Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 88
Filtrar
1.
Cell Rep ; 42(4): 112324, 2023 04 25.
Artigo em Inglês | MEDLINE | ID: mdl-37000626

RESUMO

Patient-derived organoids (PDOs) are widely heralded as a drug-screening platform to develop new anti-cancer therapies. Here, we use a drug-repurposing library to screen PDOs of colorectal cancer (CRC) to identify hidden vulnerabilities within therapy-induced phenotypes. Using a microscopy-based screen that accurately scores drug-induced cell killing, we have tested 414 putative anti-cancer drugs for their ability to switch the EGFRi/MEKi-induced cytostatic phenotype toward cytotoxicity. A majority of validated hits (9/37) are microtubule-targeting agents that are commonly used in clinical oncology, such as taxanes and vinca-alkaloids. One of these drugs, vinorelbine, is consistently effective across a panel of >25 different CRC PDOs, independent of RAS mutational status. Unlike vinorelbine alone, its combination with EGFR/MEK inhibition induces apoptosis at all stages of the cell cycle and shows tolerability and effective anti-tumor activity in vivo, setting the basis for a clinical trial to treat patients with metastatic RAS-mutant CRC.


Assuntos
Antineoplásicos , Neoplasias do Colo , Neoplasias Colorretais , Humanos , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Neoplasias Colorretais/tratamento farmacológico , Neoplasias Colorretais/genética , Neoplasias Colorretais/patologia , Vinorelbina/farmacologia , Vinorelbina/uso terapêutico , Reposicionamento de Medicamentos , Linhagem Celular Tumoral , Neoplasias do Colo/tratamento farmacológico , Neoplasias do Colo/genética , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Organoides/metabolismo
2.
Nat Cell Biol ; 23(4): 377-390, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33795873

RESUMO

Direct targeting of the downstream mitogen-activated protein kinase (MAPK) pathway to suppress extracellular-regulated kinase (ERK) activation in KRAS and BRAF mutant colorectal cancer (CRC) has proven clinically unsuccessful, but promising results have been obtained with combination therapies including epidermal growth factor receptor (EGFR) inhibition. To elucidate the interplay between EGF signalling and ERK activation in tumours, we used patient-derived organoids (PDOs) from KRAS and BRAF mutant CRCs. PDOs resemble in vivo tumours, model treatment response and are compatible with live-cell microscopy. We established real-time, quantitative drug response assessment in PDOs with single-cell resolution, using our improved fluorescence resonance energy transfer (FRET)-based ERK biosensor EKAREN5. We show that oncogene-driven signalling is strikingly limited without EGFR activity and insufficient to sustain full proliferative potential. In PDOs and in vivo, upstream EGFR activity rigorously amplifies signal transduction efficiency in KRAS or BRAF mutant MAPK pathways. Our data provide a mechanistic understanding of the effectivity of EGFR inhibitors within combination therapies against KRAS and BRAF mutant CRC.


Assuntos
Neoplasias Colorretais/tratamento farmacológico , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas B-raf/genética , Proteínas Proto-Oncogênicas p21(ras)/genética , Linhagem Celular Tumoral , Neoplasias Colorretais/genética , Neoplasias Colorretais/patologia , Receptores ErbB/genética , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Quinases de Proteína Quinase Ativadas por Mitógeno/genética , Mutação , Organoides/metabolismo , Organoides/patologia , Análise de Célula Única
3.
Small GTPases ; 11(5): 346-353, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-29388865

RESUMO

Epac1 and Rap1 mediate cAMP-induced tightening of endothelial junctions. We have previously found that one of the mechanisms is the inhibition of Rho-mediated tension in radial stress fibers by recruiting the RhoGAP ArhGAP29 in a complex containing the Rap1 effectors Rasip1 and Radil. However, other mechanisms have been proposed as well, most notably the induction of tension in circumferential actin cables by Cdc42 and its GEF FGD5. Here, we have investigated how Rap1 controls FGD5/Cdc42 and how this interconnects with Radil/Rasip1/ArhGAP29. Using endothelial barrier measurements, we show that Rho inhibition is not sufficient to explain the barrier stimulating effect of Rap1. Indeed, Cdc42-mediated tension is induced at cell-cell contacts upon Rap1 activation and this is required for endothelial barrier function. Depletion of potential Rap1 effectors identifies AF6 to mediate Rap1 enhanced tension and concomitant Rho-independent barrier function. When overexpressed in HEK293T cells, AF6 is found in a complex with FGD5 and Radil. From these results we conclude that Rap1 utilizes multiple pathways to control tightening of endothelial junctions, possibly through a multiprotein effector complex, in which AF6 functions to induce tension in circumferential actin cables.


Assuntos
Células Endoteliais/metabolismo , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Cinesinas/metabolismo , Miosinas/metabolismo , Junções Íntimas/metabolismo , Proteínas rap1 de Ligação ao GTP/metabolismo , Células Cultivadas , Células HEK293 , Humanos
4.
Nat Med ; 25(5): 838-849, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-31011202

RESUMO

Ovarian cancer (OC) is a heterogeneous disease usually diagnosed at a late stage. Experimental in vitro models that faithfully capture the hallmarks and tumor heterogeneity of OC are limited and hard to establish. We present a protocol that enables efficient derivation and long-term expansion of OC organoids. Utilizing this protocol, we have established 56 organoid lines from 32 patients, representing all main subtypes of OC. OC organoids recapitulate histological and genomic features of the pertinent lesion from which they were derived, illustrating intra- and interpatient heterogeneity, and can be genetically modified. We show that OC organoids can be used for drug-screening assays and capture different tumor subtype responses to the gold standard platinum-based chemotherapy, including acquisition of chemoresistance in recurrent disease. Finally, OC organoids can be xenografted, enabling in vivo drug-sensitivity assays. Taken together, this demonstrates their potential application for research and personalized medicine.


Assuntos
Organoides/patologia , Neoplasias Ovarianas/patologia , Adulto , Idoso , Animais , Linhagem Celular Tumoral , Ensaios de Seleção de Medicamentos Antitumorais , Feminino , Genômica , Xenoenxertos , Humanos , Camundongos SCID , Pessoa de Meia-Idade , Mutação , Neoplasias Ovarianas/tratamento farmacológico , Neoplasias Ovarianas/genética , Medicina de Precisão
5.
Oncotarget ; 10(14): 1440-1457, 2019 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-30858928

RESUMO

Anti-EGFR therapy is used to treat metastatic colorectal cancer (CRC) patients, for which initial response rates of 10-20% have been achieved. Although the presence of HER2 amplifications and oncogenic mutations in KRAS, NRAS, and BRAF are associated with EGFR-targeted therapy resistance, for a large population of CRC patients the underlying mechanism of RAS-MEK-ERK hyperactivation is not clear. Loss-of-function mutations in RASGAPs are often speculated in literature to promote CRC growth as being negative regulators of RAS, but direct experimental evidence is lacking. We generated a CRISPR-mediated knock out panel of all RASGAPs in patient-derived CRC organoids and found that only loss of NF1, but no other RASGAPs e.g. RASA1, results in enhanced RAS-ERK signal amplification and improved tolerance towards limited EGF stimulation. Our data suggests that NF1-deficient CRCs are likely not responsive to anti-EGFR monotherapy and can potentially function as a biomarker for CRC progression.

6.
PLoS One ; 13(11): e0207159, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30408122

RESUMO

Apico-basal polarity establishment is a seminal process in tissue morphogenesis. To function properly it is often imperative that epithelial cells limit apical membrane formation to a single domain. We previously demonstrated that signaling by the small GTPase Cdc42, together with its guanine nucleotide exchange factor (GEF) Tuba, is required to prevent the formation of multiple apical domains in polarized Ls174T:W4 cells, a single cell model for enterocyte polarization. To further chart the molecular signaling mechanisms that safeguard singularity during enterocyte polarization we generated knockout cells for the Cdc42 effector protein Par6A. Par6A loss results in the formation of multiple apical domains, similar to loss of Cdc42. In Par6A knockout cells, we find that active Cdc42 is more mobile at the apical membrane compared to control cells and that wild type Cdc42 is more diffusely localized throughout the cell, indicating that Par6A is required to restrict Cdc42 signaling. Par6A, Cdc42 and its GEF Tuba bind in a co-immunoprecipitation experiment and they partially colocalize at the apical membrane in polarized Ls174T:W4 cells, suggesting the formation of a trimeric complex. Indeed, in a rescue experiment using Par6A mutants, we show that the ability to establish this trimeric complex correlates with the ability to restore singularity in Par6A knockout cells. Together, these experiments therefore indicate that a Tuba/Cdc42/Par6A complex is required to ensure the formation of a single apical domain during enterocyte polarization.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Polaridade Celular/fisiologia , Proteínas do Citoesqueleto/metabolismo , Enterócitos/citologia , Enterócitos/metabolismo , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Proteína cdc42 de Ligação ao GTP/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/química , Proteínas Adaptadoras de Transdução de Sinal/genética , Linhagem Celular , Polaridade Celular/genética , Proteínas do Citoesqueleto/química , Técnicas de Inativação de Genes , Fatores de Troca do Nucleotídeo Guanina/química , Humanos , Microvilosidades/metabolismo , Microvilosidades/ultraestrutura , Estrutura Quaternária de Proteína , Transdução de Sinais , Proteína cdc42 de Ligação ao GTP/química
7.
Mol Cell Biol ; 38(12)2018 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-29581186

RESUMO

PTEN is a tumor suppressor that is frequently lost in epithelial malignancies. A part of the tumor-suppressive properties of PTEN is attributed to its function in cell polarization and consequently its role in maintaining epithelial tissue integrity. However, surprisingly little is known about the function and regulation of PTEN during epithelial cell polarization. We used clustered regularly interspaced short palindromic repeat (CRISPR)/Cas9-mediated gene disruption to delete PTEN in intestinal epithelial Ls174T:W4 cells, which upon differentiation form a microvillus-covered apical membrane (brush border) on a part of the cell cortex, independent of cell-cell junctions. We show that loss of PTEN results in the formation of a larger brush border that, in a fraction of the cells, even spans the entire plasma membrane, revealing that PTEN functions in the regulation of apical membrane size. Depletion of the phosphatase PTPL1 resulted in a similar defect. PTPL1 interacts with PTEN, and this interaction is necessary for apical membrane enrichment of PTEN. Importantly, phosphatase activity of PTPL1 is not required, indicating that PTPL1 functions as an anchor protein in this process. Our work thus demonstrates a novel function for PTEN during cell polarization in controlling apical membrane size and identifies PTPL1 as a critical apical membrane anchor for PTEN in this process.


Assuntos
Membrana Celular/metabolismo , Polaridade Celular/fisiologia , Microvilosidades/metabolismo , PTEN Fosfo-Hidrolase/metabolismo , Proteína Tirosina Fosfatase não Receptora Tipo 13/genética , Animais , Sistemas CRISPR-Cas/genética , Linhagem Celular Tumoral , Células Epiteliais/fisiologia , Técnicas de Inativação de Genes , Células HEK293 , Humanos , Masculino , Camundongos , Microvilosidades/genética , Neoplasias/patologia , PTEN Fosfo-Hidrolase/genética
8.
Artigo em Inglês | MEDLINE | ID: mdl-28778969

RESUMO

Our laboratory has studied Ras and Ras-like proteins since the discovery of the Ras oncogene 35 years ago. In this review, I will give an account of what we have done in these 35 years and indicate the main papers that have guided our research. Our efforts started with the early analysis of mutant Ras in human tumors followed by deciphering of the role of Ras in signal transduction pathways. In an attempt to interfere in Ras signaling we turned to Rap proteins. These proteins are the closest relatives of Ras and were initially identified as Ras antagonists. However, our studies revealed that the Rap signaling network primarily is involved in spatiotemporal control of cell adhesion, in part through regulation of the actin cytoskeleton. More recently we returned to Ras, trying to interfere in Ras signaling by combinatorial drug testing using the organoid technology.


Assuntos
Oncogenes/fisiologia , Proteínas rap de Ligação ao GTP/metabolismo , Proteínas ras/metabolismo , Animais , Adesão Celular , Humanos , Mutação , Transdução de Sinais/fisiologia , Proteínas rap de Ligação ao GTP/genética
9.
Mol Cell Biol ; 37(7)2017 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-28069739

RESUMO

Signaling by the small GTPase Cdc42 governs a diverse set of cellular processes that contribute to tissue morphogenesis. Since these processes often require highly localized signaling, Cdc42 activity must be clustered in order to prevent ectopic signaling. During cell polarization, apical Cdc42 signaling directs the positioning of the nascent apical membrane. However, the molecular mechanisms that drive Cdc42 clustering during polarity establishment are largely unknown. Here, we demonstrate that during cell polarization localized Cdc42 signaling is enabled via activity-dependent control of Cdc42 mobility. By performing photoconversion experiments, we show that inactive Cdc42-GDP is 30-fold more mobile than active Cdc42-GTP. This switch in apical mobility originates from a dual mechanism involving RhoGDI-mediated membrane dissociation of Cdc42-GDP and Tuba-mediated immobilization of Cdc42-GTP. Interference with either mechanism affects Cdc42 clustering and as a consequence impairs Cdc42-mediated apical membrane clustering. We therefore identify a molecular network, comprised of Cdc42, the guanine nucleotide exchange factor (GEF) Tuba, and RhoGDI, that enables differential diffusion of inactive and active Cdc42 and is required to establish localized Cdc42 signaling during enterocyte polarization.


Assuntos
Polaridade Celular , Enterócitos/citologia , Enterócitos/metabolismo , Transdução de Sinais , Proteína cdc42 de Ligação ao GTP/metabolismo , Linhagem Celular , Membrana Celular/metabolismo , Células HEK293 , Humanos , Microvilosidades/metabolismo , Ligação Proteica , Tubulina (Proteína)/metabolismo , Inibidores da Dissociação do Nucleotídeo Guanina rho-Específico
10.
Structure ; 24(12): 2039-2040, 2016 12 06.
Artigo em Inglês | MEDLINE | ID: mdl-27926831

RESUMO

In this issue of Structure, Gingras et al. (2016) show that Ras association (RA) domains of the Rap1 and Ras interacting protein Rasip1 can form a dimer in the presence and absence of the small G protein Rap1. This provides an explanation for the observed complex formation in Rap1-mediated signaling.


Assuntos
Proteínas Monoméricas de Ligação ao GTP , Proteínas rap1 de Ligação ao GTP , Transdução de Sinais , Proteínas ras
11.
Elife ; 52016 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-27845624

RESUMO

Colorectal cancer (CRC) organoids can be derived from almost all CRC patients and therefore capture the genetic diversity of this disease. We assembled a panel of CRC organoids carrying either wild-type or mutant RAS, as well as normal organoids and tumor organoids with a CRISPR-introduced oncogenic KRAS mutation. Using this panel, we evaluated RAS pathway inhibitors and drug combinations that are currently in clinical trial for RAS mutant cancers. Presence of mutant RAS correlated strongly with resistance to these targeted therapies. This was observed in tumorigenic as well as in normal organoids. Moreover, dual inhibition of the EGFR-MEK-ERK pathway in RAS mutant organoids induced a transient cell-cycle arrest rather than cell death. In vivo drug response of xenotransplanted RAS mutant organoids confirmed this growth arrest upon pan-HER/MEK combination therapy. Altogether, our studies demonstrate the potential of patient-derived CRC organoid libraries in evaluating inhibitors and drug combinations in a preclinical setting.


Assuntos
Antineoplásicos/isolamento & purificação , Antineoplásicos/farmacologia , Neoplasias Colorretais/tratamento farmacológico , Avaliação Pré-Clínica de Medicamentos/métodos , Proteínas Mutantes/antagonistas & inibidores , Organoides/efeitos dos fármacos , Proteínas ras/antagonistas & inibidores , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas , Humanos , Recombinação Genética
12.
J Cell Biol ; 210(7): 1055-63, 2015 Sep 28.
Artigo em Inglês | MEDLINE | ID: mdl-26416959

RESUMO

During yeast cell polarization localization of the small GTPase, cell division control protein 42 homologue (Cdc42) is clustered to ensure the formation of a single bud. Here we show that the disease-associated flippase ATPase class I type 8b member 1 (ATP8B1) enables Cdc42 clustering during enterocyte polarization. Loss of this regulation results in increased apical membrane size with scattered apical recycling endosomes and permits the formation of more than one apical domain, resembling the singularity defect observed in yeast. Mechanistically, we show that to become apically clustered, Cdc42 requires the interaction between its polybasic region and negatively charged membrane lipids provided by ATP8B1. Disturbing this interaction, either by ATP8B1 depletion or by introduction of a Cdc42 mutant defective in lipid binding, increases Cdc42 mobility and results in apical membrane enlargement. Re-establishing Cdc42 clustering, by tethering it to the apical membrane or lowering its diffusion, restores normal apical membrane size in ATP8B1-depleted cells. We therefore conclude that singularity regulation by Cdc42 is conserved between yeast and human and that this regulation is required to maintain healthy tissue architecture.


Assuntos
Adenosina Trifosfatases/metabolismo , Polaridade Celular/fisiologia , Enterócitos/metabolismo , Lipídeos de Membrana/metabolismo , Proteínas de Transferência de Fosfolipídeos/metabolismo , Transdução de Sinais/fisiologia , Proteína cdc42 de Ligação ao GTP/metabolismo , Adenosina Trifosfatases/genética , Animais , Linhagem Celular , Enterócitos/citologia , Humanos , Lipídeos de Membrana/genética , Camundongos , Proteínas de Transferência de Fosfolipídeos/genética , Proteína cdc42 de Ligação ao GTP/genética
13.
PLoS Biol ; 13(1): e1002038, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25603503

RESUMO

The second messenger cAMP is known to augment glucose-induced insulin secretion. However, its downstream targets in pancreatic ß-cells have not been unequivocally determined. Therefore, we designed cAMP analogues by a structure-guided approach that act as Epac2-selective agonists both in vitro and in vivo. These analogues activate Epac2 about two orders of magnitude more potently than cAMP. The high potency arises from increased affinity as well as increased maximal activation. Crystallographic studies demonstrate that this is due to unique interactions. At least one of the Epac2-specific agonists, Sp-8-BnT-cAMPS (S-220), enhances glucose-induced insulin secretion in human pancreatic cells. Selective targeting of Epac2 is thus proven possible and may be an option in diabetes treatment.


Assuntos
AMP Cíclico/análogos & derivados , AMP Cíclico/química , Fatores de Troca do Nucleotídeo Guanina/agonistas , Sítios de Ligação , Linhagem Celular Tumoral , Cristalografia por Raios X , AMP Cíclico/farmacologia , Desenho de Fármacos , Fatores de Troca do Nucleotídeo Guanina/química , Fatores de Troca do Nucleotídeo Guanina/fisiologia , Humanos , Insulina/metabolismo , Secreção de Insulina , Ilhotas Pancreáticas/efeitos dos fármacos , Ilhotas Pancreáticas/metabolismo , Modelos Moleculares , Ligação Proteica
14.
PLoS One ; 9(9): e106687, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25203140

RESUMO

Brush border formation during polarity establishment of intestinal epithelial cells is uniquely governed by the Rap2A GTPase, despite expression of the other highly similar Rap2 isoforms (Rap2B and Rap2C). We investigated the mechanisms of this remarkable specificity and found that Rap2C is spatially segregated from Rap2A signaling as it is not enriched at the apical membrane after polarization. In contrast, both Rap2A and Rap2B are similarly located at Rab11 positive apical recycling endosomes and inside the brush border. However, although Rap2B localizes similarly it is not equally activated as Rap2A during brush border formation. We reveal that the C-terminal hypervariable region allows selective activation of Rap2A, yet this selectivity does not originate from the known differential lipid modifications of this region. In conclusion, we demonstrate that Rap2 specificity during brush border formation is determined by two distinct mechanisms involving segregated localization and selective activation.


Assuntos
Enterócitos/ultraestrutura , Microvilosidades/metabolismo , Transdução de Sinais , Proteínas rap de Ligação ao GTP/metabolismo , Linhagem Celular , Enterócitos/citologia , Humanos , Isoformas de Proteínas/metabolismo , Transporte Proteico
15.
PLoS One ; 9(5): e98253, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24858808

RESUMO

PDZGEF is a guanine nucleotide exchange factor for the small G protein Rap. It was recently found that PDZGEF contributes to establishment of intestinal epithelial polarity downstream of the kinase Lkb1. By binding to phosphatidic acid enriched at the apical membrane, PDZGEF locally activates Rap2a resulting in induction of brush border formation via a pathway that includes the polarity players TNIK, Mst4 and Ezrin. Here we show that the PDZ domain of PDZGEF is essential and sufficient for targeting PDZGEF to the apical membrane of polarized intestinal epithelial cells. Inhibition of PLD and consequently production of phosphatidic acid inhibitis targeting of PDZGEF to the plasma membrane. Furthermore, localization requires specific positively charged residues within the PDZ domain. We conclude that local accumulation of PDZGEF at the apical membrane during establishment of epithelial polarity is mediated by electrostatic interactions between positively charged side chains in the PDZ domain and negatively charged phosphatidic acid.


Assuntos
Polaridade Celular/fisiologia , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Mucosa Intestinal/metabolismo , Microvilosidades/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Ácidos Fosfatídicos/metabolismo , Membrana Celular/genética , Membrana Celular/metabolismo , Membrana Celular/ultraestrutura , Fatores de Troca do Nucleotídeo Guanina/genética , Células HEK293 , Humanos , Mucosa Intestinal/ultraestrutura , Microvilosidades/genética , Microvilosidades/ultraestrutura , Proteínas do Tecido Nervoso/genética , Ácidos Fosfatídicos/genética , Proteínas rap de Ligação ao GTP/genética , Proteínas rap de Ligação ao GTP/metabolismo
16.
Nat Rev Mol Cell Biol ; 15(5): 357-62, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24739740

RESUMO

The Dishevelled, EGL-10 and pleckstrin (DEP) domain is a globular protein domain that is present in about ten human protein families with well-defined structural features. A picture is emerging that DEP domains mainly function in the spatial and temporal control of diverse signal transduction events by recruiting proteins to the plasma membrane. DEP domains can interact with various partners at the membrane, including phospholipids and membrane receptors, and their binding is subject to regulation.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Fosfoproteínas/metabolismo , Proteínas RGS/metabolismo , Transdução de Sinais , Proteínas Adaptadoras de Transdução de Sinal/química , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Membrana Celular/metabolismo , Proteínas Desgrenhadas , Proteínas de Drosophila , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Regulação da Expressão Gênica , Fatores de Troca do Nucleotídeo Guanina/química , Fatores de Troca do Nucleotídeo Guanina/genética , Humanos , Modelos Moleculares , Fosfoproteínas/química , Fosfoproteínas/genética , Ligação Proteica , Estrutura Terciária de Proteína , Proteínas RGS/química , Proteínas RGS/genética
17.
Cell Adh Migr ; 8(2): 100-7, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24714377

RESUMO

The small G-protein Rap1 plays an important role in the regulation of endothelial barrier function, a process controlled largely by cellâ€"cell adhesions and their connection to the actin cytoskeleton. During the various stages of barrier dynamics, different guanine nucleotide exchange factors (GEFs) control Rap1 activity, indicating that Rap1 integrates multiple input signals. Once activated, Rap1 induces numerous signaling cascades, together responsible for the increased endothelial barrier function. Most notably, Rap1 activation results in the inhibition of Rho to decrease radial stress fibers and the activation of Cdc42 to increase junctional actin. This implies that Rap regulates endothelial barrier function by dual control of cytoskeletal tension. The molecular details of the signaling pathways are becoming to be elucidated.


Assuntos
Citoesqueleto de Actina/metabolismo , Adesão Celular/genética , Células Endoteliais/metabolismo , Proteínas de Ligação a Telômeros/genética , Fatores de Troca do Nucleotídeo Guanina/genética , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteína KRIT1 , Proteínas Associadas aos Microtúbulos/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Complexo Shelterina , Transdução de Sinais/genética , Fibras de Estresse/genética , Proteínas de Ligação a Telômeros/metabolismo , Proteína cdc42 de Ligação ao GTP/metabolismo , Proteínas rho de Ligação ao GTP/metabolismo
18.
Dev Cell ; 27(5): 574-85, 2013 Dec 09.
Artigo em Inglês | MEDLINE | ID: mdl-24290981

RESUMO

Epithelial cell migration is crucial for the development and regeneration of epithelial tissues. Aberrant regulation of epithelial cell migration has a major role in pathological processes such as the development of cancer metastasis and tissue fibrosis. Here, we report that in response to factors that promote cell motility, the Rap guanine exchange factor RAPGEF2 is rapidly phosphorylated by I-kappa-B-kinase-ß and casein kinase-1α and consequently degraded by the proteasome via the SCF(ßTrCP) ubiquitin ligase. Failure to degrade RAPGEF2 in epithelial cells results in sustained activity of Rap1 and inhibition of cell migration induced by HGF, a potent metastatic factor. Furthermore, expression of a degradation-resistant RAPGEF2 mutant greatly suppresses dissemination and metastasis of human breast cancer cells. These findings reveal a molecular mechanism regulating migration and invasion of epithelial cells and establish a key direct link between IKKß and cell motility controlled by Rap-integrin signaling.


Assuntos
Caseína Quinase Ialfa/metabolismo , Movimento Celular/fisiologia , Células Epiteliais/citologia , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Quinase I-kappa B/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Transdução de Sinais/fisiologia , Proteínas de Peixe-Zebra/metabolismo , Animais , Animais Geneticamente Modificados , Neoplasias da Mama , Linhagem Celular Tumoral , Feminino , Células HEK293 , Xenoenxertos , Humanos , Masculino , Fosforilação/fisiologia , Proteínas Ligases SKP Culina F-Box/metabolismo , Peixe-Zebra
19.
FASEB J ; 27(10): 4122-35, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23825225

RESUMO

Previously, exchange protein directly activated by cAMP 2 (Epac2) and PKA were known to play a role in glucose-stimulated insulin secretion (GSIS) by pancreatic ß cells. The present study shows that Epac1 mRNA is also expressed by ß cells. Therefore, we generated mice and embryonic stem (ES) cells with deletion of the Epac1 gene to define its role in ß-cell biology and metabolism. The homozygous Epac1-knockout (Epac1(-/-)) mice developed impaired glucose tolerance and GSIS with deranged islet cytoarchitecture, which was confirmed by isolated islets from adult Epac1(-/-) mice. Moreover, Epac1(-/-) mice developed more severe hyperglycemia with increased ß-cell apoptosis and insulitis after multiple low-dose streptozotocin (MLDS; 40 mg/kg) treatment than Epac1(+/+) mice. Interestingly, Epac1(-/-) mice also showed metabolic defects, including increased respiratory exchange ratio (RER) and plasma triglyceride (TG), and more severe diet-induced obesity with insulin resistance, which may contributed to ß-cell dysfunction. However, islets differentiated from Epac1(-/-) ES cells showed insulin secretion defect, reduced Glut2 and PDX-1 expression, and abolished GLP-1-stimulated PCNA induction, suggesting a role of Epac1 in ß-cell function. The current study provides in vitro and in vivo evidence that Epac1 has an important role in GSIS of ß cells and phenotype resembling metabolic syndrome.


Assuntos
Fatores de Troca do Nucleotídeo Guanina/metabolismo , Células Secretoras de Insulina/metabolismo , Síndrome Metabólica/metabolismo , Animais , Glicemia , Diabetes Mellitus Experimental , Gorduras na Dieta/efeitos adversos , Células-Tronco Embrionárias , Fatores de Troca do Nucleotídeo Guanina/genética , Camundongos , Camundongos Knockout , Obesidade/etiologia , Obesidade/genética
20.
Proc Natl Acad Sci U S A ; 110(28): 11427-32, 2013 Jul 09.
Artigo em Inglês | MEDLINE | ID: mdl-23798437

RESUMO

Rap1 is a small GTPase regulating cell-cell adhesion, cell-matrix adhesion, and actin rearrangements, all processes dynamically coordinated during cell spreading and endothelial barrier function. Here, we identify the adaptor protein ras-interacting protein 1 (Rasip1) as a Rap1-effector involved in cell spreading and endothelial barrier function. Using Förster resonance energy transfer, we show that Rasip1 interacts with active Rap1 in a cellular context. Rasip1 mediates Rap1-induced cell spreading through its interaction partner Rho GTPase-activating protein 29 (ArhGAP29), a GTPase activating protein for Rho proteins. Accordingly, the Rap1-Rasip1 complex induces cell spreading by inhibiting Rho signaling. The Rasip1-ArhGAP29 pathway also functions in Rap1-mediated regulation of endothelial junctions, which controls endothelial barrier function. In this process, Rasip1 cooperates with its close relative ras-association and dilute domain-containing protein (Radil) to inhibit Rho-mediated stress fiber formation and induces junctional tightening. These results reveal an effector pathway for Rap1 in the modulation of Rho signaling and actin dynamics, through which Rap1 modulates endothelial barrier function.


Assuntos
Endotélio Vascular/fisiologia , Proteínas Ativadoras de GTPase/fisiologia , Peptídeos e Proteínas de Sinalização Intracelular/fisiologia , Proteínas rap1 de Ligação ao GTP/fisiologia , Células Cultivadas , Endotélio Vascular/citologia , Humanos , Ligação Proteica , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA