combination with Trp53 and Rb1 ablation. From auricular tumors, that also arose, we obtained tumor-derived cell lines, which were analyzed for malignancy in vitro and in vivo. By lentiviral transduction, we re-expressed Pcdh10 cDNAs. RNA-Seq analyses were performed on these cell families. RESULTS: Surprisingly, not only medulloblastomas were generated in our model but also tumors of tagged auricles (pinnae). For both tumor types, ablation of either all or only long isoforms of Pcdh10 aggravated the disease. We argued that the perichondrial stem cell compartment is at the origin of the pinnal tumors. Immunohistochemical analysis of these tumors revealed different subtypes. We obtained several pinnal-tumor derived (PTD) cell lines and analyzed these for anchorage-independent growth, invasion into collagen matrices, tumorigenicity in athymic mice. Re-expression of either the short or a long isoform of Pcdh10 in two PTD lines counteracted malignancy in all assays. RNA-Seq analyses of these two PTD lines and their respective Pcdh10-rescued cell lines allowed to identify many interesting differentially expressed genes, which were largely different in the two cell families. CONCLUSIONS: A new mouse model was generated allowing for the first time to examine the remarkable tumor suppression activity of protocadherin-10 in vivo. Despite lacking several conserved motifs, the short isoform of Pcdh10 was fully active as tumor suppressor. Our model contributes to scrutinizing the complex molecular mechanisms of tumor initiation and progression upon PCDH10 silencing in many human cancers.


Assuntos
Neoplasias Cerebelares , Meduloblastoma , Animais , Apoptose/genética , Movimento Celular/genética , Proliferação de Células/genética , Humanos , Meduloblastoma/genética , Camundongos , Isoformas de Proteínas/genética , Protocaderinas
4.
BMC Dev Biol ; 20(1): 17, 2020 08 03.
Artigo em Inglês | MEDLINE | ID: mdl-32741376

RESUMO

BACKGROUND: p120 catenin (p120ctn) is an important component in the cadherin-catenin cell adhesion complex because it stabilizes cadherin-mediated intercellular junctions. Outside these junctions, p120ctn is actively involved in the regulation of small GTPases of the Rho family, in actomyosin dynamics and in transcription regulation. We and others reported that loss of p120ctn in mouse embryos results in an embryonic lethal phenotype, but the exact developmental role of p120ctn during brain formation has not been reported. RESULTS: We combined floxed p120ctn mice with Del-Cre or Wnt1-Cre mice to deplete p120ctn from either all cells or specific brain and neural crest cells. Complete loss of p120ctn in mid-gestation embryos resulted in an aberrant morphology, including growth retardation, failure to switch from lordotic to fetal posture, and defective neural tube formation and neurogenesis. By expressing a wild-type p120ctn from the ROSA26 locus in p120ctn-null mouse embryonic stem cells, we could partially rescue neurogenesis. To further investigate the developmental role of p120ctn in neural tube formation, we generated conditional p120ctnfl/fl;Wnt1Cre knockout mice. p120ctn deletion in Wnt1-expressing cells resulted in neural tube closure defects (NTDs) and craniofacial abnormalities. These defects could not be correlated with misregulation of brain marker genes or cell proliferation. In contrast, we found that p120ctn is required for proper expression of the cell adhesion components N-cadherin, E-cadherin and ß-catenin, and of actin-binding proteins cortactin and Shroom3 at the apical side of neural folds. This region is of critical importance for closure of neural folds. Surprisingly, the lateral side of mutant neural folds showed loss of p120ctn, but not of N-cadherin, ß-catenin or cortactin. CONCLUSIONS: These results indicate that p120ctn is required for neurogenesis and neurulation. Elimination of p120ctn in cells expressing Wnt1 affects neural tube closure by hampering correct formation of specific adhesion and actomyosin complexes at the apical side of neural folds. Collectively, our results demonstrate the crucial role of p120ctn during brain morphogenesis.


Assuntos
Cateninas/metabolismo , Proteína Wnt1/metabolismo , Animais , Caderinas/genética , Caderinas/metabolismo , Cateninas/genética , Adesão Celular/genética , Adesão Celular/fisiologia , Camundongos , Camundongos Knockout , RNA não Traduzido/genética , RNA não Traduzido/metabolismo , Proteína Wnt1/genética , beta Catenina/genética , beta Catenina/metabolismo
5.
BMC Cancer ; 19(1): 598, 2019 Jun 17.
Artigo em Inglês | MEDLINE | ID: mdl-31208373

RESUMO

BACKGROUND: NANOS3 is a gene conserved throughout evolution. Despite the quite low conservation of Nanos sequences between different organisms and even between Nanos paralogs, their role in germ cell development is remarkably universal. Human Nanos3 expression is normally restricted to the gonads and the brain. However, ectopic activation of this gene has been detected in various human cancers. Until now, Nanos3 and other Nanos proteins have been studied almost exclusively in germ cell development. METHODS: Transgenic mice were generated by targeted insertion of a human Nanos3 cDNA into the ROSA26 locus. The transgene could be spatiotemporally induced by Cre recombinase activity removing an upstream floxed STOP cassette. A lung tumor model with ectopic Nanos3 expression was based on the lung-specific activation of the reverse tetracycline transactivator gene, in combination with a tetO-CMV promoter controlling Cre expression. When doxycycline was provided to the mice, Cre was activated leading to deletion of TP53 alleles and activation of both oncogenic KRasG12D and Nanos3. Appropriate controls were foreseen. Tumors and tumor-derived cell cultures were analyzed in various ways. RESULTS: We describe the successful generation of Nanos3LSL/- and Nanos3LSL/LSL mice in which an exogenous human NANOS3 gene can be activated in vivo upon Cre expression. These mice, in combination with different conditional and doxycycline-inducible Cre lines, allow the study of the role of ectopic Nanos3 expression in several cancer types. The Nanos3LSL mice were crossed with a non-small cell lung cancer (NSCLC) mouse model based on conditional expression of oncogenic KRas and homozygous loss of p53. This experiment demonstrated that ectopic expression of Nanos3 in the lungs has a significant negative effect on survival. Enhanced bronchiolar dysplasia was observed when Nanos3-expressing NSCLC mice were compared with control NSCLC mice. An allograft experiment, performed with cell cultures derived from primary lung tumors of control and Nanos3-expressing NSCLC mice, revealed lymph node metastasis in mice injected with Nanos3-expressing NSCLC cells. CONCLUSIONS: A new mouse model was generated allowing examination of Nanos3-associated pathways and investigation of the influence of ectopic Nanos3 expression in various cancer types. This model might identify Nanos3 as an interesting target in cancer therapeutics.


Assuntos
Expressão Ectópica do Gene , Camundongos , Neoplasias Experimentais/genética , Proteínas de Ligação a RNA/genética , Aloenxertos , Animais , Carcinoma Pulmonar de Células não Pequenas/genética , Doxiciclina/farmacologia , Feminino , Humanos , Integrases , Estimativa de Kaplan-Meier , Neoplasias Pulmonares/genética , Masculino , Camundongos Endogâmicos C57BL , Camundongos Nus , Camundongos Transgênicos , Transdução de Sinais/efeitos dos fármacos , Transgenes , Células Tumorais Cultivadas , Proteína Supressora de Tumor p53/genética
6.
Cell Mol Life Sci ; 75(11): 1929-1946, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29397397

RESUMO

The hallmark of Nanos proteins is their typical (CCHC)2 zinc finger motif (zf-nanos). Animals have one to four nanos genes. For example, the fruit fly and demosponge have only one nanos gene, zebrafish and humans have three, and Fugu rubripes has four. Nanos genes are mainly known for their evolutionarily preserved role in germ cell survival and pluripotency. Nanos proteins have been reported to bind the C-terminal RNA-binding domain of Pumilio to form a post-transcriptional repressor complex. Several observations point to a link between the miRNA-mediated repression complex and the Nanos/Pumilio complex. Repression of the E2F3 oncogene product is, indeed, mediated by cooperation between the Nanos/Pumilio complex and miRNAs. Another important interaction partner of Nanos is the CCR4-NOT deadenylase complex. Besides the tissue-specific contribution of Nanos proteins to normal development, their ectopic expression has been observed in several cancer cell lines and various human cancers. An inverse correlation between the expression levels of human Nanos1 and Nanos3 and E-cadherin was observed in several cancer cell lines. Loss of E-cadherin, an important cell-cell adhesion protein, contributes to tumor invasion and metastasis. Overexpression of Nanos3 induces epithelial-mesenchymal transition in lung cancer cell lines partly by repressing E-cadherin. Other than some most interesting data from Nanos knockout mice, little is known about mammalian Nanos proteins, and further research is needed. In this review, we summarize the main roles of Nanos proteins and discuss the emerging concept of Nanos proteins as oncofetal antigens.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Genômica , Mapas de Interação de Proteínas , Proteínas de Ligação a RNA/genética , Sequência de Aminoácidos , Animais , Genômica/métodos , Humanos , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patologia , Filogenia , Proteínas de Ligação a RNA/análise , Proteínas de Ligação a RNA/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Dedos de Zinco
7.
PLoS Genet ; 12(8): e1006243, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27556156

RESUMO

E-cadherin-mediated cell-cell adhesion is critical for naive pluripotency of cultured mouse embryonic stem cells (mESCs). E-cadherin-depleted mESC fail to downregulate their pluripotency program and are unable to initiate lineage commitment. To further explore the roles of cell adhesion molecules during mESC differentiation, we focused on p120 catenin (p120ctn). Although one key function of p120ctn is to stabilize and regulate cadherin-mediated cell-cell adhesion, it has many additional functions, including regulation of transcription and Rho GTPase activity. Here, we investigated the role of mouse p120ctn in early embryogenesis, mESC pluripotency and early fate determination. In contrast to the E-cadherin-null phenotype, p120ctn-null mESCs remained pluripotent, but their in vitro differentiation was incomplete. In particular, they failed to form cystic embryoid bodies and showed defects in primitive endoderm formation. To pinpoint the underlying mechanism, we undertook a structure-function approach. Rescue of p120ctn-null mESCs with different p120ctn wild-type and mutant expression constructs revealed that the long N-terminal domain of p120ctn and its regulatory domain for RhoA were dispensable, whereas its armadillo domain and interaction with E-cadherin were crucial for primitive endoderm formation. We conclude that p120ctn is not only an adaptor and regulator of E-cadherin, but is also indispensable for proper lineage commitment.


Assuntos
Caderinas/genética , Cateninas/genética , Diferenciação Celular/genética , Endoderma/crescimento & desenvolvimento , Células-Tronco Embrionárias Murinas , Animais , Blastocisto/metabolismo , Caderinas/biossíntese , Cateninas/biossíntese , Adesão Celular/genética , Linhagem da Célula/genética , Polaridade Celular/genética , Corpos Embrioides/metabolismo , Desenvolvimento Embrionário/genética , Endoderma/metabolismo , Humanos , Camundongos , Imagem Óptica , Células-Tronco Pluripotentes/metabolismo , Proteína rhoA de Ligação ao GTP/biossíntese , Proteína rhoA de Ligação ao GTP/genética , delta Catenina
8.
Histochem Cell Biol ; 149(5): 479-490, 2018 May.
Artigo em Inglês | MEDLINE | ID: mdl-29508067

RESUMO

The intercalated disc (ID) contains different kinds of intercellular junctions: gap junctions (GJs), desmosomes and areae compositae, essential for adhesion and communication between adjacent cardiomyocytes. The junctions can be identified based on their morphology when imaged using transmission electron microscopy (TEM), however, only with very limited information in the z-dimension. The application of volume EM techniques can give insight into the three-dimensional (3-D) organization of complex biological structures. In this study, we generated 3-D datasets using serial block-face scanning electron microscopy (SBF-SEM) and focused ion beam SEM (FIB-SEM), the latter resulting in datasets with 5 nm isotropic voxels. We visualized cardiomyocytes in murine ventricular heart tissue and, for the first time, we could three-dimensionally reconstruct the ID including desmosomes and GJs with 5 nm precision in a large volume. Results show in three dimensions a highly folded structure of the ID, with the presence of GJs and desmosomes in both plicae and interplicae regions. We observed close contact of GJs with mitochondria and a variable spatial distribution of the junctions. Based on measurements of the shape of the intercellular junctions in 3-D, it is seen that GJs and desmosomes vary in size, depending on the region within the ID. This demonstrates that volume EM is essential to visualize morphological changes and its potential to quantitatively determine structural changes between normal and pathological conditions, e.g., cardiomyopathies.


Assuntos
Imageamento Tridimensional , Junções Intercelulares/ultraestrutura , Miócitos Cardíacos/ultraestrutura , Animais , Camundongos , Microscopia Eletrônica de Varredura , Miócitos Cardíacos/citologia , Fenótipo
9.
Exp Cell Res ; 358(1): 3-9, 2017 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-28268172

RESUMO

Cadherin genes encode a superfamily of conserved transmembrane proteins that share an adhesive ectodomain composed of tandem cadherin repeats. More than 100 human cadherin superfamily members have been identified, which can be classified into three families: major cadherins, protocadherins and cadherin-related proteins. These superfamily members are involved in diverse fundamental cellular processes including cell-cell adhesion, morphogenesis, cell recognition and signaling. Epithelial cadherin (E-cadherin) is the founding cadherin family member. Its cytoplasmic tail interacts with the armadillo catenins, p120 and ß-catenin. Further, α-catenin links the cadherin/armadillo catenin complex to the actin filament network. Even genomes of ancestral metazoan species such as cnidarians and placozoans encode a limited number of distinct cadherins and catenins, emphasizing the conservation and functional importance of these gene families. Moreover, a large expansion of the cadherin and catenin families coincides with the emergence of vertebrates and reflects a major functional diversification in higher metazoans. Here, we revisit and review the functions, phylogenetic classifications and co-evolution of the cadherin and catenin protein families.


Assuntos
Caderinas/metabolismo , Cateninas/metabolismo , Adesão Celular/fisiologia , Membrana Celular/metabolismo , Proteínas do Citoesqueleto/metabolismo , Animais , Humanos , Morfogênese/fisiologia
10.
Cell Mol Life Sci ; 74(3): 525-541, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27497926

RESUMO

The superfamily of armadillo repeat proteins is a fascinating archetype of modular-binding proteins involved in various fundamental cellular processes, including cell-cell adhesion, cytoskeletal organization, nuclear import, and molecular signaling. Despite their diverse functions, they all share tandem armadillo (ARM) repeats, which stack together to form a conserved three-dimensional structure. This superhelical armadillo structure enables them to interact with distinct partners by wrapping around them. Despite the important functional roles of this superfamily, a comprehensive analysis of the composition, classification, and phylogeny of this protein superfamily has not been reported. Furthermore, relatively little is known about a subset of ARM proteins, and some of the current annotations of armadillo repeats are incomplete or incorrect, often due to high similarity with HEAT repeats. We identified the entire armadillo repeat superfamily repertoire in the human genome, annotated each armadillo repeat, and performed an extensive evolutionary analysis of the armadillo repeat proteins in both metazoan and premetazoan species. Phylogenetic analyses of the superfamily classified them into several discrete branches with members showing significant sequence homology, and often also related functions. Interestingly, the phylogenetic structure of the superfamily revealed that about 30 % of the members predate metazoans and represent an ancient subset, which is gradually evolving to acquire complex and highly diverse functions.


Assuntos
Proteínas do Domínio Armadillo/genética , Filogenia , Sequência de Aminoácidos , Animais , Proteínas do Domínio Armadillo/química , Proteínas do Domínio Armadillo/classificação , Proteínas do Domínio Armadillo/metabolismo , Evolução Biológica , Evolução Molecular , Humanos , Modelos Moleculares , Conformação Proteica , Alinhamento de Sequência
11.
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
12.
J Cell Sci ; 127(Pt 12): 2603-13, 2014 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-24931943

RESUMO

Pluripotent embryonic stem cells (ESCs) can self-renew or differentiate into any cell type within an organism. Here, we focus on the roles of cadherins and catenins - their cytoplasmic scaffold proteins - in the fate, maintenance and differentiation of mammalian ESCs. E-cadherin is a master stem cell regulator that is required for both mouse ESC (mESC) maintenance and differentiation. E-cadherin interacts with key components of the naive stemness pathway and ablating it prevents stem cells from forming well-differentiated teratomas or contributing to chimeric animals. In addition, depleting E-cadherin converts naive mouse ESCs into primed epiblast-like stem cells (EpiSCs). In line with this, a mesenchymal-to-epithelial transition (MET) occurs during reprogramming of somatic cells towards induced pluripotent stem cells (iPSCs), leading to downregulation of N-cadherin and acquisition of high E-cadherin levels. ß-catenin exerts a dual function; it acts in cadherin-based adhesion and in WNT signaling and, although WNT signaling is important for stemness, the adhesive function of ß-catenin might be crucial for maintaining the naive state of stem cells. In addition, evidence is rising that other junctional proteins are also important in ESC biology. Thus, precisely regulated levels and activities of several junctional proteins, in particular E-cadherin, safeguard naive pluripotency and are a prerequisite for complete somatic cell reprogramming.


Assuntos
Moléculas de Adesão Celular/metabolismo , Células-Tronco Embrionárias/fisiologia , Junções Intercelulares/fisiologia , Animais , Cateninas/fisiologia , Adesão Celular , Diferenciação Celular , Proliferação de Células , Humanos
13.
Eur J Immunol ; 45(10): 2898-910, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26173741

RESUMO

Plakophilin-3 (PKP3) is a member of the armadillo protein family, which is important in cell-cell contacts and signaling during development and tumorigenesis. In conventional facilities, PKP3-deficient mice (PKP3(-/-)) develop spontaneous dermatitis, indicating a possible involvement of PKP3 in inflammatory responses. Here, we show that PKP3 deficiency sensitizes mice to irritant contact dermatitis induced by phorbol myristate acetate (PMA). This sensitization occurred in mice with PKP3 deficiency in the hematopoietic system (PKP3(-/-hem)), but not if the deficiency was specific to skin keratinocytes (PKP3(-/-ker)). In a model of dextran sulfate sodium induced colitis, ubiquitous PKP3 deletion, but not intestinal epithelial PKP3 deficiency (PKP3(-/-IEC)), impaired survival from disease. Interestingly, PKP3(-/-hem) mice also displayed increased sensitivity to dextran sulfate sodium induced colitis. Finally, PKP3(-/-) mice were more sensitive to the lethality of lipopolysaccharide (LPS) injection than wild-type (WT) mice, and this phenotype was associated with increased intestinal permeability. PKP3(-/-IEC) mice did not reproduce the enhanced endotoxin reactivity of PKP3(-/-) mice, in contrast to PKP3(-/-hem) mice. Finally, in vitro stimulation of WT neutrophils with LPS or PMA increased Pkp3 expression. In conclusion, our data highlight a novel role for hematopoietic PKP3 in the regulation of both locally and systemically induced immune responses. Nonetheless, further research is needed to unravel the underlying mechanism.


Assuntos
Colite/imunologia , Dermatite de Contato/imunologia , Regulação da Expressão Gênica/imunologia , Neutrófilos/imunologia , Placofilinas/imunologia , Animais , Colite/induzido quimicamente , Dermatite de Contato/genética , Dermatite de Contato/patologia , Sulfato de Dextrana/toxicidade , Regulação da Expressão Gênica/efeitos dos fármacos , Inflamação/induzido quimicamente , Inflamação/genética , Inflamação/imunologia , Inflamação/patologia , Lipopolissacarídeos/toxicidade , Camundongos , Camundongos Knockout , Neutrófilos/patologia , Placofilinas/genética , Acetato de Tetradecanoilforbol/toxicidade
14.
J Pathol ; 237(1): 25-37, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25904364

RESUMO

We have explored the role of the human NANOS3 gene in lung tumour progression. We show that NANOS3 is over-expressed by invasive lung cancer cells and is a prognostic marker for non-small cell lung carcinomas (NSCLCs). NANOS3 gene expression is restricted in testis and brain and is regulated by epigenetic events. It is up-regulated in cultured cells undergoing epithelial - mesenchymal transition (EMT). NANOS3 over-expression in human NSCLC cell lines enhances their invasiveness by up-regulating EMT, whereas its silencing induces mesenchymal - epithelial transition. NANOS3 represses E-cadherin at the transcriptional level and up-regulates vimentin post-transcriptionally. Also, we show that NANOS3 binds mRNAs encoding vimentin and regulates the length of their poly(A) tail. Finally, NANOS3 can also protect vimentin mRNA from microRNA-mediated repression. We thus demonstrate a role for NANOS3 in the acquisition of invasiveness by human lung tumour cells and propose a new mechanism of post-transcriptional regulation of EMT.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/metabolismo , Movimento Celular , Transição Epitelial-Mesenquimal , Neoplasias Pulmonares/metabolismo , Proteínas de Ligação a RNA/metabolismo , Vimentina/metabolismo , Antígenos CD , Caderinas/genética , Caderinas/metabolismo , Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/mortalidade , Carcinoma Pulmonar de Células não Pequenas/patologia , Linhagem Celular Tumoral , Epigênese Genética , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Estimativa de Kaplan-Meier , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/mortalidade , Neoplasias Pulmonares/patologia , Masculino , MicroRNAs/metabolismo , Pessoa de Meia-Idade , Invasividade Neoplásica , Prognóstico , RNA Mensageiro/metabolismo , Proteínas de Ligação a RNA/genética , Transdução de Sinais , Fatores de Tempo , Transcrição Gênica , Transfecção , Vimentina/genética
15.
Am J Physiol Lung Cell Mol Physiol ; 308(3): L253-8, 2015 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-25480337

RESUMO

Ten to 25% of adult asthma is occupational induced, a subtype caused by exposure to workplace chemicals. A recent genomewide association study identified single-nucleotide polymorphisms in the cardiac protein αT-catenin (αT-cat) that correlated with the incidence and severity of toluene diisocyanate (TDI) occupational asthma. αT-cat is a critical mediator of cell-cell adhesion and is predominantly expressed in cardiomyocytes, but its connection to asthma remains unknown. Therefore, we sought to determine the primary αT-cat-expressing cell type in the lung and its contribution to lung physiology in a murine model of TDI asthma. We show that αT-cat is expressed in lung within the cardiac sheath of pulmonary veins. Mechanically ventilated αT-cat knockout (KO) mice exhibit a significantly increased pressure-volume curve area compared with wild-type (WT) mice, suggesting that αT-cat loss affects lung hysteresis. Using a murine model of TDI asthma, we find that αT-cat KO mice show increased airway hyperresponsiveness to methacholine compared with WT mice. Bronchoalveolar lavage reveals only a mild macrophage-dominant inflammation that is not significantly different between WT and KO mice. These data suggest that αT-cat may contribute to asthma through a mechanism independent of inflammation and related to heart and pulmonary vein dysfunction.


Assuntos
Poluentes Atmosféricos/toxicidade , Asma Ocupacional/metabolismo , Tolueno 2,4-Di-Isocianato/toxicidade , alfa Catenina/metabolismo , Animais , Asma Ocupacional/induzido quimicamente , Células Cultivadas , Feminino , Humanos , Junções Intercelulares/metabolismo , Pulmão/irrigação sanguínea , Pulmão/metabolismo , Pulmão/patologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Miócitos Cardíacos/metabolismo , Veias Pulmonares/metabolismo , Veias Pulmonares/patologia
16.
Am J Physiol Lung Cell Mol Physiol ; 309(7): L725-35, 2015 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-26209277

RESUMO

Genetic studies have identified Protocadherin-1 (PCDH1) and Mothers against decapentaplegic homolog-3 (SMAD3) as susceptibility genes for asthma. PCDH1 is expressed in bronchial epithelial cells and has been found to interact with SMAD3 in yeast two-hybrid (Y2H) overexpression assays. Here, we test whether PCDH1 and SMAD3 interact at endogenous protein levels in bronchial epithelial cells and evaluate the consequences thereof for transforming growth factor-ß1 (TGF-ß1)-induced gene transcription. We performed Y2H screens and coimmunoprecipitation (co-IP) experiments of PCDH1 and SMAD3 in HEK293T and 16HBE14o(-) (16HBE) cell lines. Activity of a SMAD3-driven luciferase reporter gene in response to TGF-ß1 was measured in BEAS-2B cells transfected with PCDH1 and in 16HBE cells transfected with PCDH1-small-interfering RNA (siRNA). TGF-ß1-induced gene expression was quantified in BEAS-2B clones overexpressing PCDH1 and in human primary bronchial epithelial cells (PBECs) transfected with PCDH1-siRNA. We confirm PCDH1 and SMAD3 interactions by Y2H and by co-IP in HEK293T cells overexpressing both proteins, and at endogenous protein levels in 16HBE cells. TGF-ß-induced activation of a SMAD3-driven reporter was reduced by exogenous PCDH1 in BEAS2B cells, whereas it was increased by siRNA-mediated knockdown of endogenous PCDH1 in 16HBE cells. Overexpression of PCDH1 suppressed expression of TGF-ß target genes in BEAS-2B cells, whereas knockdown of PCDH1 in human PBECs increased TGF-ß-induced gene expression. In conclusion, we demonstrate that PCDH1 binds to SMAD3 and regulates its activation by TGF-ß signaling in bronchial epithelial cells. We propose that PCDH1 and SMAD3 act in a single pathway in asthma susceptibility that affects sensitivity of the airway epithelium to TGF-ß.


Assuntos
Brônquios/metabolismo , Caderinas/metabolismo , Células Epiteliais/metabolismo , Mucosa Respiratória/metabolismo , Proteína Smad3/metabolismo , Transcrição Gênica , Fator de Crescimento Transformador beta1/metabolismo , Asma/genética , Asma/metabolismo , Asma/patologia , Brônquios/patologia , Caderinas/genética , Células Epiteliais/patologia , Células HEK293 , Humanos , Ligação Proteica , Protocaderinas , Mucosa Respiratória/patologia , Proteína Smad3/genética , Fator de Crescimento Transformador beta1/genética , Técnicas do Sistema de Duplo-Híbrido
17.
BMC Cancer ; 15: 391, 2015 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-25958384

RESUMO

BACKGROUND: NBPF1 (Neuroblastoma Breakpoint Family, member 1) was originally identified in a neuroblastoma patient on the basis of its disruption by a chromosomal translocation t(1;17)(p36.2;q11.2). Considering this genetic defect and the frequent genomic alterations of the NBPF1 locus in several cancer types, we hypothesized that NBPF1 is a tumor suppressor. Decreased expression of NBPF1 in neuroblastoma cell lines with loss of 1p36 heterozygosity and the marked decrease of anchorage-independent clonal growth of DLD1 colorectal carcinoma cells with induced NBPF1 expression further suggest that NBPF1 functions as tumor suppressor. However, little is known about the mechanisms involved. METHODS: Expression of NBPF was analyzed in human skin and human cervix by immunohistochemistry. The effects of NBPF1 on the cell cycle were evaluated by flow cytometry. We investigated by real-time quantitative RT-PCR the expression profile of a panel of genes important in cell cycle regulation. Protein levels of CDKN1A-encoded p21(CIP1/WAF1) were determined by western blotting and the importance of p53 was shown by immunofluorescence and by a loss-of-function approach. LC-MS/MS analysis was used to investigate the proteome of DLD1 colon cancer cells with induced NBPF1 expression. Possible biological interactions between the differentially regulated proteins were investigated with the Ingenuity Pathway Analysis tool. RESULTS: We show that NBPF is expressed in the non-proliferative suprabasal layers of squamous stratified epithelia of human skin and cervix. Forced expression of NBPF1 in HEK293T cells resulted in a G1 cell cycle arrest that was accompanied by upregulation of the cyclin-dependent kinase inhibitor p21(CIP1/WAF1) in a p53-dependent manner. Additionally, forced expression of NBPF1 in two p53-mutant neuroblastoma cell lines also resulted in a G1 cell cycle arrest and CDKN1A upregulation. However, CDKN1A upregulation by NBPF1 was not observed in the DLD1 cells, which demonstrates that NBPF1 exerts cell-specific effects. In addition, proteome analysis of NBPF1-overexpressing DLD1 cells identified 32 differentially expressed proteins, of which several are implicated in carcinogenesis. CONCLUSIONS: We demonstrated that NBPF1 exerts different tumor suppressive effects, depending on the cell line analyzed, and provide new clues into the molecular mechanism of the enigmatic NBPF proteins.


Assuntos
Proteínas de Transporte/genética , Pontos de Checagem da Fase G1 do Ciclo Celular/genética , Neuroblastoma/genética , Proteínas Supressoras de Tumor/genética , Proteínas de Transporte/metabolismo , Linhagem Celular Tumoral , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Epitélio/metabolismo , Epitélio/patologia , Expressão Gênica , Genes Reporter , Células HEK293 , Humanos , Família Multigênica , Neuroblastoma/metabolismo , Proteoma , Proteômica , Transdução de Sinais , Transfecção , Proteína Supressora de Tumor p53/metabolismo , Proteínas Supressoras de Tumor/metabolismo
18.
Nucleic Acids Res ; 41(Web Server issue): W531-4, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23620286

RESUMO

The most important mechanism in the regulation of transcription is the binding of a transcription factor (TF) to a DNA sequence called the TF binding site (TFBS). Most binding sites are short and degenerate, which makes predictions based on their primary sequence alone somewhat unreliable. We present a new web tool that implements a flexible and extensible algorithm for predicting TFBS. The algorithm makes use of both direct (the sequence) and several indirect readout features of protein-DNA complexes (biophysical properties such as bendability or the solvent-excluded surface of the DNA). This algorithm significantly outperforms state-of-the-art approaches for in silico identification of TFBS. Users can submit FASTA sequences for analysis in the PhysBinder integrative algorithm and choose from >60 different TF-binding models. The results of this analysis can be used to plan and steer wet-lab experiments. The PhysBinder web tool is freely available at http://bioit.dmbr.ugent.be/physbinder/index.php.


Assuntos
DNA/química , Software , Fatores de Transcrição/química , Algoritmos , Sítios de Ligação , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/metabolismo , Humanos , Internet , Regiões Promotoras Genéticas , Análise de Sequência de DNA , Telomerase/genética , Fatores de Transcrição/metabolismo
19.
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
20.
Nat Rev Cancer ; 5(12): 956-64, 2005 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16294216

RESUMO

Kaiso belongs to the zinc finger and broad-complex, tramtrack and bric-a-brac/poxvirus and zinc finger (BTB/POZ) protein family that has been implicated in tumorigenesis. Kaiso was first discovered in a complex with the armadillo-domain protein p120ctn and later shown to function as a transcriptional repressor. As p120ctn seems to relieve Kaiso-mediated repression, its altered intracellular localization in some cancer cells might result in aberrant Kaiso nuclear activity. Intriguingly, Kaiso's target genes include both methylated and sequence-specific recognition sites. The latter include genes that are modulated by the canonical Wnt (beta-catenin-T-cell factor) signalling pathway. Further interest in Kaiso stems from findings that its cytoplasmic versus nuclear localization is modulated by complex cues from the microenvironment.


Assuntos
Neoplasias/genética , Fatores de Transcrição/genética , Animais , Proteínas do Domínio Armadillo/genética , Cateninas , Moléculas de Adesão Celular , Proteínas do Citoesqueleto/genética , Humanos , Fosfoproteínas , Proteínas Repressoras/genética , Xenopus , Proteínas de Xenopus/genética , beta Catenina/genética , delta Catenina
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA