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.
Cell Commun Signal ; 22(1): 85, 2024 01 30.
Artigo em Inglês | MEDLINE | ID: mdl-38291468

RESUMO

K-Ras is the most frequently mutated Ras variant in pancreatic, colon and non-small cell lung adenocarcinoma. Activating mutations in K-Ras result in increased amounts of active Ras-GTP and subsequently a hyperactivation of effector proteins and downstream signaling pathways. Here, we demonstrate that oncogenic K-Ras(V12) regulates tumor cell migration by activating the phosphatidylinositol 3-kinases (PI3-K)/Akt pathway and induces the expression of E-cadherin and neural cell adhesion molecule (NCAM) by upregulation of Akt3. In vitro interaction and co-precipitation assays identified PI3-Kα as a bona fide effector of active K-Ras4B but not of H-Ras or N-Ras, resulting in enhanced Akt phosphorylation. Moreover, K-Ras(V12)-induced PI3-K/Akt activation enhanced migration in all analyzed cell lines. Interestingly, Western blot analyses with Akt isoform-specific antibodies as well as qPCR studies revealed, that the amount and the activity of Akt3 was markedly increased whereas the amount of Akt1 and Akt2 was downregulated in EGFP-K-Ras(V12)-expressing cell clones. To investigate the functional role of each Akt isoform and a possible crosstalk of the isoforms in more detail, each isoform was stably depleted in PANC-1 pancreatic and H23 lung carcinoma cells. Akt3, the least expressed Akt isoform in most cell lines, is especially upregulated and active in Akt2-depleted cells. Since expression of EGFP-K-Ras(V12) reduced E-cadherin-mediated cell-cell adhesion by induction of polysialylated NCAM, Akt3 was analyzed as regulator of E-cadherin and NCAM. Western blot analyses revealed pronounced reduction of E-cadherin and NCAM in the Akt3-kd cells, whereas Akt1 and Akt2 depletion upregulated E-cadherin, especially in H23 lung carcinoma cells. In summary, we identified oncogenic K-Ras4B as a key regulator of PI3-Kα-Akt signaling and Akt3 as a crucial regulator of K-Ras4B-induced modulation of E-cadherin and NCAM expression and localization.


Assuntos
Adenocarcinoma , Neoplasias Pulmonares , Neoplasias Pancreáticas , Humanos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Moléculas de Adesão de Célula Nervosa , Caderinas , Neoplasias Pulmonares/genética , Isoformas de Proteínas , Fosfatidilinositol 3-Quinases/metabolismo , Pulmão/metabolismo , Neoplasias Pancreáticas/patologia
2.
Int J Numer Method Biomed Eng ; 39(4): e3679, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36606741

RESUMO

The primary cause for cancer-related death is metastasis, and although this phenomenon is the hallmark of cancer, it remains poorly understood. Since studies on the underlying mechanisms are still demanding by experimental means prognostic tools based on computer models can be of great value, not only for elucidating metastasis formation but also for assessing the prospective benefits as well as risks of a therapy for patients with advanced cancer. Here, we present an agent-based model (ABM), describing the complete process of platelet-assisted extravasation of circulating tumor cells (CTCs) from the chemoattraction of blood platelets by the CTCs up to the embedding of the CTCs in the epithelial tissue by computational means. From the simulation results, we conclude that the ABM produces results in consistency with experimental observations, which opens new perspectives for the development of computer models for predicting the efficacity of drug-based tumor therapies and assisting precision medicine approaches.


Assuntos
Células Neoplásicas Circulantes , Humanos , Células Neoplásicas Circulantes/patologia , Plaquetas/patologia , Prognóstico , Simulação por Computador
3.
Int J Mol Sci ; 23(8)2022 Apr 12.
Artigo em Inglês | MEDLINE | ID: mdl-35457076

RESUMO

A Clostridioides difficile infection (CDI) is the most common nosocomial infection worldwide. The main virulence factors of pathogenic C. difficile are TcdA and TcdB, which inhibit small Rho-GTPases. The inhibition of small Rho-GTPases leads to the so-called cytopathic effect, a reorganization of the actin cytoskeleton, an impairment of the colon epithelium barrier function and inflammation. Additionally, TcdB induces a necrotic cell death termed pyknosis in vitro independently from its glucosyltransferases, which are characterized by chromatin condensation and ROS production. To understand the underlying mechanism of this pyknotic effect, we conducted a large-scale phosphoproteomic study. We included the analysis of alterations in the phosphoproteome after treatment with TcdA, which was investigated for the first time. TcdA exhibited no glucosyltransferase-independent necrotic effect and was, thus, a good control to elucidate the underlying mechanism of the glucosyltransferase-independent effect of TcdB. We found RAS to be a central upstream regulator of the glucosyltransferase-independent effect of TcdB. The inhibition of RAS led to a 68% reduction in necrosis. Further analysis revealed apolipoprotein C-III (APOC3) as a possible crucial factor of CDI-induced inflammation in vivo.


Assuntos
Toxinas Bacterianas , Clostridioides difficile , Infecções por Clostridium , Proteínas de Bactérias/metabolismo , Toxinas Bacterianas/metabolismo , Clostridioides , Enterotoxinas/metabolismo , Células Epiteliais/metabolismo , GTP Fosfo-Hidrolases , Glucosiltransferases/metabolismo , Humanos , Inflamação , Necrose
4.
Int J Mol Sci ; 23(4)2022 Feb 11.
Artigo em Inglês | MEDLINE | ID: mdl-35216127

RESUMO

The annual meeting "Signal Transduction-Receptors, Mediators and Genes" of the Signal Transduction Society (STS) is an interdisciplinary conference which is open to all scientists sharing a common interest in the elucidation of the signaling pathways mediating physiological or pathological processes in the health and disease of humans, animals, plants, fungi, prokaryotes, and protists. The 24th meeting on signal transduction was held from 15 to 17 November 2021 in Weimar, Germany. As usual, keynote presentations by invited scientists introduced the respective workshops, and were followed by speakers chosen from the submitted abstracts. A special workshop focused on "Target Identification and Interaction". Ample time was reserved for the discussion of the presented data during the workshops. Unfortunately, due to restrictions owing to the SARS-CoV-2 pandemic, the poster sessions-and thus intensive scientific discussions at the posters-were not possible. In this report, we provide a concise summary of the various workshops and further aspects of the scientific program.


Assuntos
Transdução de Sinais/fisiologia , Pesquisa Biomédica , Alemanha , Sociedades Científicas
5.
Int J Mol Sci ; 22(17)2021 Aug 28.
Artigo em Inglês | MEDLINE | ID: mdl-34502237

RESUMO

Neural crest (NC) cells are highly migratory cells that contribute to various vertebrate tissues, and whose migratory behaviors resemble cancer cell migration and invasion. Information exchange via dynamic NC cell-cell contact is one mechanism by which the directionality of migrating NC cells is controlled. One transmembrane protein that is most likely involved in this process is protein tyrosine kinase 7 (PTK7), an evolutionary conserved Wnt co-receptor that is expressed in cranial NC cells and several tumor cells. In Xenopus, Ptk7 is required for NC migration. In this study, we show that the Ptk7 protein is dynamically localized at cell-cell contact zones of migrating Xenopus NC cells and required for contact inhibition of locomotion (CIL). Using deletion constructs of Ptk7, we determined that the extracellular immunoglobulin domains of Ptk7 are important for its transient accumulation and that they mediate homophilic binding. Conversely, we found that ectopic expression of Ptk7 in non-NC cells was able to prevent NC cell invasion. However, deletion of the extracellular domains of Ptk7 abolished this effect. Thus, Ptk7 is sufficient at protecting non-NC tissue from NC cell invasion, suggesting a common role of PTK7 in contact inhibition, cell invasion, and tissue integrity.


Assuntos
Moléculas de Adesão Celular/metabolismo , Diferenciação Celular , Movimento Celular , Inibição de Contato , Neoplasias Pulmonares/metabolismo , Crista Neural/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo , Animais , Polaridade Celular , Humanos , Neoplasias Pulmonares/patologia , Xenopus laevis
6.
Development ; 147(10)2020 05 22.
Artigo em Inglês | MEDLINE | ID: mdl-32366678

RESUMO

Directional migration during embryogenesis and tumor progression faces the challenge that numerous external signals need to converge to precisely control cell movement. The Rho guanine exchange factor (GEF) Trio is especially well suited to relay signals, as it features distinct catalytic domains to activate Rho GTPases. Here, we show that Trio is required for Xenopus cranial neural crest (NC) cell migration and cartilage formation. Trio cell-autonomously controls protrusion formation of NC cells and Trio morphant NC cells show a blebbing phenotype. Interestingly, the Trio GEF2 domain is sufficient to rescue protrusion formation and migration of Trio morphant NC cells. We show that this domain interacts with the DEP/C-terminus of Dishevelled (DVL). DVL - but not a deletion construct lacking the DEP domain - is able to rescue protrusion formation and migration of Trio morphant NC cells. This is likely mediated by activation of Rac1, as we find that DVL rescues Rac1 activity in Trio morphant embryos. Thus, our data provide evidence for a novel signaling pathway, whereby Trio controls protrusion formation of cranial NC cells by interacting with DVL to activate Rac1.


Assuntos
Movimento Celular/genética , Proteínas Desgrenhadas/metabolismo , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Crista Neural/citologia , Proteínas Serina-Treonina Quinases/metabolismo , Transdução de Sinais/genética , Proteínas de Xenopus/metabolismo , Xenopus laevis/embriologia , Animais , Proteínas Desgrenhadas/genética , Fatores de Troca do Nucleotídeo Guanina/genética , Células HEK293 , Humanos , Crista Neural/embriologia , Fenótipo , Plasmídeos/genética , Ligação Proteica/genética , Domínios Proteicos , Proteínas Serina-Treonina Quinases/genética , Transfecção , Proteínas de Xenopus/genética , Proteínas rac1 de Ligação ao GTP/metabolismo , Proteína rhoA de Ligação ao GTP/metabolismo
7.
Int J Mol Sci ; 21(8)2020 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-32326408

RESUMO

The annual meeting "Signal Transduction-Receptors, Mediators and Genes" of the Signal Transduction Society (STS) is an interdisciplinary conference open to all scientists sharing the common interest in elucidating the signalling pathways underlying the physiological or pathological processes in health and disease of humans, animals, plants, fungi, prokaryotes and protists. The 23rd meeting on signal transduction was held from 4-6 November 2019 in Weimar, Germany, and focused on "Trends in Cancer and Infection". As usual, keynote presentations by invited scientists introduced the respective workshops and were followed by speakers chosen from the submitted abstracts. Ample time had been reserved for discussion of the presented data during the workshops. In this report, we provide a concise summary of the various workshops and further aspects of the scientific program.


Assuntos
Doenças Transmissíveis/imunologia , Neoplasias/imunologia , Neoplasias/metabolismo , Transdução de Sinais , Animais , Doenças Transmissíveis/metabolismo , Doenças Transmissíveis/microbiologia , Doenças Transmissíveis/parasitologia , Alemanha , Humanos , Neoplasias/genética , Transdução de Sinais/imunologia
8.
Biol Chem ; 401(4): 517-531, 2020 03 26.
Artigo em Inglês | MEDLINE | ID: mdl-31811797

RESUMO

Rac1 is a ubiquitously expressed Rho GTPase and an important regulator of the actin cytoskeleton. Its splice variant Rac1b exhibits a 19-amino acid (aa) in-frame insertion and is predominantly active. Both proteins were described in tumorigenesis or metastasis. We investigated the contribution of Rac1 and Rac1b to tumor progression of human non-small-cell lung adenocarcinoma (NSCLA). Rac1 protein was present in 8/8 NSCLA cell lines analyzed, whereas Rac1b was expressed in only 6/8. In wound-healing assays, enhanced green fluorescence protein (EGFP)-Rac1 slightly decreased cell migration, whereas proliferation was increased in both, Rac1- and Rac1b-expressing cells. In the in vivo chorioallantoic invasion model, EGFP-Rac1-expressing cells formed more invasive tumors compared to EGFP-Rac1b. This increased invasiveness correlated with enhanced phosphorylation of p38α, AKT and glycogen synthase kinase 3ß (GSK3ß), and activation of serum response- and Smad-dependent gene promoters by Rac1. In contrast, Rac1b solely activated the mitogen-activated protein kinase (MAPK) JNK2, together with TCF/LEF1- and nuclear factor kappa B (NFκB)-responsive gene reporters. Rac1b, as Rac1, phosphorylated p38α, AKT and GSK3ß. Knockdown of the splicing factor epithelial splicing regulatory protein 1 (ESRP1), which mediates out-splicing of exon 3b from Rac1 pre-messenger RNA, resulted in increased Rac1b messenger RNA (mRNA) and suppression of the epithelial-mesenchymal transition (EMT)-associated transcription factor ZEB1. Our data demonstrate different signaling and functional activities of Rac1 and Rac1b and an important role for Rac1 in lung cancer metastasis.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/metabolismo , Neoplasias Pulmonares/metabolismo , Proteínas rac1 de Ligação ao GTP/metabolismo , Carcinoma Pulmonar de Células não Pequenas/patologia , Transição Epitelial-Mesenquimal/genética , Humanos , Neoplasias Pulmonares/patologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Transdução de Sinais/genética , Células Tumorais Cultivadas , Proteínas rac1 de Ligação ao GTP/análise , Proteínas rac1 de Ligação ao GTP/genética
9.
Cancers (Basel) ; 12(1)2019 Dec 20.
Artigo em Inglês | MEDLINE | ID: mdl-31861875

RESUMO

K-Ras is the most prominent driver of oncogenesis and no effective K-Ras inhibitors have been established despite decades of intensive research. Identifying new K-Ras-binding proteins and their interaction domains offers the opportunity for defining new approaches in tackling oncogenic K-Ras. We have identified Galectin-8 as a novel, direct binding protein for K-Ras4B by mass spectrometry analyses and protein interaction studies. Galectin-8 is a tandem-repeat Galectin and it is widely expressed in lung and pancreatic carcinoma cells. siRNA-mediated depletion of Galectin-8 resulted in increased K-Ras4B content and ERK1/2 activity in lung and pancreatic carcinoma cells. Moreover, cell migration and cell proliferation were inhibited by the depletion of Galectin-8. The K-Ras4B-Galectin-8 interaction is indispensably associated with the farnesylation of K-Ras4B. The lysine-rich polybasic domain (PBD), a region that is unique for K-Ras4B as compared to H- and N-Ras, stabilizes the interaction and accounts for the specificity. Binding assays with the deletion mutants of Galectin-8, comprising either of the two carbohydrate recognition domains (CRD), revealed that K-Ras4B only interacts with the N-CRD, but not with the C-CRD. Structural modeling uncovers a potential binding pocket for the hydrophobic farnesyl chain of K-Ras4B and a cluster of negatively charged amino acids for interaction with the positively charged lysine residues in the N-CRD. Our results demonstrate that Galectin-8 is a new binding partner for K-Ras4B and it interacts via the N-CRD with the farnesylated PBD of K-Ras, thereby modulating the K-Ras effector pathways as well as cell proliferation and migration.

10.
Int J Mol Sci ; 20(1)2019 Jan 08.
Artigo em Inglês | MEDLINE | ID: mdl-30626122

RESUMO

The annual meeting "Signal Transduction-Receptors, Mediators, and Genes" of the Signal Transduction Society (STS) is an interdisciplinary conference open to all scientists sharing the common interest in elucidating signaling pathways in physiological or pathological processes in humans, animals, plants, fungi, prokaryotes, and protists. On the occasion of the 20th anniversary of the STS, the 22nd joint meeting took place in Weimar from 5⁻7 November 2018. With the focus topic "Signaling: From Past to Future" the evolution of the multifaceted research concerning signal transduction since foundation of the society was highlighted. Invited keynote speakers introduced the respective workshop topics and were followed by numerous speakers selected from the submitted abstracts. All presentations were lively discussed during the workshops. Here, we provide a concise summary of the various workshops and further aspects of the scientific program.


Assuntos
Transdução de Sinais , Animais , Morte Celular , Modelos Animais de Doenças , Humanos , Camundongos , Neoplasias/metabolismo , Neoplasias/patologia , Neurônios/metabolismo , Proteína Supressora de Tumor p53/metabolismo
11.
Int J Mol Sci ; 19(2)2018 Feb 12.
Artigo em Inglês | MEDLINE | ID: mdl-29439515

RESUMO

The annual "Joint Meeting Signal Transduction-Receptors, Mediators and Genes" of the Signal Transduction Society (STS) aims to be an interdisciplinary forum for researchers who share a common interest in deciphering signal transduction pathways in normal or transformed cells, in health and disease, in humans and animal models, or in plants or bacteria. The special focus of the 21st annual Joint Meeting, which took place from 8-10 November 2017 in Weimar, was the topic "Metabolism in Health and Disease" and covered multiple aspects of this highly exciting and fast developing research field. Invited keynote speakers introduced the impact of metabolism on tumor immunology, immune cell signaling, and posttranslational modifications in three specific workshops to the audience. Various other aspects of signal transduction were intensively discussed in five additional workshops. Here, we give an overview of the various workshops and further aspects of the scientific program.


Assuntos
Transdução de Sinais , Sociedades Científicas , Distinções e Prêmios , Alemanha , Metabolismo
12.
Sci Rep ; 7(1): 17313, 2017 12 11.
Artigo em Inglês | MEDLINE | ID: mdl-29229918

RESUMO

Prompted by earlier findings that the Rac1-related isoform Rac1b inhibits transforming growth factor (TGF)-ß1-induced canonical Smad signalling, we studied here whether Rac1b also impacts TGF-ß1-dependent non-Smad signalling such as the MKK6-p38 and MEK-ERK mitogen-activated protein kinase (MAPK) pathways and epithelial-mesenchymal transition (EMT). Transient depletion of Rac1b protein in pancreatic cancer cells by RNA interference increased the extent and duration of TGF-ß1-induced phosphorylation of p38 MAPK in a Smad4-independent manner. Rac1b depletion also strongly increased basal ERK activation - independent of the kinase function of the TGF-ß type I receptor ALK5 - and sensitised cells towards further upregulation of phospho-ERK levels by TGF-ß1, while ectopic overexpression of Rac1b had the reverse effect. Rac1b depletion increased an EMT phenotype as evidenced by cell morphology, gene expression of EMT markers, cell migration and growth inhibition. Inhibition of MKK6-p38 or MEK-ERK signalling partially relieved the Rac1b depletion-dependent increase in TGF-ß1-induced gene expression and cell migration. Rac1b depletion also enhanced TGF-ß1 autoinduction of crucial TGF-ß pathway components and decreased that of TGF-ß pathway inhibitors. Our results show that Rac1b antagonises TGF-ß1-dependent EMT by inhibiting MKK6-p38 and MEK-ERK signalling and by controlling gene expression in a way that favors attenuation of TGF-ß signalling.


Assuntos
Transição Epitelial-Mesenquimal , MAP Quinase Quinase 6/metabolismo , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Neoplasias Pancreáticas/patologia , Fator de Crescimento Transformador beta1/farmacologia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Proteínas rac1 de Ligação ao GTP/metabolismo , Movimento Celular , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/metabolismo , Fosforilação , Transdução de Sinais , Células Tumorais Cultivadas
13.
Circ Res ; 121(4): 424-438, 2017 Aug 04.
Artigo em Inglês | MEDLINE | ID: mdl-28620066

RESUMO

RATIONALE: Acute pulmonary oxygen sensing is essential to avoid life-threatening hypoxemia via hypoxic pulmonary vasoconstriction (HPV) which matches perfusion to ventilation. Hypoxia-induced mitochondrial superoxide release has been suggested as a critical step in the signaling pathway underlying HPV. However, the identity of the primary oxygen sensor and the mechanism of superoxide release in acute hypoxia, as well as its relevance for chronic pulmonary oxygen sensing, remain unresolved. OBJECTIVES: To investigate the role of the pulmonary-specific isoform 2 of subunit 4 of the mitochondrial complex IV (Cox4i2) and the subsequent mediators superoxide and hydrogen peroxide for pulmonary oxygen sensing and signaling. METHODS AND RESULTS: Isolated ventilated and perfused lungs from Cox4i2-/- mice lacked acute HPV. In parallel, pulmonary arterial smooth muscle cells (PASMCs) from Cox4i2-/- mice showed no hypoxia-induced increase of intracellular calcium. Hypoxia-induced superoxide release which was detected by electron spin resonance spectroscopy in wild-type PASMCs was absent in Cox4i2-/- PASMCs and was dependent on cysteine residues of Cox4i2. HPV could be inhibited by mitochondrial superoxide inhibitors proving the functional relevance of superoxide release for HPV. Mitochondrial hyperpolarization, which can promote mitochondrial superoxide release, was detected during acute hypoxia in wild-type but not Cox4i2-/- PASMCs. Downstream signaling determined by patch-clamp measurements showed decreased hypoxia-induced cellular membrane depolarization in Cox4i2-/- PASMCs compared with wild-type PASMCs, which could be normalized by the application of hydrogen peroxide. In contrast, chronic hypoxia-induced pulmonary hypertension and pulmonary vascular remodeling were not or only slightly affected by Cox4i2 deficiency, respectively. CONCLUSIONS: Cox4i2 is essential for acute but not chronic pulmonary oxygen sensing by triggering mitochondrial hyperpolarization and release of mitochondrial superoxide which, after conversion to hydrogen peroxide, contributes to cellular membrane depolarization and HPV. These findings provide a new model for oxygen-sensing processes in the lung and possibly also in other organs.


Assuntos
Complexo IV da Cadeia de Transporte de Elétrons/metabolismo , Pulmão/metabolismo , Mitocôndrias/metabolismo , Oxigênio/metabolismo , Animais , Hipóxia Celular/fisiologia , Linhagem Celular Tumoral , Complexo IV da Cadeia de Transporte de Elétrons/genética , Feminino , Humanos , Masculino , Potencial da Membrana Mitocondrial/fisiologia , Camundongos , Camundongos Knockout , Mitocôndrias/genética
14.
Biochim Biophys Acta ; 1863(8): 2027-36, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27155083

RESUMO

Pharmacological inhibition of oxygen sensing prolyl hydroxylase domain enzymes (PHDs) has been shown to preserve renal structure and function in various models of kidney disease. Since transforming growth factor ß-1 (TGFß-1) is one of the major mediators of kidney injury, we investigated if inhibition of PHDs with subsequent stabilization of hypoxia inducible transcription factors (HIF) might interfere with TGFß-1 signaling with special emphasis on its target gene connective tissue growth factor (CTGF). Overnight incubation of human renal tubular cells, primary cells and cell lines, with the PDH inhibitor DMOG increased Smad3 expression, but barely affected Smad2. Both Smads were translocated into the nucleus upon activation of the cells with TGFß-1. Interestingly, Smad3 nuclear localization was enhanced upon pretreatment of the cells with DMOG for several hours, whereas nuclear Smad2 was reduced. This differential localization was independent of Smad2/3 phosphorylation. Reduced nuclear Smad2 correlated with impaired CTGF secretion in DMOG-treated cells and transient downregulation of Smad2 interfered with TGFß-1-induced CTGF synthesis. Furthermore, YAP was confirmed as indispensable transcription factor involved in CTGF synthesis. Nuclear localization of YAP and TAZ was reduced in DMOG-treated cells. Our data thus provide evidence for DMOG-mediated reduction of CTGF expression by regulating the nuclear localization of the transcription factors Smad2, YAP and TAZ. Prolonged inhibition of PHDs was necessary to achieve alterations in cellular localization suggesting an indirect HIF-mediated effect. This mechanism might be extended to other transcription factors and target genes, and may thus represent a novel mechanism of negative regulation of gene expression by PHD inhibition.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Fator de Crescimento do Tecido Conjuntivo/biossíntese , Túbulos Renais/metabolismo , Fosfoproteínas/metabolismo , Prolil Hidroxilases/fisiologia , Proteína Smad2/metabolismo , Proteína Smad3/metabolismo , Fatores de Transcrição/metabolismo , Transporte Ativo do Núcleo Celular , Aciltransferases , Proteínas Adaptadoras de Transdução de Sinal/genética , Aminoácidos Dicarboxílicos/farmacologia , Hipóxia Celular/genética , Células Cultivadas , Fator de Crescimento do Tecido Conjuntivo/antagonistas & inibidores , Fator de Crescimento do Tecido Conjuntivo/fisiologia , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Túbulos Renais/citologia , Oxigênio/metabolismo , Fosfoproteínas/genética , Cultura Primária de Células , Interferência de RNA , RNA Interferente Pequeno/genética , Proteína Smad2/genética , Fator de Crescimento Transformador beta1/fisiologia , Proteínas de Sinalização YAP
15.
PLoS One ; 10(3): e0121589, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25816094

RESUMO

Morphological alterations of cells can lead to modulation of gene expression. An essential link is the MKL1-dependent activation of serum response factor (SRF), which translates changes in the ratio of G- and F-actin into mRNA transcription. SRF activation is only partially characterized in non-transformed epithelial cells. Therefore, the impact of GTPases of the Rho family and changes in F-actin structures were analyzed in renal proximal tubular epithelial cells. Activation of SRF signaling was compared to the regulation of a known MKL1/SRF target gene, connective tissue growth factor (CTGF). In the human proximal tubular cell line HKC-8 overexpression of two actin mutants either favoring or preventing the formation of F-actin fibers regulated SRF-mediated transcription as well as CTGF expression. Only overexpression of constitutively active RhoA activated SRF-dependent gene expression whereas no effect was detected upon overexpression of Rac1 mutants. To elucidate the functional role of Rho kinases as downstream mediators of RhoA, pharmacological inhibition and genetic inhibition by transient siRNA knock down were compared. Upon stimulation with lysophosphatidic acid (LPA) Rho kinase inhibitors partially suppressed SRF-mediated transcription, whereas interference with Rho kinase expression by siRNA reduced activation of SRF, but barely affected CTGF expression. Together with the partial inhibition of CTGF expression by the pharmacological inhibitors Y27432 and H1154, Rho kinases seem to be less important in mediating RhoA signaling related to CTGF expression in HKC-8 epithelial cells. Short term pharmacological inhibition of Rac1 activity by EHT1864 reduced SRF-dependent CTGF expression in HKC-8 cells, but was overcome by a stimulatory effect after prolonged incubation after 4-6 h. Similarly, human primary cells of proximal but not of distal tubular origin showed inhibitory as well as stimulatory effects of Rac1 inhibition. Thus, RhoA signaling activates MKL1-SRF-mediated CTGF expression in proximal tubular cells, whereas Rac1 signaling is more complex with adaptive cellular responses.


Assuntos
Actinas/metabolismo , Células Epiteliais/metabolismo , Túbulos Renais Proximais/metabolismo , Transdução de Sinais , Proteínas rac1 de Ligação ao GTP/metabolismo , Proteína rhoA de Ligação ao GTP/metabolismo , Células Cultivadas , Fator de Crescimento do Tecido Conjuntivo/metabolismo , Células Epiteliais/citologia , Humanos , Túbulos Renais Proximais/citologia , Lisofosfolipídeos/farmacologia , Mutação , RNA Interferente Pequeno/farmacologia , Fator de Resposta Sérica/metabolismo , Transdução de Sinais/efeitos dos fármacos , Proteínas rac1 de Ligação ao GTP/genética , Proteína rhoA de Ligação ao GTP/genética
16.
Oncotarget ; 5(1): 277-90, 2014 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-24378395

RESUMO

Transforming growth factor (TGF)-ß1 promotes progression of pancreatic ductal adenocarcinoma (PDAC) by enhancing epithelial-mesenchymal transition, cell migration/invasion, and metastasis, in part by cooperating with the small GTPase Rac1. Prompted by the observation of higher expression of Rac1b, an alternatively spliced Rac1 isoform, in pancreatic ductal epithelial cells and in patients with chronic pancreatitis vs. PDAC, as well as in long-time vs. short-time survivors among PDAC patients, we asked whether Rac1b might negatively affect TGF-ß1 prometastatic function. Interestingly, the non-malignant pancreatic ductal epithelial cell line H6c7 exhibited a higher ratio of active Rac1b to total Rac1b than the TGF-ß1-responsive PDAC cell lines Panc-1 and Colo357. Notably, siRNA-mediated silencing of Rac1b increased TGF-ß1/Smad-dependent migratory activities in H6c7, Colo357, and Panc-1 cells, while ectopic overexpression of Rac1b in Panc-1 cells attenuated TGF-ß1-induced cell motility. Depletion of Rac1b in Panc-1 cells enhanced TGF-ß1/Smad-dependent expression of promoter-reporter genes and of the endogenous Slug gene. Moreover, Rac1b depletion resulted in a higher and more sustained C-terminal phosphorylation of Smad3 and Smad2, suggesting that Rac1b is involved in Smad2/3 dephosphorylation/inactivation. Since pharmacologic or siRNA-mediated inhibition of Smad3 but not Smad2 was able to alleviate the Rac1b siRNA effect on TGF-ß1-induced cell migration, our results suggests that Rac1b inhibits TGF-ß1-induced cell motility in pancreatic ductal epithelial cells by blocking the function of Smad3. Moreover, Rac1b may act as an endogenous inhibitor of Rac1 in TGF-ß1-mediated migration and possibly metastasis. Hence, it could be exploited for diagnostic/prognostic purposes or even therapeutically in late-stage PDAC as an antimetastatic agent.


Assuntos
Carcinoma Ductal Pancreático/patologia , Movimento Celular/fisiologia , Neoplasias Pancreáticas/patologia , Proteína Smad2/metabolismo , Proteína Smad3/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Proteínas rac1 de Ligação ao GTP/metabolismo , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/metabolismo , Linhagem Celular Tumoral , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Transição Epitelial-Mesenquimal , Guanosina Trifosfato/metabolismo , Humanos , Ductos Pancreáticos/citologia , Ductos Pancreáticos/metabolismo , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Fosforilação , Transdução de Sinais , Transfecção , Proteínas rac1 de Ligação ao GTP/genética
17.
Cell Commun Signal ; 11: 80, 2013 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-24144209

RESUMO

BACKGROUND: Hypoxia is a major driving force in vascularization and vascular remodeling. Pharmacological inhibition of prolyl hydroxylases (PHDs) leads to an oxygen-independent and long-lasting activation of hypoxia-inducible factors (HIFs). Whereas effects of HIF-stabilization on transcriptional responses have been thoroughly investigated in endothelial cells, the molecular details of cytoskeletal changes elicited by PHD-inhibition remain largely unknown. To investigate this important aspect of PHD-inhibition, we used a spheroid-on-matrix cell culture model. RESULTS: Microvascular endothelial cells (glEND.2) were organized into spheroids. Migration of cells from the spheroids was quantified and analyzed by immunocytochemistry. The PHD inhibitor dimethyloxalyl glycine (DMOG) induced F-actin stress fiber formation in migrating cells, but only weakly affected microvascular endothelial cells firmly attached in a monolayer. Compared to control spheroids, the residual spheroids were larger upon PHD inhibition and contained more cells with tight VE-cadherin positive cell-cell contacts. Morphological alterations were dependent on stabilization of HIF-1α and not HIF-2α as shown in cells with stable knockdown of HIF-α isoforms. DMOG-treated endothelial cells exhibited a reduction of immunoreactive Rac-1 at the migrating front, concomitant with a diminished Rac-1 activity, whereas total Rac-1 protein remained unchanged. Two chemically distinct Rac-1 inhibitors mimicked the effects of DMOG in terms of F-actin fiber formation and orientation, as well as stabilization of residual spheroids. Furthermore, phosphorylation of p21-activated kinase PAK downstream of Rac-1 was reduced by DMOG in a HIF-1α-dependent manner. Stabilization of cell-cell contacts associated with decreased Rac-1 activity was also confirmed in human umbilical vein endothelial cells. CONCLUSIONS: Our data demonstrates that PHD inhibition induces HIF-1α-dependent cytoskeletal remodeling in endothelial cells, which is mediated essentially by a reduction in Rac-1 signaling.


Assuntos
Citoesqueleto de Actina/metabolismo , Células Endoteliais/efeitos dos fármacos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Microvasos/efeitos dos fármacos , Inibidores de Prolil-Hidrolase/farmacologia , Proteínas rac1 de Ligação ao GTP/metabolismo , Citoesqueleto de Actina/ultraestrutura , Actinas/metabolismo , Aminoácidos Dicarboxílicos/farmacologia , Movimento Celular , Células Endoteliais/fisiologia , Células Endoteliais/ultraestrutura , Células Endoteliais da Veia Umbilical Humana , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Microvasos/fisiologia , Microvasos/ultraestrutura , Transdução de Sinais , Esferoides Celulares/citologia , Esferoides Celulares/fisiologia , Quinases Ativadas por p21/metabolismo , Proteínas rac1 de Ligação ao GTP/genética , Quinases Associadas a rho/metabolismo
19.
Arch Biochem Biophys ; 524(1): 48-55, 2012 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-22583808

RESUMO

The molecular mechanisms leading to tumor progression and acquisition of a metastatic phenotype are highly complex and only partially understood. The spatiotemporal regulation of E-cadherin-mediated adherens junctions is essential for normal epithelia function and tissue integrity. Perturbation of the E-cadherin complex assembly is a key event in epithelial-mesenchymal transition and is directed by a huge number of mechanisms that differ greatly with regard to cell types and tissues. The reduction in intercellular adhesion interferes with tissue integrity and allows cancer cells to disseminate from the primary tumor thereby initiating cancer metastasis. In the present review we will summarize the current findings about the influence of Rho GTPases on the formation and maintenance of adherens junction and will then proceed to discuss the involvement of p120-catenin on cell-cell adhesion and tumor cell migration.


Assuntos
Junções Aderentes/metabolismo , Cateninas/metabolismo , Proteínas rho de Ligação ao GTP/metabolismo , Junções Aderentes/patologia , Animais , Adesão Celular , Humanos , Metástase Neoplásica/patologia , Neoplasias/metabolismo , Neoplasias/patologia , delta Catenina
20.
Int J Radiat Oncol Biol Phys ; 81(5): 1506-14, 2011 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-21985943

RESUMO

PURPOSE: It is known that postirradiation survival of tumor cells presenting mutated K-RAS is mediated through autocrine activation of epidermal growth factor receptor (EGFR). In this study the molecular mechanism of radioresistance of cells overexpressing mutated K-RAS(V12) was investigated. METHODS AND MATERIALS: Head-and-neck cancer cells (FaDu) presenting wild-type K-RAS were transfected with empty vector or vector expressing mutated K-RAS(V12). The effect of K-RAS(V12) on autocrine production of EGFR ligands, activation of EGFR downstream pathways, DNA damage repair, and postirradiation survival was analyzed. RESULTS: Conditioned medium collected from K-RAS(V12)-transfected cells enhanced activation of the phosphatidylinositol-3-kinase-Akt pathway and increased postirradiation survival of wild-type K-RAS parental cells when compared with controls. These effects were reversed by amphiregulin (AREG)-neutralizing antibody. In addition, secretion of the EGFR ligands AREG and transforming growth factor α was significantly increased upon overexpression of K-RAS(V12). Expression of mutated K-RAS(V12) resulted in an increase in radiation-induced DNA-dependent protein kinase catalytic subunit (DNA-PKcs) phosphorylation at S2056. This increase was accompanied by increased repair of DNA double-strand breaks. Abrogation of DNA-PKcs phosphorylation by serum depletion or AREG-neutralizing antibody underscored the role of autocrine production of EGFR ligands, namely, AREG, in regulating DNA-PKcs activation in K-RAS mutated cells. CONCLUSIONS: These data indicate that radioresistance of K-RAS mutated tumor cells is at least in part due to constitutive production of EGFR ligands, which mediate enhanced repair of DNA double-strand breaks through the EGFR-phosphatidylinositol-3-kinase-Akt cascade.


Assuntos
Reparo do DNA/fisiologia , Proteína Quinase Ativada por DNA/metabolismo , Receptores ErbB/metabolismo , Glicoproteínas/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Proteínas Proto-Oncogênicas p21(ras)/fisiologia , Tolerância a Radiação/fisiologia , Fator de Crescimento Transformador alfa/metabolismo , Anfirregulina , Anticorpos Monoclonais/farmacologia , Linhagem Celular Tumoral , Sobrevivência Celular/fisiologia , Meios de Cultivo Condicionados , Quebras de DNA de Cadeia Dupla , Família de Proteínas EGF , Genes ras , Glicoproteínas/imunologia , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/imunologia , Ligantes , Mutação , Fosfatidilinositol 3-Quinase/metabolismo , Fosforilação , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Proto-Oncogênicas p21(ras)/genética , Transdução de Sinais/fisiologia , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...