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










Base de dados
Intervalo de ano de publicação
1.
Front Cell Dev Biol ; 8: 618898, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33604334

RESUMO

Cytoskeletal structure and its regulation are essential for maintenance of the differentiated state of specific types of cells and their adaptation to physiologic and pathophysiologic conditions. Renal glomerular capillaries, composed of podocytes, endothelial cells, and the glomerular basement membrane, have distinct structural and biophysical properties and are the site of injury in many glomerular diseases. Calcineurin inhibitors, immunosuppressant drugs used for organ transplantation and auto-immune diseases, can protect podocytes and glomerular capillaries from injury by preserving podocyte cytoskeletal structure. These drugs cause complications including hypertension and hyperkalemia which are mediated by WNK (With No Lysine) kinases as well as vasculopathy with glomerulopathy. WNK kinases and their target kinases oxidative stress-responsive kinase 1 (OSR1) and SPS1-related proline/alanine-rich kinase (SPAK) have fundamental roles in angiogenesis and are activated by calcineurin inhibitors, but the actions of these agents on kidney vasculature, and glomerular capillaries are not fully understood. We investigated WNK1 expression in cultured podocytes and isolated mouse glomerular capillaries to determine if WNK1 contributes to calcineurin inhibitor-induced preservation of podocyte and glomerular structure. WNK1 and OSR1/SPAK are expressed in podocytes, and in a pattern similar to podocyte synaptopodin in glomerular capillaries. Calcineurin inhibitors increased active OSR1/SPAK in glomerular capillaries, the Young's modulus (E) of glomeruli, and the F/G actin ratio, effects all blocked by WNK inhibition. In glomeruli, WNK inhibition caused reduced and irregular synaptopodin-staining, abnormal capillary and foot process structures, and increased deformability. In cultured podocytes, FK506 activated OSR1/SPAK, increased lamellipodia, accelerated cell migration, and promoted traction force. These actions of FK506 were reduced by depletion of WNK1. Collectively, these results demonstrate the importance of WNK1 in regulation of the podocyte actin cytoskeleton, biophysical properties of glomerular capillaries, and slit diaphragm structure, all of which are essential to normal kidney function.

3.
Am J Physiol Renal Physiol ; 311(1): F16-27, 2016 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-27009339

RESUMO

The With no Lysine [K] (WNK) family of enzymes are central in the regulation of blood pressure. WNKs have been implicated in hereditary hypertension disorders, mainly through control of the activity and levels of ion cotransporters and channels. Actions of WNKs in the kidney have been heavily investigated, and recent studies have provided insight into not only the regulation of these enzymes but also how mutations in WNKs and their interacting partners contribute to hypertensive disorders. Defining the roles of WNKs in the cardiovascular system will provide clues about additional mechanisms by which WNKs can regulate blood pressure. This review summarizes recent developments in the regulation of the WNK signaling cascade and its role in regulation of blood pressure.


Assuntos
Hipertensão/enzimologia , Hipertensão/genética , Peptídeos e Proteínas de Sinalização Intracelular/genética , Proteínas Serina-Treonina Quinases/genética , Animais , Pressão Sanguínea/genética , Pressão Sanguínea/fisiologia , Humanos , Transdução de Sinais/genética
4.
Postdoc J ; 3(6): 71-87, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-26140278

RESUMO

Phosphoinositide 3-kinases (PI3Ks) are central regulators of cellular responses to extracellular stimuli, and are involved in growth, proliferation, migration, and metabolism. The Class I PI3Ks are activated by Receptor Tyrosine Kinases (RTKs) or G Protein-Coupled Receptors (GPCRs), and their signaling is commonly deregulated in disease conditions. Among the class I PI3Ks, the p110ß isoform is unique in being activated by both RTKs and GPCRs, and its ability to bind Rho-GTPases and Rab5. Recent studies have characterized these p110ß interacting partners, defining the binding mechanisms and regulation, and thus provide insight into the function of this kinase in physiology and disease. This review summarizes the developments in p110ß research, focusing on the interacting partners and their role in p110ß-mediated signaling.

5.
Methods Mol Biol ; 1298: 271-81, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25800850

RESUMO

Isoform-specific signaling by Class IA PI 3-kinases depends in part on the interactions between distinct catalytic subunits and upstream regulatory proteins. From among the class IA catalytic subunits (p110α, p110ß, and p110δ), p110ß has unique properties. Unlike the other family members, p110ß directly binds to Gßγ subunits, downstream from activated G-protein coupled receptors, and to activated Rab5. Furthermore, the Ras-binding domain (RBD) of p110ß binds to Rac and Cdc42 but not to Ras. Defining mutations that specifically disrupt these regulatory interactions is critical for defining their role in p110ß signaling. This chapter describes the approach that was used to identify the Rab5 binding site in p110ß, and discusses methods for the analysis of p110ß-Rab5 interactions.


Assuntos
Domínio Catalítico , Fosfatidilinositol 3-Quinases/química , Fosfatidilinositol 3-Quinases/metabolismo , Mapeamento de Interação de Proteínas/métodos , Proteínas rab5 de Ligação ao GTP/metabolismo , Guanosina 5'-O-(3-Tiotrifosfato)/química , Guanosina Difosfato/química , Células HEK293 , Humanos , Proteínas Imobilizadas/química , Proteínas Imobilizadas/genética , Proteínas Imobilizadas/isolamento & purificação , Proteínas Imobilizadas/metabolismo , Fosfatidilinositol 3-Quinases/genética , Ligação Proteica , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas rab5 de Ligação ao GTP/química , Proteínas rab5 de Ligação ao GTP/genética , Proteínas rab5 de Ligação ao GTP/isolamento & purificação
6.
Proc Natl Acad Sci U S A ; 111(45): 15999-6004, 2014 Nov 11.
Artigo em Inglês | MEDLINE | ID: mdl-25362046

RESUMO

The with no lysine (K) (WNK) family of enzymes is best known for control of blood pressure through regulation of the function and membrane localization of ion cotransporters. In mice, global as well as endothelial-specific WNK1 gene disruption results in embryonic lethality due to angiogenic and cardiovascular defects. WNK1(-/-) embryos can be rescued by endothelial-specific expression of a constitutively active form of the WNK1 substrate protein kinase OSR1 (oxidative stress responsive 1). Using human umbilical vein endothelial cells (HUVECs), we explored mechanisms underlying the requirement of WNK1-OSR1 signaling for vascular development. WNK1 is required for cord formation in HUVECs, but the actions of the two major WNK1 effectors, OSR1 and its close relative SPAK (STE20/SPS1-related proline-, alanine-rich kinase), are distinct. SPAK is important for endothelial cell proliferation, whereas OSR1 is required for HUVEC chemotaxis and invasion. We also identified the zinc-finger transcription factor Slug in WNK1-mediated control of endothelial functions. Our study identifies a separation of functions for the WNK1-activated protein kinases OSR1 and SPAK in mediating proliferation, invasion, and gene expression in endothelial cells and an unanticipated link between WNK1 and Slug that is important for angiogenesis.


Assuntos
Células Endoteliais da Veia Umbilical Humana/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Animais , Proliferação de Células/fisiologia , Regulação da Expressão Gênica/fisiologia , Células Endoteliais da Veia Umbilical Humana/citologia , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Camundongos , Camundongos Knockout , Antígenos de Histocompatibilidade Menor , Neovascularização Fisiológica/fisiologia , Proteínas Serina-Treonina Quinases/genética , Transdução de Sinais/fisiologia , Fatores de Transcrição da Família Snail , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Proteína Quinase 1 Deficiente de Lisina WNK
7.
Trends Endocrinol Metab ; 25(5): 274-82, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24751357

RESUMO

Autophagy is an important catabolic cellular process that eliminates damaged and unnecessary cytoplasmic proteins and organelles. Basal autophagy occurs during normal physiological conditions, but the activity of this process can be significantly altered in human diseases. Thus, defining the regulatory inputs and signals that control autophagy is essential. Nutrients are key modulators of autophagy. Although autophagy is generally accepted to be regulated in a cell-autonomous fashion, recent studies suggest that nutrients can modulate autophagy in a systemic manner by inducing the secretion of hormones and neurotransmitters that regulate G protein-coupled receptors (GPCRs). Emerging studies show that GPCRs also regulate autophagy by directly detecting extracellular nutrients. We review the role of GPCRs in autophagy regulation, highlighting their potential as therapeutic drug targets.


Assuntos
Autofagia/fisiologia , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais/fisiologia , Animais , Humanos
8.
Proc Natl Acad Sci U S A ; 110(47): 18862-7, 2013 Nov 19.
Artigo em Inglês | MEDLINE | ID: mdl-24190998

RESUMO

Phosphoinositide 3-kinase gamma (PI3Kγ) has profound roles downstream of G-protein-coupled receptors in inflammation, cardiac function, and tumor progression. To gain insight into how the enzyme's activity is shaped by association with its p101 adaptor subunit, lipid membranes, and Gßγ heterodimers, we mapped these regulatory interactions using hydrogen-deuterium exchange mass spectrometry. We identify residues in both the p110γ and p101 subunits that contribute critical interactions with Gßγ heterodimers, leading to PI3Kγ activation. Mutating Gßγ-interaction sites of either p110γ or p101 ablates G-protein-coupled receptor-mediated signaling to p110γ/p101 in cells and severely affects chemotaxis and cell transformation induced by PI3Kγ overexpression. Hydrogen-deuterium exchange mass spectrometry shows that association with the p101 regulatory subunit causes substantial protection of the RBD-C2 linker as well as the helical domain of p110γ. Lipid interaction massively exposes that same helical site, which is then stabilized by Gßγ. Membrane-elicited conformational change of the helical domain could help prepare the enzyme for Gßγ binding. Our studies and others identify the helical domain of the class I PI3Ks as a hub for diverse regulatory interactions that include the p101, p87 (also known as p84), and p85 adaptor subunits; Rab5 and Gßγ heterodimers; and the ß-adrenergic receptor kinase.


Assuntos
Classe Ib de Fosfatidilinositol 3-Quinase/química , Classe Ib de Fosfatidilinositol 3-Quinase/metabolismo , Modelos Moleculares , Fosfatidilinositol 3-Quinases/metabolismo , Conformação Proteica , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais/fisiologia , Animais , Quimiotaxia , Classe Ib de Fosfatidilinositol 3-Quinase/genética , Medição da Troca de Deutério , Ativação Enzimática , Células HEK293 , Humanos , Espectrometria de Massas , Camundongos , Microscopia Confocal , Células NIH 3T3 , Receptores Acoplados a Proteínas G/agonistas , Transdução de Sinais/genética , Proteínas ras/metabolismo
9.
Exp Cell Res ; 319(20): 3065-80, 2013 Dec 10.
Artigo em Inglês | MEDLINE | ID: mdl-24120736

RESUMO

Connexins (Cx), gap junction (GJ) proteins, are regarded as tumor suppressors, and Cx43 expression is often down regulated in breast tumors. We assessed the effect of Cx43 over-expression in 2D and 3D cultures of two breast adenocarcinoma cell lines: MCF-7 and MDA-MB-231. While Cx43 over-expression decreased proliferation of 2D and 3D cultures of MCF-7 by 56% and 80% respectively, MDA-MB-231 growth was not altered in 2D cultures, but exhibited 35% reduction in 3D cultures. C-terminus truncated Cx43 did not alter proliferation. Untransfected MCF-7 cells formed spherical aggregates in 3D cultures, and MDA-MB-231 cells formed stellar aggregates. However, MCF-7 cells over-expressing Cx43 formed smaller sized clusters and Cx43 expressing MDA-MB-231 cells lost their stellar morphology. Extravasation ability of both MCF-7 and MDA-MB-231 cells was reduced by 60% and 30% respectively. On the other hand, silencing Cx43 in MCF10A cells, nonneoplastic human mammary cell line, increased proliferation in both 2D and 3D cultures, and disrupted acinar morphology. Although Cx43 over-expression did not affect total levels of ß-catenin, α-catenin and ZO-2, it decreased nuclear levels of ß-catenin in 2D and 3D cultures of MCF-7 cells, and in 3D cultures of MDA-MB-231 cells. Cx43 associated at the membrane with α-catenin, ß-catenin and ZO-2 in 2D and 3D cultures of MCF-7 cells, and only in 3D conditions in MDA-MB-231 cells. This study suggests that Cx43 exerts tumor suppressive effects in a context-dependent manner where GJ assembly with α-catenin, ß-catenin and ZO-2 may be implicated in reducing growth rate, invasiveness, and, malignant phenotype of 2D and 3D cultures of MCF-7 cells, and 3D cultures of MDA-MB-231 cells, by sequestering ß-catenin away from nucleus.


Assuntos
Conexina 43/genética , Conexina 43/metabolismo , Neoplasias/genética , beta Catenina/metabolismo , Perfilação da Expressão Gênica , Humanos , Células MCF-7 , Neoplasias/metabolismo , Neoplasias/patologia , Fenótipo , Ligação Proteica , Reação em Cadeia da Polimerase em Tempo Real , Células Tumorais Cultivadas , beta Catenina/genética
10.
PLoS One ; 8(5): e63833, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23734178

RESUMO

The PI3-kinase pathway is commonly activated in tumors, most often by loss of PTEN lipid phosphatase activity or the amplification or mutation of p110α. Oncogenic mutants have commonly been found in p110α, but rarely in any of the other catalytic subunits of class I PI3-kinases. We here characterize a p110ß helical domain mutation, E633K, first identified in a Her2-positive breast cancer. The mutation increases basal p110ß activity, but does not affect activation of p85/p110ß dimers by phosphopeptides or Gßγ. Expression of the mutant causes increases in Akt and S6K1 activation, transformation, chemotaxis, proliferation and survival in low serum. E633 is conserved among class I PI3 Ks, and its mutation in p110ß is also activating. Interestingly, the E633K mutant occurs near a region that interacts with membranes in activated PI 3-kinases, and its mutation abrogates the requirement for an intact Ras-binding domain in p110ß-mediated transformation. We propose that the E633K mutant activates p110ß by enhancing its basal association with membranes. This study presents the first analysis of an activating oncogenic mutation of p110ß.


Assuntos
Substituição de Aminoácidos , Neoplasias da Mama/genética , Classe Ia de Fosfatidilinositol 3-Quinase/genética , Mutação , Sequência de Aminoácidos , Animais , Western Blotting , Neoplasias da Mama/enzimologia , Membrana Celular/metabolismo , Movimento Celular/genética , Proliferação de Células , Sobrevivência Celular/genética , Transformação Celular Neoplásica/genética , Classe Ia de Fosfatidilinositol 3-Quinase/metabolismo , Ativação Enzimática/genética , Feminino , Células HEK293 , Humanos , Lipossomos/metabolismo , Camundongos , Células NIH 3T3 , Fosfotransferases/metabolismo , Ligação Proteica , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Quinases S6 Ribossômicas 70-kDa/metabolismo , Homologia de Sequência de Aminoácidos , Células Sf9
11.
Mol Cell ; 50(1): 29-42, 2013 Apr 11.
Artigo em Inglês | MEDLINE | ID: mdl-23434372

RESUMO

Autophagy is an evolutionarily conserved membrane trafficking process. Induction of autophagy in response to nutrient limitation or cellular stress occurs by similar mechanisms in organisms from yeast to mammals. Unlike yeast, metazoan cells rely more on growth factor signaling for a wide variety of cellular activities including nutrient uptake. How growth factor availability regulates autophagy is poorly understood. Here we show that, upon growth factor limitation, the p110ß catalytic subunit of the class IA phosphoinositide 3-kinases (PI3Ks) dissociates from growth factor receptor complexes and increases its interaction with the small GTPase Rab5. This p110ß-Rab5 association maintains Rab5 in its guanosine triphosphate (GTP)-bound state and enhances the Rab5-Vps34 interaction that promotes autophagy. p110ß mutants that fail to interact with Rab5 are defective in autophagy promotion. Hence, in mammalian cells, p110ß acts as a molecular sensor for growth factor availability and induces autophagy by activating a Rab5-mediated signaling cascade.


Assuntos
Autofagia , Peptídeos e Proteínas de Sinalização Intercelular/deficiência , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas rab5 de Ligação ao GTP/metabolismo , Animais , Classe I de Fosfatidilinositol 3-Quinases/deficiência , Classe I de Fosfatidilinositol 3-Quinases/genética , Classe III de Fosfatidilinositol 3-Quinases/metabolismo , Guanosina Trifosfato/metabolismo , Células HEK293 , Humanos , Camundongos , Camundongos Knockout , Mutação , Fosfatidilinositol 3-Quinases/deficiência , Fosfatidilinositol 3-Quinases/genética , Transdução de Sinais , Transfecção
12.
Trends Pharmacol Sci ; 34(3): 149-53, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23411347

RESUMO

Phosphoinositide (PI) 3-kinases are essential regulators of cellular proliferation, survival, metabolism, and motility that are frequently dysregulated in human disease. The design of inhibitors to target the PI 3-kinase/mTOR pathway is a major area of investigation by both academic laboratories and the pharmaceutical industry. This review focuses on the Class IA PI 3-kinase p110ß, which plays a unique role in thrombogenesis and in the growth of tumors with deletion or loss-of-function mutation of the Phosphatase and Tensin Homolog (PTEN) lipid phosphatase. Several p110ß-selective inhibitors that target the ATP-binding site in the kinase domain have been identified. However, recent discoveries regarding the regulatory mechanisms that control p110ß activity suggest alternative strategies by which to disrupt signaling by this PI 3-kinase isoform. This review summarizes the current status of p110ß-specific inhibitors and discusses how these new insights into p110 regulation might be used to devise novel pharmacological inhibitors.


Assuntos
Classe I de Fosfatidilinositol 3-Quinases/antagonistas & inibidores , Inibidores Enzimáticos/farmacologia , Animais , Classe I de Fosfatidilinositol 3-Quinases/química , Classe I de Fosfatidilinositol 3-Quinases/metabolismo , Desenho de Fármacos , Inibidores Enzimáticos/química , Humanos , Modelos Moleculares
13.
Sci Signal ; 5(253): ra89, 2012 Dec 04.
Artigo em Inglês | MEDLINE | ID: mdl-23211529

RESUMO

Synergistic activation by heterotrimeric guanine nucleotide-binding protein (G protein)-coupled receptors (GPCRs) and receptor tyrosine kinases distinguishes p110ß from other class IA phosphoinositide 3-kinases (PI3Ks). Activation of p110ß is specifically implicated in various physiological and pathophysiological processes, such as the growth of tumors deficient in phosphatase and tensin homolog deleted from chromosome 10 (PTEN). To determine the specific contribution of GPCR signaling to p110ß-dependent functions, we identified the site in p110ß that binds to the Gßγ subunit of G proteins. Mutation of this site eliminated Gßγ-dependent activation of PI3Kß (a dimer of p110ß and the p85 regulatory subunit) in vitro and in cells, without affecting basal activity or phosphotyrosine peptide-mediated activation. Disrupting the p110ß-Gßγ interaction by mutation or with a cell-permeable peptide inhibitor blocked the transforming capacity of PI3Kß in fibroblasts and reduced the proliferation, chemotaxis, and invasiveness of PTEN-null tumor cells in culture. Our data suggest that specifically targeting GPCR signaling to PI3Kß could provide a therapeutic approach for tumors that depend on p110ß for growth and metastasis.


Assuntos
Transformação Celular Neoplásica/metabolismo , Fibroblastos/metabolismo , Subunidades beta da Proteína de Ligação ao GTP/metabolismo , Subunidades gama da Proteína de Ligação ao GTP/metabolismo , Proteínas de Neoplasias/metabolismo , Neoplasias/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Linhagem Celular , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/patologia , Classe I de Fosfatidilinositol 3-Quinases , Fibroblastos/patologia , Subunidades beta da Proteína de Ligação ao GTP/genética , Subunidades gama da Proteína de Ligação ao GTP/genética , Humanos , Invasividade Neoplásica , Metástase Neoplásica , Proteínas de Neoplasias/genética , Neoplasias/genética , Neoplasias/patologia , Neoplasias/terapia , Fosfatidilinositol 3-Quinases/genética , Receptores Acoplados a Proteínas G/genética , Transdução de Sinais/genética
15.
Proc Natl Acad Sci U S A ; 107(46): 19897-902, 2010 Nov 16.
Artigo em Inglês | MEDLINE | ID: mdl-21030680

RESUMO

Class I PI3-kinases signal downstream of receptor tyrosine kinases and G protein-coupled receptors and have been implicated in tumorigenesis. Although the oncogenic potential of the PI3-kinase subunit p110α requires its mutational activation, other p110 isoforms can induce transformation when overexpressed in the wild-type state. In wild-type p110α, N345 in the C2 domain forms hydrogen bonds with D560 and N564 in the inter-SH2 (iSH2) domain of p85, and mutations of p110α or p85 that disrupt this interface lead to increased basal activity and transformation. Sequence analysis reveals that N345 in p110α aligns with K342 in p110ß. This difference makes wild-type p110ß analogous to a previously described oncogenic mutant, p110α-N345K. We now show that p110ß is inhibited by p85 to a lesser extent than p110α and is not differentially inhibited by wild-type p85 versus p85 mutants that disrupt the C2-iSH2 domain interface. Similar results were seen in soft agar and focus-formation assays, where p110ß was similar to p110α-N345K in transforming potential. Inhibition of p110ß by p85 was enhanced by a K342N mutation in p110ß, which led to decreased activity in vitro, decreased basal Akt and ribosomal protein S6 kinase (S6K1) activation, and decreased transformation in NIH 3T3 cells. Moreover, unlike wild-type p110ß, p110ß-K342N was differentially regulated by wild-type and mutant p85, suggesting that the inhibitory C2-iSH2 interface is functional in this mutant. This study shows that the enhanced transforming potential of p110ß is the result of its decreased inhibition by p85, due to the disruption of an inhibitory C2-iSH2 domain interface.


Assuntos
Domínio Catalítico , Transformação Celular Neoplásica/patologia , Fosfatidilinositol 3-Quinases/metabolismo , Sequência de Aminoácidos , Substituição de Aminoácidos/genética , Animais , Classe I de Fosfatidilinositol 3-Quinases , Células HEK293 , Humanos , Camundongos , Dados de Sequência Molecular , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Células NIH 3T3 , Fosfatidilinositol 3-Quinases/química , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Transdução de Sinais , Relação Estrutura-Atividade , Domínios de Homologia de src
16.
Oncotarget ; 1(8): 729-33, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21321382

RESUMO

The PI3K pathway is frequently activated in tumors, most commonly through p110α mutation or PTEN deletion. In contrast to p110α, p110ß is oncogenic when over-expressed in the wild-type state, suggesting that its regulation by p85 is different than that of p110α. In this perspective, we summarize recent data concerning the regulation of p110ß, which shows that wild-type p110ß acts like an oncogenic mutant of p110α. We also discuss the significance of this altered regulation in tumor models of PTEN deletion, as well as the potential implications of the unique p110ß regulation on GPCR-driven tumorigenesis.


Assuntos
Classe Ia de Fosfatidilinositol 3-Quinase/fisiologia , Neoplasias/genética , Neoplasias/patologia , Animais , Sobrevivência Celular/genética , Sobrevivência Celular/fisiologia , Classe Ia de Fosfatidilinositol 3-Quinase/genética , Classe Ia de Fosfatidilinositol 3-Quinase/metabolismo , Humanos , Modelos Biológicos , Neoplasias/metabolismo , PTEN Fosfo-Hidrolase/genética , PTEN Fosfo-Hidrolase/metabolismo , PTEN Fosfo-Hidrolase/fisiologia , Multimerização Proteica , Transdução de Sinais/genética , Transdução de Sinais/fisiologia
17.
Cancer Cell ; 16(6): 463-74, 2009 Dec 08.
Artigo em Inglês | MEDLINE | ID: mdl-19962665

RESUMO

Members of the mammalian phosphoinositide-3-OH kinase (PI3K) family of proteins are critical regulators of various cellular process including cell survival, growth, proliferation, and motility. Oncogenic activating mutations in the p110alpha catalytic subunit of the heterodimeric p110/p85 PI3K enzyme are frequent in human cancers. Here we show the presence of frequent mutations in p85alpha in colon cancer, a majority of which occurs in the inter-Src homology-2 (iSH2) domain. These mutations uncouple and retain p85alpha's p110-stabilizing activity, while abrogating its p110-inhibitory activity. The p85alpha mutants promote cell survival, AKT activation, anchorage-independent cell growth, and oncogenesis in a p110-dependent manner.


Assuntos
Neoplasias do Colo/patologia , Mutação , Fosfatidilinositol 3-Quinases/metabolismo , Animais , Linhagem Celular , Transformação Celular Neoplásica , Neoplasias do Colo/enzimologia , Ativação Enzimática , Humanos , Camundongos , Fosfatidilinositol 3-Quinases/química , Fosfatidilinositol 3-Quinases/genética
18.
Cell Commun Signal ; 7: 4, 2009 Mar 12.
Artigo em Inglês | MEDLINE | ID: mdl-19284610

RESUMO

Connexins constitute a large family of trans-membrane proteins that allow intercellular communication and the transfer of ions and small signaling molecules between cells. Recent studies have revealed complex translational and post-translational mechanisms that regulate connexin synthesis, maturation, membrane transport and degradation that in turn modulate gap junction intercellular communication. With the growing myriad of connexin interacting proteins, including cytoskeletal elements, junctional proteins, and enzymes, gap junctions are now perceived, not only as channels between neighboring cells, but as signaling complexes that regulate cell function and transformation. Connexins have also been shown to form functional hemichannels and have roles altogether independent of channel functions, where they exert their effects on proliferation and other aspects of life and death of the cell through mostly-undefined mechanisms. This review provides an updated overview of current knowledge of connexins and their interacting proteins, and it describes connexin modulation in disease and tumorigenesis.

19.
J Med Liban ; 56(1): 35-41, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-19534089

RESUMO

Familial Mediterranean fever (FMF) is the earliest known autoinflammatory disease, characterized by symptoms such as arthritis, peritonitis, pleuritis, erysipelas-like erythema, and most importantly amyloidosis. This disease is very common in populations of the Mediterranean area, and due to its high carrier frequency and occurrence rate in these populations, it has been the focus of much research work. Such research has allowed greater insights into the genetics of FMF, leading to the discovery of the responsible gene in 1997 and the determination of mutations and their effect on the phenotype of patients, as well as the interactions and roles of the pyrin protein, which seems to have various roles in regulation of innate immunity, inflammation, and apoptosis. Colchicine has been used as preventive treatment since 1972, and recent studies have allowed the determination of its mode of action.


Assuntos
Colchicina/uso terapêutico , Proteínas do Citoesqueleto/genética , Febre Familiar do Mediterrâneo/tratamento farmacológico , Febre Familiar do Mediterrâneo/genética , Moduladores de Tubulina/uso terapêutico , Adolescente , Adulto , Alelos , Criança , Pré-Escolar , Análise Mutacional de DNA , Éxons/genética , Febre Familiar do Mediterrâneo/diagnóstico , Genética Populacional , Genótipo , Humanos , Lactente , Líbano , Fenótipo , Pirina
20.
Open Clin Cancer J ; 1: 1-5, 2007 Nov 08.
Artigo em Inglês | MEDLINE | ID: mdl-18665243

RESUMO

Carcinoembryonic antigen and vascular endothelial growth factors are among the most important prognostic markers of colorectal cancer. Testing for these markers independently has been of limited value in screening for this tumor. The aim of this study is to determine the importance of simultaneous blood CEA and VEGF level determinations in diagnosis of colorectal cancer. Thirty-six patients diagnosed with colorectal cancer along with eight healthy controls were tested by ELISA for CEA and VEGF levels in serum and plasma, respectively. The positive predictive value of these markers was 95.4% for CEA and 89.5% for VEGF, and for combined CEA and VEGF was also high at 88%. Combined CEA and VEGF blood level assay constitutes a useful panel in detecting patients with colorectal cancer. Positive results allow selection of a subgroup of patients with a high tumor risk; therefore, such tests comprise valuable tumor diagnostic tests to add to current detection methods.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...