Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Diabetologia ; 62(7): 1275-1290, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31001672

RESUMO

AIMS/HYPOTHESIS: Previous studies have shown that diabetes mellitus destabilises the integrity of the microvasculature in different organs by damaging the interaction between pericytes and endothelial cells. In bone marrow, pericytes exert trophic functions on endothelial cells and haematopoietic cells through paracrine mechanisms. However, whether bone marrow pericytes are a target of diabetes-induced damage remains unknown. Here, we investigated whether type 2 diabetes can affect the abundance and function of bone marrow pericytes. METHODS: We conducted an observational clinical study comparing the abundance and molecular/functional characteristics of CD146+ pericytes isolated from the bone marrow of 25 individuals without diabetes and 14 individuals with uncomplicated type 2 diabetes, referring to our Musculoskeletal Research Unit for hip reconstructive surgery. RESULTS: Immunohistochemistry revealed that diabetes causes capillary rarefaction and compression of arteriole size in bone marrow, without changing CD146+ pericyte counts. These data were confirmed by flow cytometry on freshly isolated bone marrow cells. We then performed an extensive functional and molecular characterisation of immunosorted CD146+ pericytes. Type 2 diabetes caused a reduction in pericyte proliferation, viability, migration and capacity to support in vitro angiogenesis, while inducing apoptosis. AKT is a key regulator of the above functions and its phosphorylation state is reportedly reduced in the bone marrow endothelium of individuals with diabetes. Surprisingly, we could not find a difference in AKT phosphorylation (at either Ser473 or Thr308) in bone marrow pericytes from individuals with and without diabetes. Nonetheless, the angiocrine signalling reportedly associated with AKT was found to be significantly downregulated, with lower levels of fibroblast growth factor-2 (FGF2) and C-X-C motif chemokine ligand 12 (CXCL12), and activation of the angiogenesis inhibitor angiopoietin 2 (ANGPT2). Transfection with the adenoviral vector carrying the coding sequence for constitutively active myristoylated AKT rescued functional defects and angiocrine signalling in bone marrow pericytes from diabetic individuals. Furthermore, an ANGPT2 blocking antibody restored the capacity of pericytes to promote endothelial networking. CONCLUSIONS/INTERPRETATION: This is the first demonstration of pericyte dysfunction in bone marrow of people with type 2 diabetes. An altered angiocrine signalling from pericytes may participate in bone marrow microvascular remodelling in individuals with diabetes.


Assuntos
Células da Medula Óssea/patologia , Diabetes Mellitus Tipo 2/patologia , Pericitos/patologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Células Endoteliais/patologia , Feminino , Citometria de Fluxo , Humanos , Imuno-Histoquímica , Masculino , Pessoa de Meia-Idade , Transdução de Sinais/fisiologia
2.
Diabetologia ; 62(7): 1315, 2019 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-31115642

RESUMO

Unfortunately, three errors were made in the conversion of HbA1c to per cent values.

3.
Lancet Oncol ; 13(6): e249-58, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22652233

RESUMO

Since the identification of microRNAs (miRNAs) in 1993, and the subsequent discovery of their highly conserved nature in 2000, the amount of research into their function--particularly how they contribute to malignancy--has greatly increased. This class of small RNA molecules control gene expression and provide a previously unknown control mechanism for protein synthesis. As such, it is unsurprising that miRNAs are now known to play an essential part in malignancy, functioning as tumour suppressors and oncogenes. This Review summarises the present understanding of how miRNAs operate at the molecular level; how their dysregulation is a crucial part of tumour formation, maintenance, and metastasis; how they can be used as biomarkers for disease type and grade; and how miRNA-based treatments could be used for diverse types of malignancies.


Assuntos
Biomarcadores Tumorais/genética , Terapia Genética/métodos , MicroRNAs/genética , Terapia de Alvo Molecular/métodos , Neoplasias/genética , Neoplasias/terapia , Antineoplásicos/uso terapêutico , Feminino , Previsões , Regulação Neoplásica da Expressão Gênica , Genes Supressores de Tumor , Humanos , Masculino , MicroRNAs/metabolismo , Neoplasias/fisiopatologia , Medicina de Precisão/tendências , Resultado do Tratamento , Reino Unido
4.
Am J Physiol Cell Physiol ; 302(9): C1394-404, 2012 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-22322975

RESUMO

Sustained overactivation of RhoA is a common component for the pathogenesis of several cardiovascular disorders, including hypertension. Although activity of Rho proteins depends on Rho exchange factors (Rho-GEFs), the identity of Rho-GEFs expressed in vascular smooth muscle cells (VSMC) and participating in the control of Rho protein activity and Rho-dependent functions remains unknown. To address this question, we analyzed by quantitative RT-PCR the expression profile of 28 RhoA-GEFs in arteries of normotensive (saline-treated) and hypertensive (ANG II-treated) rats. Sixteen RhoA-GEFs were downregulated in mesenteric arteries of hypertensive rats, among which nine are also downregulated in cultured VSMC stimulated by ANG II (100 nM, 48 h), suggesting a direct effect of ANG II. Inhibition of type 1 ANG II receptors (losartan, 1 µM) or Rho kinase (fasudil, 10 µM) prevented ANG II-induced RhoA-GEF downregulation. Functionally, ANG II-induced downregulation of RhoA-GEFs is associated with decreased Rho kinase activation in response to endothelin-1, norepinephrine, and U-46619. This work thus identifies a group of RhoA-GEFs that controls RhoA and RhoA-dependent functions in VSMC, and a negative feedback of RhoA/Rho kinase activity on the expression of these RhoA-GEFs that may play an adaptative role to limit RhoA/Rho kinase activation.


Assuntos
Retroalimentação Fisiológica/fisiologia , Fatores de Troca do Nucleotídeo Guanina/biossíntese , Hipertensão/fisiopatologia , Músculo Liso Vascular/metabolismo , Quinases Associadas a rho/metabolismo , Angiotensina II/metabolismo , Angiotensina II/toxicidade , Animais , Artérias/metabolismo , Western Blotting , Perfilação da Expressão Gênica , Hipertensão/induzido quimicamente , Masculino , Músculo Liso Vascular/fisiopatologia , RNA Interferente Pequeno , Ratos , Ratos Endogâmicos WKY , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/fisiologia , Transfecção
5.
Biochem Soc Trans ; 38(6): 1565-70, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21118127

RESUMO

T2D (Type 2 diabetes mellitus) is a major health issue that has reached epidemic status worldwide. T2D is a progressive metabolic disorder characterized by reduced insulin sensitivity, insulin resistance and pancreatic ß-cell dysfunction. Improper treatment of TD2 can lead to severe complications such as heart disease, stroke, kidney failure, blindness and nerve damage. The aetiology and molecular mechanisms of T2D are not fully understood, but compelling evidence points to a link between T2D, obesity, dyslipidaemia and insulin resistance. Although T2D seems to be strongly linked to environmental factors such as nutrition and lifestyle, studies have shown that genetic factors, such as polymorphisms associated with metabolic genes, imprinting, fetal programming and miRNA (microRNA) expression, could also contribute to the development of this disease. miRNAs are small 22-25-nt-long untranslated RNAs that negatively regulate the translation of mRNAs. miRNAs are involved in a large number of biological functions such as development, metabolism, immunity and diseases such as cancer, cardiovascular diseases and diabetes. The present review examines the various miRNAs that have been identified as being potentially involved in T2D, focusing on the insulin-sensitive organs: white adipose tissue, liver, skeletal muscle and the insulin-producing pancreatic ß-cells.


Assuntos
Diabetes Mellitus Tipo 2/genética , MicroRNAs/genética , MicroRNAs/metabolismo , Tecido Adiposo/fisiologia , Animais , Diabetes Mellitus Tipo 2/etiologia , Regulação da Expressão Gênica , Glucose/metabolismo , Humanos , Insulina/metabolismo , Resistência à Insulina , Células Secretoras de Insulina/metabolismo , Metabolismo dos Lipídeos , Músculo Esquelético/fisiologia , Obesidade/metabolismo
6.
Diabetes Care ; 43(7): 1520-1529, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32358022

RESUMO

OBJECTIVE: In patients with type 2 diabetes (T2D) and critical limb ischemia (CLI), migration of circulating CD34+ cells predicted cardiovascular mortality at 18 months after revascularization. This study aimed to provide long-term validation and mechanistic understanding of the biomarker. RESEARCH DESIGN AND METHODS: The association between CD34+ cell migration and cardiovascular mortality was reassessed at 6 years after revascularization. In a new series of T2D-CLI and control subjects, immuno-sorted bone marrow CD34+ cells were profiled for miRNA expression and assessed for apoptosis and angiogenesis activity. The differentially regulated miRNA-21 and its proapoptotic target, PDCD4, were titrated to verify their contribution in transferring damaging signals from CD34+ cells to endothelial cells. RESULTS: Multivariable regression analysis confirmed that CD34+ cell migration forecasts long-term cardiovascular mortality. CD34+ cells from T2D-CLI patients were more apoptotic and less proangiogenic than those from control subjects and featured miRNA-21 downregulation, modulation of several long noncoding RNAs acting as miRNA-21 sponges, and upregulation of the miRNA-21 proapoptotic target PDCD4. Silencing miR-21 in control CD34+ cells phenocopied the T2D-CLI cell behavior. In coculture, T2D-CLI CD34+ cells imprinted naive endothelial cells, increasing apoptosis, reducing network formation, and modulating the TUG1 sponge/miRNA-21/PDCD4 axis. Silencing PDCD4 or scavenging reactive oxygen species protected endothelial cells from the negative influence of T2D-CLI CD34+ cells. CONCLUSIONS: Migration of CD34+ cells predicts long-term cardiovascular mortality in T2D-CLI patients. An altered paracrine signaling conveys antiangiogenic and proapoptotic features from CD34+ cells to the endothelium. This damaging interaction may increase the risk for life-threatening complications.


Assuntos
Antígenos CD34/metabolismo , Proteínas Reguladoras de Apoptose/metabolismo , Doenças Cardiovasculares/mortalidade , Diabetes Mellitus Tipo 2 , Células Endoteliais/fisiologia , Isquemia/diagnóstico , MicroRNAs/metabolismo , Proteínas de Ligação a RNA/metabolismo , Adulto , Idoso , Antígenos CD34/sangue , Proteínas Reguladoras de Apoptose/sangue , Proteínas Reguladoras de Apoptose/genética , Biomarcadores/sangue , Biomarcadores/metabolismo , Células Sanguíneas/fisiologia , Doenças Cardiovasculares/sangue , Doenças Cardiovasculares/diagnóstico , Estudos de Casos e Controles , Movimento Celular/genética , Células Cultivadas , Estado Terminal , Diabetes Mellitus Tipo 2/complicações , Diabetes Mellitus Tipo 2/diagnóstico , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/mortalidade , Angiopatias Diabéticas/diagnóstico , Angiopatias Diabéticas/metabolismo , Angiopatias Diabéticas/mortalidade , Células Endoteliais/metabolismo , Extremidades/irrigação sanguínea , Feminino , Células Endoteliais da Veia Umbilical Humana , Humanos , Isquemia/sangue , Isquemia/mortalidade , Masculino , MicroRNAs/sangue , MicroRNAs/genética , Pessoa de Meia-Idade , Valor Preditivo dos Testes , Prognóstico , Proteínas de Ligação a RNA/sangue , Proteínas de Ligação a RNA/genética , Transdução de Sinais/fisiologia
7.
Diabetes ; 67(7): 1380-1394, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29703845

RESUMO

Fat accumulates in bone marrow (BM) of patients with diabetes. In this study, we investigated the mechanisms and consequences of this phenomenon. BM mesenchymal stromal cells (BM-MSCs) from patients with type 2 diabetes (T2D) constitutively express adipogenic markers and robustly differentiate into adipocytes (ADs) upon in vitro induction as compared with BM-MSCs from subjects without diabetes. Moreover, BM-ADs from subjects with T2D (T2D BM-ADs) paracrinally stimulate a transcriptional adipogenic program in BM-MSCs. Antagonism of MCP-1, a chemokine pivotally expressed in T2D BM-ADs, prevented the T2D BM-AD secretome from converting BM-MSCs into ADs. Mechanistic validation of human data was next performed in an obese T2D mouse model. Systemic antagonism of MCP-1 improved metabolic control, reduced BM fat, and increased osteocyte density. It also indirectly re-established the abundance of long-term versus short-term hematopoietic stem cells. We reveal a diabetic feedback loop in which 1) BM-MSCs are constitutively inclined to make ADs, and 2) mature BM-ADs, via secreted MCP-1, relentlessly fuel BM-MSC determination into new fat. Pharmacological inhibition of MCP-1 signaling can contrast this vicious cycle, restoring, at least in part, the balance between adipogenesis and hematopoiesis in BM from subjects with T2D.


Assuntos
Adipócitos/metabolismo , Células da Medula Óssea/patologia , Quimiocina CCL2/metabolismo , Diabetes Mellitus Tipo 2 , Células-Tronco Hematopoéticas/patologia , Células-Tronco Mesenquimais/metabolismo , Adipócitos/efeitos dos fármacos , Adipócitos/patologia , Adiposidade/efeitos dos fármacos , Adiposidade/genética , Adulto , Idoso , Idoso de 80 Anos ou mais , Células da Medula Óssea/metabolismo , Contagem de Células , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Células Cultivadas , Quimiocina CCL2/antagonistas & inibidores , Quimiocina CCL2/genética , Quimiocina CCL2/farmacologia , Diabetes Mellitus Tipo 2/complicações , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patologia , Retroalimentação Fisiológica/fisiologia , Feminino , Hematopoese/efeitos dos fármacos , Hematopoese/genética , Células-Tronco Hematopoéticas/efeitos dos fármacos , Células-Tronco Hematopoéticas/fisiologia , Humanos , Metabolismo dos Lipídeos/efeitos dos fármacos , Metabolismo dos Lipídeos/genética , Masculino , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/patologia , Pessoa de Meia-Idade
8.
Pharmacol Ther ; 171: 30-42, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-27916653

RESUMO

Pericytes are a heterogeneous population of cells located in the blood vessel wall. They were first identified in the 19th century by Rouget, however their biological role and potential for drug targeting have taken time to be recognised. Isolation of pericytes from several different tissues has allowed a better phenotypic and functional characterization. These findings revealed a tissue-specific, multi-functional group of cells with multilineage potential. Given this emerging evidence, pericytes have acquired specific roles in pathobiological events in vascular diseases. In this review article, we will provide a compelling overview of the main diseases in which pericytes are involved, from well-established mechanisms to the latest findings. Pericyte involvement in diabetes and cancer will be discussed extensively. In the last part of the article we will review therapeutic approaches for these diseases in light of the recently acquired knowledge. To unravel pericyte-related vascular pathobiological events is pivotal not only for more tailored treatments of disease but also to establish pericytes as a therapeutic tool.


Assuntos
Isquemia/fisiopatologia , Pericitos/citologia , Doenças Vasculares/fisiopatologia , Animais , Diabetes Mellitus/fisiopatologia , Diabetes Mellitus/terapia , Humanos , Isquemia/terapia , Terapia de Alvo Molecular , Neoplasias/patologia , Neoplasias/terapia , Doenças Vasculares/terapia
9.
Endocrinology ; 157(11): 4246-4256, 2016 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-27583789

RESUMO

Obesity during pregnancy has a long-term effect on the health of the offspring including risk of developing the metabolic syndrome. Using a mouse model of maternal diet-induced obesity, we employed a genome-wide approach to investigate the microRNA (miRNA) and miRNA transcription profile in adipose tissue to understand mechanisms through which this occurs. Male offspring of diet-induced obese mothers, fed a control diet from weaning, showed no differences in body weight or adiposity at 8 weeks of age. However, offspring from the obese dams had up-regulated cytokine (Tnfα; P < .05) and chemokine (Ccl2 and Ccl7; P < .05) signaling in their adipose tissue. This was accompanied by reduced expression of miR-706, which we showed can directly regulate translation of the inflammatory proteins IL-33 (41% up-regulated; P < .05) and calcium/calmodulin-dependent protein kinase 1D (30% up-regulated; P < .01). We conclude that exposure to obesity during development primes an inflammatory environment in adipose tissue that is independent of offspring adiposity. Programming of adipose tissue miRNAs that regulate expression of inflammatory signaling molecules may be a contributing mechanism.


Assuntos
Tecido Adiposo/metabolismo , Troca Materno-Fetal/fisiologia , Obesidade/metabolismo , Obesidade/fisiopatologia , Tecido Adiposo/imunologia , Adiposidade/genética , Adiposidade/fisiologia , Animais , Peso Corporal/fisiologia , Quimiocinas/metabolismo , Citocinas/metabolismo , Feminino , Interleucina-33/metabolismo , Masculino , Fenômenos Fisiológicos da Nutrição Materna , Troca Materno-Fetal/genética , Camundongos , MicroRNAs , Gravidez
10.
Diabetes ; 65(12): 3691-3704, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27600065

RESUMO

Critical limb ischemia (CLI), foot ulcers, former amputation, and impaired regeneration are independent risk factors for limb amputation in subjects with diabetes. The present work investigates whether and by which mechanism diabetes negatively impacts on functional properties of muscular pericytes (MPs), which are resident stem cells committed to reparative angiomyogenesis. We obtained muscle biopsy samples from patients with diabetes who were undergoing major limb amputation and control subjects. Diabetic muscles collected at the rim of normal tissue surrounding the plane of dissection showed myofiber degeneration, fat deposition, and reduction of MP vascular coverage. Diabetic MPs (D-MPs) display ultrastructural alterations, a differentiation bias toward adipogenesis at the detriment of myogenesis and an inhibitory activity on angiogenesis. Furthermore, they have an imbalanced redox state, with downregulation of the antioxidant enzymes superoxide dismutase 1 and catalase, and activation of the pro-oxidant protein kinase C isoform ß-II (PKCßII)-dependent p66Shc signaling pathway. A reactive oxygen species scavenger or, even more effectively, clinically approved PKCßII inhibitors restore D-MP angiomyogenic activity. Inhibition of the PKCßII-dependent p66Shc signaling pathway could represent a novel therapeutic approach for the promotion of muscle repair in individuals with diabetes.


Assuntos
Isquemia/metabolismo , Músculo Esquelético/metabolismo , Pericitos/metabolismo , Proteína Quinase C beta/metabolismo , Idoso , Western Blotting , Proliferação de Células/efeitos dos fármacos , Ensaio de Imunoadsorção Enzimática , Feminino , Citometria de Fluxo , Células Endoteliais da Veia Umbilical Humana , Humanos , Imuno-Histoquímica , Técnicas In Vitro , Masculino , Microscopia Eletrônica de Transmissão , Músculo Esquelético/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Pericitos/efeitos dos fármacos , Ftalimidas/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/efeitos dos fármacos
11.
Mol Metab ; 3(3): 325-33, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24749062

RESUMO

We determined the effects of maternal diet-induced obesity on offspring adipose tissue insulin signalling and miRNA expression in the aetiology of insulin resistance in later life. Although body composition and glucose tolerance of 8-week-old male offspring of obese dams were not dysregulated, serum insulin was significantly (p<0.05) elevated. Key insulin signalling proteins in adipose tissue were down-regulated, including the insulin receptor, catalytic (p110ß) and regulatory (p85α) subunits of PI3K as well as AKT1 and 2 (all p<0.05). The largest reduction observed was in IRS-1 protein (p<0.001), which was regulated post-transcriptionally. Concurrently, miR-126, which targets IRS-1, was up-regulated (p<0.05). These two features were maintained in isolated primary pre-adipocytes and differentiated adipocytes in-vitro. We have therefore established that maternal diet-induced obesity programs adipose tissue insulin resistance. We hypothesise that maintenance of the phenotype in-vitro strongly suggests that this mechanism is cell autonomous and may drive insulin resistance in later life.

12.
Cardiovasc Res ; 86(1): 131-40, 2010 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-19969623

RESUMO

AIMS: Rho guanine nucleotide exchange factors (Rho GEFs) are responsible for Rho protein activation by catalyzing the exchange of GDP for GTP. Although overactivation of Rho proteins is a common component of the pathogenesis of vascular disorders, the molecular mechanisms and the Rho GEFs regulating Rho protein activity and Rho-dependent functions in vascular smooth muscle cells (VSMC) are still unknown. The aim of this study was thus to identify Rho GEFs involved in the regulation of VSMC functions. METHODS AND RESULTS: By a functional screening based on small interfering RNA (siRNA)-mediated silencing of 27 Rho GEFs, we found that only silencing of the Rho GEF Vav3 inhibited rat VSMC proliferation. Conversely, overexpression of Vav3 potentiated VSMC proliferation, whereas the catalytically inactive Vav3 mutant had no effect. The stimulatory effect of Vav3 on VSMC proliferation was inhibited by the Src tyrosine kinase inhibitor SU6656 and by co-expression of the dominant-negative Rac1-N17 mutant. In agreement with this observation, expression of Vav3 induced enrichment of Rac1 to the membrane, activation of its effector PAK, and stimulated VSMC migration. Increased levels of Vav3 transcripts were found in stented arteries and arteries from hypertensive rats. Furthermore, siRNA targeting Vav3 decreased arterial wall cell proliferation. CONCLUSION: The Rho GEF Vav3 controls VSMC proliferation and migration through activation of Rac1/PAK signalling. Vav3 is a convergent point mediating Rac1 activation in response to different upstream mediators that promote VSMC proliferation and migration and thus appears to be a new potential therapeutic target that could be used to limit vascular proliferative diseases.


Assuntos
Movimento Celular/fisiologia , Músculo Liso Vascular/citologia , Músculo Liso Vascular/fisiologia , Proteínas Proto-Oncogênicas c-vav/metabolismo , Proteínas rho de Ligação ao GTP/metabolismo , Animais , Aorta/citologia , Aorta/fisiologia , Divisão Celular/fisiologia , Células Cultivadas , Inibidores Enzimáticos/farmacologia , NG-Nitroarginina Metil Éster/farmacologia , Proteínas Proto-Oncogênicas c-vav/genética , RNA Interferente Pequeno , Ratos , Ratos Endogâmicos WKY , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Quinases Ativadas por p21/metabolismo , Proteínas rac1 de Ligação ao GTP/metabolismo , Proteínas rho de Ligação ao GTP/genética
13.
Genes Dev ; 16(4): 427-38, 2002 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-11850406

RESUMO

Mechanisms that regulate signal propagation through the ERK/MAPK pathway are still poorly understood. Several proteins are suspected to play critical roles in this process. One of these is Kinase Suppressor of Ras (KSR), a component previously identified in RAS-dependent genetic screens in Drosophila and Caenorhabditis elegans. Here, we show that KSR functions upstream of MEK within the ERK/MAPK module. In agreement with this, we found that KSR facilitates the phosphorylation of MEK by RAF. We further show that KSR associates independently with RAF and MEK, and that these interactions lead to the formation of a RAF/MEK complex, thereby positioning RAF in close proximity to its substrate MEK. These findings suggest that KSR functions as a scaffold that assembles the RAF/MEK functional pair.


Assuntos
Proteínas de Drosophila/fisiologia , Drosophila melanogaster/enzimologia , Sistema de Sinalização das MAP Quinases/fisiologia , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Proteínas Quinases/fisiologia , Proteínas Proto-Oncogênicas c-raf/fisiologia , Animais , Células Cultivadas , Proteínas de Drosophila/genética , Drosophila melanogaster/citologia , Drosophila melanogaster/genética , Substâncias Macromoleculares , Fosforilação , Proteínas Quinases/genética , Processamento de Proteína Pós-Traducional , Proteínas Recombinantes de Fusão/fisiologia , Transfecção , Proteínas ras/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA