Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 65
Filtrar
1.
Cell ; 161(5): 967-970, 2015 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-26000477

RESUMO

RAF links RAS, one of the most potent human oncogenes, to its effector ERK and to proliferation. This role is evolutionarily conserved, but while simpler multicellular organisms express one RAF, mammals have three. This Minireview highlights common and divergent features of RAF paralogs, their signaling outputs, and roles in tumorigenesis.


Assuntos
Carcinogênese , Sistema de Sinalização das MAP Quinases , Proteínas Proto-Oncogênicas c-raf/metabolismo , Animais , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Humanos , Neoplasias/metabolismo , Proteínas Proto-Oncogênicas c-raf/química , Proteínas Proto-Oncogênicas c-raf/genética , Transdução de Sinais
2.
PLoS Pathog ; 17(7): e1009697, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-34237114

RESUMO

Listeria monocytogenes (L. monocytogenes) is a food-borne bacterial pathogen. Innate immunity to L. monocytogenes is profoundly affected by type I interferons (IFN-I). Here we investigated host metabolism in L. monocytogenes-infected mice and its potential control by IFN-I. Accordingly, we used animals lacking either the IFN-I receptor (IFNAR) or IRF9, a subunit of ISGF3, the master regulator of IFN-I-induced genes. Transcriptomes and metabolite profiles showed that L. monocytogenes infection induces metabolic rewiring of the liver. This affects various metabolic pathways including fatty acid (FA) metabolism and oxidative phosphorylation and is partially dependent on IFN-I signaling. Livers and macrophages from Ifnar1-/- mice employ increased glutaminolysis in an IRF9-independent manner, possibly to readjust TCA metabolite levels due to reduced FA oxidation. Moreover, FA oxidation inhibition provides protection from L. monocytogenes infection, explaining part of the protection of Irf9-/- and Ifnar1-/- mice. Our findings define a role of IFN-I in metabolic regulation during L. monocytogenes infection. Metabolic differences between Irf9-/- and Ifnar1-/- mice may underlie the different susceptibility of these mice against lethal infection with L. monocytogenes.


Assuntos
Interferon Tipo I/metabolismo , Listeria monocytogenes/metabolismo , Listeriose/metabolismo , Fígado/metabolismo , Animais , Ácidos Graxos/metabolismo , Interferon Tipo I/imunologia , Listeria monocytogenes/imunologia , Listeriose/imunologia , Fígado/imunologia , Camundongos , Camundongos Endogâmicos C57BL
3.
FASEB J ; 36(9): e22478, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35916021

RESUMO

The dynamics of the actin cytoskeleton and its connection to endothelial cell-cell junctions determine the barrier function of endothelial cells. The proper regulation of barrier opening/closing is necessary for the normal function of vessels, and its dysregulation can result in chronic and acute inflammation leading to edema formation. By using atomic force microscopy, we show here that thrombin-induced permeability of human umbilical vein endothelial cells, associated with actin stress fiber formation, stiffens the cell center. The depletion of the MEK/ERK kinase BRAF reduces thrombin-induced permeability prevents stress fiber formation and cell stiffening. The peripheral actin ring becomes stabilized by phosphorylated myosin light chain, while cofilin is excluded from the cell periphery. All these changes can be reverted by the inhibition of ROCK, but not of the MEK/ERK module. We propose that the balance between the binding of cofilin and myosin to F-actin in the cell periphery, which is regulated by the activity of ROCK, determines the local dynamics of actin reorganization, ultimately driving or preventing stress fiber formation.


Assuntos
Actinas , Proteínas Proto-Oncogênicas B-raf , Citoesqueleto de Actina/metabolismo , Fatores de Despolimerização de Actina/metabolismo , Actinas/metabolismo , Células Cultivadas , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Cadeias Leves de Miosina/metabolismo , Fosforilação , Proteínas Proto-Oncogênicas B-raf/metabolismo , Trombina/metabolismo
4.
BMC Genomics ; 23(1): 817, 2022 Dec 09.
Artigo em Inglês | MEDLINE | ID: mdl-36494623

RESUMO

BACKGROUND: Quantitative proteomics has become an increasingly prominent tool in the study of life sciences. A substantial hurdle for many biologists are, however, the intricacies involved in the associated high throughput data analysis. RESULTS: In order to facilitate this task for users with limited background knowledge, we have developed amica, a freely available open-source web-based software that accepts proteomic input files from different sources. amica provides quality control, differential expression, biological network and over-representation analysis on the basis of minimal user input. Scientists can use amica's query interface interactively to compare multiple conditions and rapidly identify enriched or depleted proteins. They can visualize their results using customized output graphics, and ultimately export the results in a tab-separated format that can be shared with collaborators. The code for the application, input data and documentation can be accessed online at https://github.com/tbaccata/amica and is also incorporated in the web application. CONCLUSIONS: The strong emphasis on dynamic user interactions, the integration of various databases and the option to download processed data, facilitate the analysis of complex proteomic data for both first-time users and experienced bioinformaticians. A freely available version of amica is available at https://bioapps.maxperutzlabs.ac.at/app/amica .


Assuntos
Proteômica , Software , Proteômica/métodos , Proteínas/metabolismo , Bases de Dados Factuais , Internet
5.
Mol Cell ; 50(1): 43-55, 2013 Apr 11.
Artigo em Inglês | MEDLINE | ID: mdl-23453810

RESUMO

The Raf/MEK/ERK and PI3K/Akt pathways are prominent effectors of oncogenic Ras. These pathways negatively regulate each other, but the mechanism involved is incompletely understood. We now identify MEK1 as an essential regulator of lipid/protein phosphatase PTEN, through which it controls phosphatidylinositol-3-phosphate accumulation and AKT signaling. MEK1 ablation stabilizes AKT activation and, in vivo, causes a lupus-like autoimmune disease and myeloproliferation. Mechanistically, MEK1 is necessary for PTEN membrane recruitment as part of a ternary complex containing the multidomain adaptor MAGI1. Complex formation is independent of MEK1 kinase activity but requires phosphorylation of T292 on MEK1 by activated ERK. Thus, inhibiting the ERK pathway reduces PTEN membrane recruitment, increasing phosphatidylinositol-3-phosphate accumulation and AKT activation. Our data offer a conceptual framework for the observation that activation of the PI3K pathway frequently mediate resistance to MEK inhibitors and for the promising results obtained by combined MEK/PI3K inhibition in preclinical cancer models.


Assuntos
Membrana Celular/enzimologia , Fibroblastos/enzimologia , Linfócitos/enzimologia , MAP Quinase Quinase 1/metabolismo , PTEN Fosfo-Hidrolase/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Tolerância a Antígenos Próprios , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Doenças Autoimunes/enzimologia , Doenças Autoimunes/genética , Doenças Autoimunes/imunologia , Células COS , Moléculas de Adesão Celular , Morte Celular , Membrana Celular/efeitos dos fármacos , Membrana Celular/imunologia , Membrana Celular/patologia , Chlorocebus aethiops , Ativação Enzimática , MAP Quinases Reguladas por Sinal Extracelular/antagonistas & inibidores , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Feminino , Fibroblastos/efeitos dos fármacos , Fibroblastos/imunologia , Fibroblastos/patologia , Genótipo , Guanilato Quinases , Ativação Linfocitária , Linfócitos/imunologia , Linfócitos/patologia , MAP Quinase Quinase 1/antagonistas & inibidores , MAP Quinase Quinase 1/deficiência , MAP Quinase Quinase 1/genética , Masculino , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Complexos Multienzimáticos , Transtornos Mieloproliferativos/enzimologia , Transtornos Mieloproliferativos/genética , Transtornos Mieloproliferativos/imunologia , Fenótipo , Fosfatos de Fosfatidilinositol/metabolismo , Fosforilação , Inibidores de Proteínas Quinases/farmacologia , Transporte Proteico , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Interferência de RNA , Transdução de Sinais , Fatores de Tempo , Transfecção , Tirosina
6.
Development ; 143(2): 348-55, 2016 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-26657770

RESUMO

Extracellular phosphate plays a key role in growth plate maturation by inducing Erk1/2 (Mapk3/1) phosphorylation, leading to hypertrophic chondrocyte apoptosis. The Raf kinases induce Mek1/2 (Map2k1/2) and Erk1/2 phosphorylation; however, a role for Raf kinases in endochondral bone formation has not been identified. Ablation of both A-Raf (Araf) and B-Raf (Braf) in chondrocytes does not alter growth plate maturation. Because c-Raf (Raf1) phosphorylation is increased by extracellular phosphate and c-Raf is the predominant isoform expressed in hypertrophic chondrocytes, chondrocyte-specific c-Raf knockout mice (c-Raf(f/f);ColII-Cre(+)) were generated to define a role for c-Raf in growth plate maturation. In vivo studies demonstrated that loss of c-Raf in chondrocytes leads to expansion of the hypertrophic layer of the growth plate, with decreased phospho-Erk1/2 immunoreactivity and impaired hypertrophic chondrocyte apoptosis. However, cultured hypertrophic chondrocytes from these mice did not exhibit impairment of phosphate-induced Erk1/2 phosphorylation. Studies performed to reconcile the discrepancy between the in vitro and in vivo hypertrophic chondrocyte phenotypes revealed normal chondrocyte differentiation in c-Raf(f/f);ColII-Cre(+) mice and lack of compensatory increase in the expression of A-Raf and B-Raf. However, VEGF (Vegfa) immunoreactivity in the hypertrophic chondrocytes of c-Raf(f/f);ColII-Cre(+) mice was significantly reduced, associated with increased ubiquitylation of VEGF protein. Thus, c-Raf plays an important role in growth plate maturation by regulating vascular invasion, which is crucial for replacement of terminally differentiated hypertrophic chondrocytes by bone.


Assuntos
Lâmina de Crescimento/citologia , Lâmina de Crescimento/metabolismo , Proteínas Proto-Oncogênicas c-raf/metabolismo , Animais , Apoptose/genética , Apoptose/fisiologia , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Células Cultivadas , Condrócitos/citologia , Condrócitos/metabolismo , Feminino , Masculino , Camundongos , Osteogênese/genética , Osteogênese/fisiologia , Proteínas Proto-Oncogênicas c-raf/genética
7.
J Biol Chem ; 292(8): 3164-3171, 2017 02 24.
Artigo em Inglês | MEDLINE | ID: mdl-28073913

RESUMO

Hypophosphatemia causes rickets by impairing hypertrophic chondrocyte apoptosis. Phosphate induction of MEK1/2-ERK1/2 phosphorylation in hypertrophic chondrocytes is required for phosphate-mediated apoptosis and growth plate maturation. MEK1/2 can be activated by numerous molecules including Raf isoforms. A- and B-Raf ablation in chondrocytes does not alter skeletal development, whereas ablation of C-Raf decreases hypertrophic chondrocyte apoptosis and impairs vascularization of the growth plate. However, ablation of C-Raf does not impair phosphate-induced ERK1/2 phosphorylation in vitro, but leads to rickets by decreasing VEGF protein stability. To determine whether Raf isoforms are required for phosphate-induced hypertrophic chondrocyte apoptosis, mice lacking all three Raf isoforms in chondrocytes were generated. Raf deletion caused neonatal death and a significant expansion of the hypertrophic chondrocyte layer of the growth plate, accompanied by decreased cleaved caspase-9. This was associated with decreased phospho-ERK1/2 immunoreactivity in the hypertrophic chondrocyte layer and impaired vascular invasion. These data further demonstrated that Raf kinases are required for phosphate-induced ERK1/2 phosphorylation in cultured hypertrophic chondrocytes and perform essential, but partially redundant roles in growth plate maturation.


Assuntos
Condrócitos/metabolismo , Condrogênese , Lâmina de Crescimento/crescimento & desenvolvimento , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Proteínas Proto-Oncogênicas A-raf/metabolismo , Proteínas Proto-Oncogênicas B-raf/metabolismo , Proteínas Proto-Oncogênicas c-raf/metabolismo , Animais , Apoptose , Desenvolvimento Ósseo , Células Cultivadas , Condrócitos/citologia , Condrócitos/patologia , Lâmina de Crescimento/metabolismo , Camundongos Endogâmicos C57BL , Fosfatos/metabolismo , Fosforilação , Isoformas de Proteínas/metabolismo , Quinases raf/metabolismo
8.
Biochem Soc Trans ; 45(1): 27-36, 2017 Feb 08.
Artigo em Inglês | MEDLINE | ID: mdl-28202657

RESUMO

The RAS/ERK pathway has been intensely studied for about three decades, not least because of its role in human pathologies. ERK activation is observed in the majority of human cancers; in about one-third of them, it is driven by mutational activation of pathway components. The pathway is arguably one of the best targets for molecule-based pharmacological intervention, and several small-molecule inhibitors are in clinical use. Genetically engineered mouse models have greatly contributed to our understanding of signaling pathways in development, tissue homeostasis, and disease. In the specific case of the RAS/ERK pathway, they have revealed unique biological roles of structurally and functionally similar proteins, new kinase-independent effectors, and unsuspected relationships with other cascades. This short review summarizes the contribution of mouse models to our current understanding of the pathway.


Assuntos
MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Sistema de Sinalização das MAP Quinases , Neoplasias/metabolismo , Proteínas ras/metabolismo , Animais , Modelos Animais de Doenças , Humanos , Camundongos , Mutação , Neoplasias/classificação , Neoplasias/genética , Microambiente Tumoral/genética , Proteínas ras/genética
10.
Trends Biochem Sci ; 35(12): 660-8, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20621483

RESUMO

The three-tiered Raf-MEK-ERK kinase module is activated downstream of Ras and has been traditionally linked to cellular proliferation. Mammals have three Raf, two Mek and two Erk genes. Recently, the analysis of protein-protein interactions in the pathway has begun to provide a rationale for the redundancy within each tier. New results show that the MEK-ERK-activating unit consists of Raf hetero- and homodimers; downstream of Raf, MEK1-MEK2 heterodimers and ERK dimers are required for temporal and spatial pathway regulation. Finally, C-Raf mediates pathway crosstalk downstream of Ras by directly binding to and inhibiting kinases engaged in other signaling cascades. Given the roles of these interactions in tumorigenesis, their study will provide new opportunities for molecule-based therapies that target the pathway.


Assuntos
MAP Quinase Quinase Quinases/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Transdução de Sinais , Quinases raf/metabolismo , Animais , Dimerização , Retroalimentação , Humanos , Neoplasias/metabolismo
11.
J Cell Biol ; 223(9)2024 Sep 02.
Artigo em Inglês | MEDLINE | ID: mdl-38874393

RESUMO

Hematopoietic stem cells (HSCs) continuously replenish mature blood cells with limited lifespans. To maintain the HSC compartment while ensuring output of differentiated cells, HSCs undergo asymmetric cell division (ACD), generating two daughter cells with different fates: one will proliferate and give rise to the differentiated cells' progeny, and one will return to quiescence to maintain the HSC compartment. A balance between MEK/ERK and mTORC1 pathways is needed to ensure HSC homeostasis. Here, we show that activation of these pathways is spatially segregated in premitotic HSCs and unequally inherited during ACD. A combination of genetic and chemical perturbations shows that an ERK-dependent mechanism determines the balance between pathways affecting polarity, proliferation, and metabolism, and thus determines the frequency of asymmetrically dividing HSCs. Our data identify druggable targets that modulate HSC fate determination at the level of asymmetric division.


Assuntos
Divisão Celular Assimétrica , Células-Tronco Hematopoéticas , Animais , Células-Tronco Hematopoéticas/metabolismo , Células-Tronco Hematopoéticas/citologia , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Diferenciação Celular , Transdução de Sinais , Proliferação de Células , Linhagem da Célula , Camundongos , Sistema de Sinalização das MAP Quinases , Camundongos Endogâmicos C57BL , Polaridade Celular
12.
Int J Mol Sci ; 14(10): 19361-84, 2013 Sep 25.
Artigo em Inglês | MEDLINE | ID: mdl-24071938

RESUMO

The mammalian skin is the largest organ of the body and its outermost layer, the epidermis, undergoes dynamic lifetime renewal through the activity of somatic stem cell populations. The EGFR-Ras-Raf pathway has a well-described role in skin development and tumor formation. While research mainly focuses on its role in cutaneous tumor initiation and maintenance, much less is known about Ras signaling in the epidermal stem cells, which are the main targets of skin carcinogenesis. In this review, we briefly discuss the properties of the epidermal stem cells and review the role of EGFR-Ras-Raf signaling in keratinocyte stem cells during homeostatic and pathological conditions.


Assuntos
Epiderme/fisiologia , Receptores ErbB/genética , Folículo Piloso/fisiologia , Neoplasias Cutâneas/fisiopatologia , Células-Tronco/fisiologia , Quinases raf/genética , Proteínas ras/genética , Animais , Epiderme/metabolismo , Receptores ErbB/metabolismo , Folículo Piloso/metabolismo , Humanos , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/metabolismo , Células-Tronco/metabolismo , Quinases raf/metabolismo , Proteínas ras/metabolismo
13.
FEBS J ; 290(1): 73-75, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-35332671

RESUMO

In this special interview series, we profile members of The FEBS Journal editorial board to highlight their research focus, perspectives on the journal and future directions in their field. Manuela Baccarini is Professor of Cell Signaling at the University of Vienna, Coordinator of the International PhD Program 'Signaling Mechanisms in Cellular Homeostasis' and Director of the Vienna BioCenter PhD Program, a graduate school of the University and Medical University of Vienna in collaboration with the Institute of Molecular Pathology and the Austrian Academy of Sciences, Institute for Medical Biotechnology and Gregor Mendel Institute, as well as EMBO member and corresponding member of the Austrian Academy of Sciences. She has served as an editorial board member of The FEBS Journal since 2016.


Assuntos
Biotecnologia , Humanos
14.
Cell Rep ; 42(12): 113583, 2023 12 26.
Artigo em Inglês | MEDLINE | ID: mdl-38096057

RESUMO

Selective autophagy mediates the removal of harmful material from the cytoplasm. This cargo material is selected by cargo receptors, which orchestrate its sequestration within double-membrane autophagosomes and subsequent lysosomal degradation. The cargo receptor p62/SQSTM1 is present in cytoplasmic condensates, and a fraction of them are constantly delivered into lysosomes. However, the molecular composition of the p62 condensates is incompletely understood. To obtain insights into their composition, we develop a method to isolate these condensates and find that p62 condensates are enriched in components of the translation machinery. Furthermore, p62 interacts with translation initiation factors, and eukaryotic initiation factor 2α (eIF2α) and eIF4E are degraded by autophagy in a p62-dependent manner. Thus, p62-mediated autophagy may in part be linked to down-regulation of translation initiation. The p62 condensate isolation protocol developed here may facilitate the study of their contribution to cellular quality control and their roles in health and disease.


Assuntos
Condensados Biomoleculares , Fator de Iniciação 2 em Eucariotos , Fator de Iniciação 4E em Eucariotos , Proteínas de Ligação a RNA , Humanos , Células HEK293 , Proteínas de Ligação a RNA/metabolismo , Condensados Biomoleculares/efeitos dos fármacos , Condensados Biomoleculares/metabolismo , Fator de Iniciação 2 em Eucariotos/antagonistas & inibidores , Fator de Iniciação 2 em Eucariotos/metabolismo , Fator de Iniciação 4E em Eucariotos/antagonistas & inibidores , Fator de Iniciação 4E em Eucariotos/metabolismo , Autofagia/efeitos dos fármacos , Autofagia/genética , Wortmanina/farmacologia
15.
Autophagy ; 19(1): 152-162, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-35435804

RESUMO

Impaired degradation of the transcriptional coactivator YAP1 and IL6ST (interleukin 6 cytokine family signal transducer), two proteins deregulated in liver cancer, has been shown to promote tumor growth. Here, we demonstrate that YAP1 and IL6ST are novel substrates of chaperone-mediated autophagy (CMA) in human hepatocellular carcinoma (HCC) and hepatocyte cell lines. Knockdown of the lysosomal CMA receptor LAMP2A increases protein levels of YAP1 and IL6ST, without changes in mRNA expression. Additionally, both proteins show KFERQ-dependent binding to the CMA chaperone HSPA8 and accumulate into isolated lysosomes after stimulation of CMA by prolonged starvation. We further show that LAMP2A downregulation promotes the proliferation and migration in HCC cells and a human hepatocyte cell line, and that it does so in a YAP1- and IL6ST-dependent manner. Finally, LAMP2A expression is downregulated, and YAP1 and IL6ST expression is upregulated, in human HCC biopsies. Taken together, our work reveals a novel mechanism that controls the turnover of two cancer-relevant proteins and suggests a tumor suppressor function of CMA in the liver, advocating for the exploitation of CMA activity for diagnostic and therapeutic purposes.Abbreviations: ACTB: actin beta; ATG5: autophagy related 5; ATG7: autophagy related 7; CMA: chaperone-mediated autophagy; eMI: endosomal microautophagy; HCC: hepatocellular carcinoma; HSPA8: heat shock protein family A (Hsp70) member 8; IL6ST: interleukin 6 cytokine family signal transducer; JAK: Janus kinase; LAMP1: lysosomal associated membrane protein 1; LAMP2A: lysosomal associated membrane protein 2A; MAPK8: mitogen-activated protein kinase 8; P6: pyridine 6; SQSTM1: sequestosome 1; TUBA: tubulin alpha; VDAC1: voltage dependent anion channel 1; VP: verteporfin; YAP1: Yes1 associated transcriptional regulator.


Assuntos
Carcinoma Hepatocelular , Autofagia Mediada por Chaperonas , Neoplasias Hepáticas , Humanos , Carcinoma Hepatocelular/metabolismo , Autofagia/fisiologia , Neoplasias Hepáticas/metabolismo , Interleucina-6/metabolismo , Linhagem Celular , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proliferação de Células , Lisossomos/metabolismo , Proteína 2 de Membrana Associada ao Lisossomo/metabolismo , Receptor gp130 de Citocina/metabolismo
16.
Oncogene ; 42(20): 1649-1660, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-37020037

RESUMO

More than 30% of all human cancers are driven by RAS mutations and activating KRAS mutations are present in 40% of colorectal cancer (CRC) in the two main CRC subgroups, MSS (Microsatellite Stable) and MSI (Microsatellite Instable). Studies in RAS-driven tumors have shown essential roles of the RAS effectors RAF and specifically of RAF1, which can be dependent or independent of RAF's ability to activate the MEK/ERK module. In this study, we demonstrate that RAF1, but not its kinase activity, plays a crucial role in the proliferation of both MSI and MSS CRC cell line-derived spheroids and patient-derived organoids, and independently of KRAS mutation status. Moreover, we could define a RAF1 transcriptomic signature which includes genes that contribute to STAT3 activation, and could demonstrate that RAF1 ablation decreases STAT3 phosphorylation in all CRC spheroids tested. The genes involved in STAT3 activation as well as STAT3 targets promoting angiogenesis were also downregulated in human primary tumors expressing low levels of RAF1. These results indicate that RAF1 could be an attractive therapeutic target in both MSI and MSS CRC regardless of their KRAS status and support the development of selective RAF1 degraders rather than RAF1 inhibitors for clinical use in combination therapies.


Assuntos
Neoplasias Colorretais , Humanos , Neoplasias Colorretais/genética , Neoplasias Colorretais/patologia , Proteínas Proto-Oncogênicas p21(ras)/genética , Proteínas Proto-Oncogênicas B-raf/genética , Repetições de Microssatélites , Mutação , Instabilidade de Microssatélites , Proliferação de Células/genética , Fator de Transcrição STAT3/genética
17.
FASEB J ; 25(11): 3884-95, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21817126

RESUMO

Regulation of glucose homeostasis by insulin depends on pancreatic ß-cell growth, survival, and function. Raf-1 kinase is a major downstream target of several growth factors that promote proliferation and survival of many cell types, including the pancreatic ß cells. We have previously reported that insulin protects ß cells from apoptosis and promotes proliferation by activating Raf-1 signaling in cultured human islets, mouse islets, and MIN6 cells. As Raf-1 activity is critical for basal apoptosis and insulin secretion in vitro, we hypothesized that Raf-1 may play an important role in glucose homeostasis in vivo. To test this hypothesis, we utilized the Cre-loxP recombination system to obtain a pancreatic ß-cell-specific ablation of Raf-1 kinase gene (RIPCre(+/+):Raf-1(flox/flox)) and a complete set of littermate controls (RIPCre(+/+):Raf-1(wt/wt)). RIPCre(+/+):Raf-1(flox/flox) mice were viable, and no effects on weight gain were observed. RIPCre(+/+):Raf-1(flox/flox) mice had increased fasting blood glucose levels and impaired glucose tolerance but normal insulin tolerance compared to littermate controls. Insulin secretion in vivo and in isolated islets was markedly impaired, but there was no apparent effect on the exocytosis machinery. However, islet insulin protein and insulin 2 mRNA, but not insulin 1 mRNA, were dramatically reduced in Raf-1-knockout mice. Analysis of insulin 2 knockout mice demonstrated that this reduction in mRNA was sufficient to impair in vivo insulin secretion. Our data further indicate that Raf-1 specifically and acutely regulates insulin 2 mRNA via negative action on Foxo1, which has been shown to selectively control the insulin 2 gene. This work provides the first direct evidence that Raf-1 signaling is essential for the regulation of basal insulin transcription and the supply of releasable insulin in vivo.


Assuntos
Glicemia/metabolismo , Fatores de Transcrição Forkhead/fisiologia , Células Secretoras de Insulina/fisiologia , Insulina/metabolismo , Proteínas Proto-Oncogênicas c-raf/fisiologia , Animais , Apoptose/fisiologia , Proliferação de Células , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Feminino , Proteína Forkhead Box O1 , Homeostase , Insulina/biossíntese , Secreção de Insulina , Células Secretoras de Insulina/patologia , Masculino , Camundongos , Camundongos Knockout
19.
Nat Neurosci ; 10(5): 598-607, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17396120

RESUMO

To define the role of the Raf serine/threonine kinases in nervous system development, we conditionally targeted B-Raf and C-Raf, two of the three known mammalian Raf homologs, using a mouse line expressing Cre recombinase driven by a nestin promoter. Targeting of B-Raf, but not C-Raf, markedly attenuated baseline phosphorylation of Erk in neural tissues and led to growth retardation. Conditional elimination of B-Raf in dorsal root ganglion (DRG) neurons did not interfere with survival, but instead caused marked reduction in expression of the glial cell line-derived neurotrophic factor receptor Ret at postnatal stages, associated with a profound reduction in levels of transcription factor CBF-beta. Elimination of both alleles of Braf, which encodes B-Raf, and one allele of Raf1, which encodes C-Raf, affected DRG neuron maturation as well as proprioceptive axon projection toward the ventral horn in the spinal cord. Finally, conditional elimination of all Braf and Raf1 alleles strongly reduced neurotrophin-dependent axon growth in vitro as well as cutaneous axon terminal arborization in vivo. We conclude that Raf function is crucial for several aspects of DRG neuron development, including differentiation and axon growth.


Assuntos
Axônios/fisiologia , Diferenciação Celular/fisiologia , Neurônios Aferentes/citologia , Neurônios Aferentes/fisiologia , Transdução de Sinais/fisiologia , Quinases raf/fisiologia , Animais , Diferenciação Celular/efeitos dos fármacos , Sobrevivência Celular , Células Cultivadas , Embrião de Mamíferos , Éxons , Gânglios Espinais/citologia , Expressão Gênica/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mutação/fisiologia , Fator de Crescimento Neural/farmacologia , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Neurônios Aferentes/efeitos dos fármacos , Ratos , Transdução de Sinais/genética , Células-Tronco/efeitos dos fármacos , Transfecção , Quinases raf/genética
20.
Sci Signal ; 14(682)2021 05 11.
Artigo em Inglês | MEDLINE | ID: mdl-33975980

RESUMO

Members of the RAF family of serine-threonine kinases are intermediates in the mitogen-activated protein kinase and extracellular signal-regulated kinase (MAPK-ERK) signaling pathway, which controls key differentiation processes in B cells. By analyzing mice with B cell-specific deletion of Raf1, Braf, or both, we showed that Raf-1 and B-Raf acted together in mediating the positive selection of pre-B and transitional B cells as well as in initiating plasma cell differentiation. However, genetic or chemical inactivation of RAFs led to increased ERK phosphorylation in mature B cells. ERK activation in the absence of Raf-1 and B-Raf was mediated by multiple RAF-independent pathways, with phosphoinositide 3-kinase (PI3K) playing an important role. Furthermore, we found that ERK phosphorylation strongly increased during the transition from activated B cells to pre-plasmablasts. This increase in ERK phosphorylation did not occur in B cells lacking both Raf-1 and B-Raf, which most likely explains the partial block of plasma cell differentiation in mice lacking both RAFs. Collectively, our data indicate that B-Raf and Raf-1 are not necessary to mediate ERK phosphorylation in naïve or activated B cells but are essential for mediating the marked increase in ERK phosphorylation during the transition from activated B cells to pre-plasmablasts.


Assuntos
Linfócitos B/citologia , MAP Quinases Reguladas por Sinal Extracelular , Plasmócitos/citologia , Proteínas Proto-Oncogênicas c-raf , Animais , Diferenciação Celular , MAP Quinases Reguladas por Sinal Extracelular/genética , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Camundongos , Fosfatidilinositol 3-Quinases , Fosforilação , Proteínas Proto-Oncogênicas c-raf/genética , Proteínas Proto-Oncogênicas c-raf/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA