Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
Biol. Res ; 54: 23-23, 2021. ilus, tab
Artigo em Inglês | LILACS | ID: biblio-1505793

RESUMO

The Ras family of small Guanosine Triphosphate (GTP)-binding proteins (G proteins) represents one of the main components of intracellular signal transduction required for normal cardiac growth, but is also critically involved in the development of cardiac hypertrophy and heart failure. The present review provides an update on the role of the H-, K- and N-Ras genes and their related pathways in cardiac diseases. We focus on cardiac hypertrophy and heart failure, where Ras has been studied the most. We also review other cardiac diseases, like genetic disorders related to Ras. The scope of the review extends from fundamental concepts to therapeutic applications. Although the three Ras genes have a nearly identical primary structure, there are important functional differences between them: H-Ras mainly regulates cardiomyocyte size, whereas K-Ras regulates cardiomyocyte proliferation. N-Ras is the least studied in cardiac cells and is less associated to cardiac defects. Clinically, oncogenic H-Ras causes Costello syndrome and facio-cutaneous-skeletal syndromes with hypertrophic cardiomyopathy and arrhythmias. On the other hand, oncogenic K-Ras and alterations of other genes of the Ras-Mitogen-Activated Protein Kinase (MAPK) pathway, like Raf, cause Noonan syndrome and cardio-facio-cutaneous syndromes characterized by cardiac hypertrophy and septal defects. We further review the modulation by Ras of key signaling pathways in the cardiomyocyte, including: (i) the classical Ras-Raf-MAPK pathway, which leads to a more physiological form of cardiac hypertrophy; as well as other pathways associated with pathological cardiac hypertrophy, like (ii) The SAPK (stress activated protein kinase) pathways p38 and JNK; and (iii) The alternative pathway Raf-Calcineurin-Nuclear Factor of Activated T cells (NFAT). Genetic alterations of Ras isoforms or of genes in the Ras-MAPK pathway result in Ras-opathies, conditions frequently associated with cardiac hypertrophy or septal defects among other cardiac diseases. Several studies underline the potential role of H- and K-Ras as a hinge between physiological and pathological cardiac hypertrophy, and as potential therapeutic targets in cardiac hypertrophy and failure. Highlights - The Ras (Rat Sarcoma) gene family is a group of small G proteins - Ras is regulated by growth factors and neurohormones affecting cardiomyocyte growth and hypertrophy - Ras directly affects cardiomyocyte physiological and pathological hypertrophy - Genetic alterations of Ras and its pathways result in various cardiac phenotypes? - Ras and its pathway are differentially regulated in acquired heart disease - Ras modulation is a promising therapeutic target in various cardiac conditions.


Assuntos
Humanos , Cardiopatias Congênitas , Síndrome de Noonan , Transdução de Sinais , Cardiomegalia , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Sistema de Sinalização das MAP Quinases
2.
An. bras. dermatol ; 94(4): 458-460, July-Aug. 2019. tab, graf
Artigo em Inglês | LILACS | ID: biblio-1038300

RESUMO

Abstract: Melanoma is widely known as the most lethal skin cancer. Specific tumor-related mortality can be significantly reduced if diagnosis and treatment are properly performed during initial phases of the disease. The current search for biomarkers in early-stage melanomas is a high-priority challenge for physicians and researchers. We aimed to assess the immunoexpression of BRAFV600E and KIT in a case series consisting of 44 early-stage melanomas. Formalin-fixed paraffin-embedded samples were systematically evaluated using a semi-quantitative method based on scores of percentage and intensity for immunostained tumor cells. We observed significant concordance between BRAFV600E and KIT immunoexpression in thin invasive melanomas. Our findings corroborate previous evidence showing abnormal expression of proteins associated with MAPK intracellular signaling pathway in early-stage melanomas.


Assuntos
Humanos , Masculino , Feminino , Adulto , Pessoa de Meia-Idade , Idoso , Idoso de 80 Anos ou mais , Adulto Jovem , Neoplasias Cutâneas/patologia , Proteínas Proto-Oncogênicas c-kit/análise , Proteínas Proto-Oncogênicas B-raf/análise , Melanoma/patologia , Imuno-Histoquímica , Estudos Retrospectivos , Progressão da Doença , Proteínas Proto-Oncogênicas c-kit/genética , Proteínas Quinases Ativadas por Mitógeno/análise , Proteínas Proto-Oncogênicas B-raf/genética , Mutação , Estadiamento de Neoplasias
3.
Biol. Res ; 52: 41, 2019. tab, graf
Artigo em Inglês | LILACS | ID: biblio-1019505

RESUMO

BACKGROUND: Di-N-butyl-phthalate (DBP) is an endocrine disrupting substance. We investigated the adverse effect of DBP on testis of male rat and reveal its potential mechanism of MAPK signaling pathway involved this effect in vivo and in vitro. Gonadal hormone, sperm quality, morphological change and the activation status of JNK, ERK1/2 and p38 was determined in vivo. Primary Sertoli cell was established and cultivated with JNK, ERK1/2 inhibitors, then determine the cell viability, apoptosis and the expression of p-JNK, p-ERK1/2. Data in this study were presented as mean ± SD and determined by one-way analysis of variance (ANOVA) followed by Bonferroni's test. Difference was considered statistically significant at P < 0.05. RESULTS: In vivo experiment, DBP impaired the normal structure of testicular tissue, reduced testosterone levels in blood serum, decreased sperm count and increased sperm abnormality, p-ERK1/2 and p-JNK in rat testicular tissue increased in a dose-dependent manner. In vitro studies, DBP could decrease the viability of Sertoli cells and increase p-ERK1/2 and p-JNK. Cell apoptosis in SP600125 + DBP group was significantly lower than in DBP group (P < 0.05). p-JNK was not significantly decreased in SP600125 + DBP group, while p-ERK1/2 was significantly decreased in U0126 + DBP group. CONCLUSIONS: These results suggest that DBP can lead to testicular damage and the activation of ERK1/2 and JNK pathways, the JNK signaling pathway may be primarily associated with its effect.


Assuntos
Animais , Masculino , Ratos , Testículo/lesões , Testículo/metabolismo , Transdução de Sinais/fisiologia , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Sistema de Sinalização das MAP Quinases/fisiologia , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Dibutilftalato/farmacologia , Testículo/efeitos dos fármacos , Ratos Sprague-Dawley , Proteínas Quinases Ativadas por Mitógeno/fisiologia , Proteínas Quinases JNK Ativadas por Mitógeno/fisiologia
4.
São Paulo; s.n; s.n; 2019. 108 p. ilus, graf, tab.
Tese em Português | LILACS | ID: biblio-1008521

RESUMO

Os inibidores de BRAF (iBRAFs) e de MEK (iMEK), inauguraram uma nova classe de medicamentos, a terapia direcionada, no combate ao melanoma metastático. Entretanto, os pacientes adquirem resistência ao tratamento em poucos meses. Além disso, a imunoterapia vem ganhando espaço no tratamento do câncer, incluindo o melanoma, porém, com alguns aspectos inexplorados. Dentro deste tema, a enzima IDO vem despertando um grande interesse pela participação nos mecanismos de imunotolerância, imunoescape e progressão tumoral. A IDO é responsável pelo consumo e depleção do triptofano, produzindo a quinurenina. Ela está presente em diversos tipos celulares, incluindo células do sistema imune e células tumorais. Este trabalho objetivou avaliar a expressão de IDO durante a progressão da doença - desde do nevo até o melanoma metastático e também avaliar a regulação de IDO induzido por IFN-γ após tratamento com iBRAF em linhagens parentais e resistentes ao iBRAF, buscando-se os mecanismos moleculares. Por fim, objetivou-se entender os efeitos do 1-metil-triptofano (1-MT), um inibidor de IDO, tanto na sua capacidade de inibir a atividade de IDO quanto na sua influência na capacidade clonogênica. O estudo de bioinformática sobre o repositório público GSE12391 mostrou que o nível de expressão gênica de IDO foi superior nos estágios mais avançado da doença. Além disso, todas amostras de melanoma primário de pacientes apresentaram a imunomarcação de IDO, enquanto que nenhuma amostra de nevo apresentou tal marcação. Adicionalmente, a ocorrência de IDO se deu nos infiltrados linfoides, em células mononucleares do sistema imune. Duas análises de bioinformática de expressão gênica demonstraram que a IDO estava expressa positivamente na fase de resistência ao iBRAF. Ademais, os resultados de expressão proteica mostraram que a inibição de via MAPK (tanto por iBRAF quanto por iMEK) conseguiu modular a expressão de IDO, sendo que a maioria das linhagens apresentou uma diminuição de IDO. A atividade de IDO, medida através da produção de quinurenina, por HPLC se mostrou em consonância com os resultados de expressão proteica, exceto pela linhagem WM164 que não apresentou atividade enzimática, embora a proteína estivesse presente. Por fim, o 1-MT conseguiu inibir de maneira eficiente a enzima IDO, bloqueando a produção de quinurenina. Além de que, o 1-MT reduziu a capacidade clonogênica de maneira dose-dependente. Portanto, conclui-se que a expressão de IDO é crescente conforme a progressão do melanoma, que a inibição da via MAPK regulou a expressão de IDO e que o 1-MT reduz a capacidade clonogênica, além da sua função primária de inibir IDO


BRAF and MEK inhibitors (BRAFi and MEKi) has launched a new class of medication, the target therapy, to combat metastatic melanoma. Nevertheless, patients acquired resistance to the treatment in few months. Additionally, immunotherapy has been gaining space in cancer treatment, including melanoma, but some aspects need to be explored. Inside this theme, IDO enzyme has called the attention due to its participation in the mechanisms of immune tolerance, scape and tumor progression. IDO is responsible for tryptophan consume e depletion, producing kynurenine. It is present in different cells, including cells from immune system and tumor cells. This work purposed evaluate IDO expression during disease progression - since nevus until metastatic melanoma and also, evaluate IFN-γ-induced IDO regulation after BRAFi treatment in parental and resistant melanoma cell lines, seeking the molecular mechanisms. Lastly, it was evaluated the effects of 1-methyltryptopahn (1-MT), an IDO inhibitor, by its ability to inhibit IDO and also by its influency on the clonogenic capability. Bioinformatic study performed on GSE12391 showed that gene expression level of IDO was superior in the most advanced stages of the disease. Additionally, all sample of patient's primary melanoma presented IDO immunostaining, whereas, no nevus samples presented such staining. Besides, IDO occurrence was in the lymphoid infiltrates, in mononuclear cells from immune system. Two bioinformatic analysis of gene expression demonstrated that IDO was differentially overexpressed during BRAFi resistance stage. Moreover, protein expression results presented that MAPK pathway inhibition (both by BRAFi and by MEKy) was able to modulate IDO expression, and most of the cell lines presented an IDO downregulation. IDO activity, measured through kynurenine production, by HPLC was consonant with protein expression results, except by WM164 cell line, which did not present enzymatic activity, albeit the protein was present. By the end, 1-MT could inhibit efficiently IDO enzyme, blocking kynurenine production. Furthermore, 1-MT reduced clonogenic capability in a dosedependent manner. Therefore, it was concluded that IDO expression increases along with melanoma progression, MAPK pathway inhibition regulated IDO expression and 1-MT reduced clonogenic capability, besides its primary function of IDO inhibitor


Assuntos
Progressão da Doença , Indolamina-Pirrol 2,3,-Dioxigenase/análise , Melanoma/prevenção & controle , Biologia Computacional/instrumentação , Proteínas Quinases Ativadas por Mitógeno/análise
5.
West Indian med. j ; 67(2): 98-104, Apr.-June 2018. tab, graf
Artigo em Inglês | LILACS | ID: biblio-1045825

RESUMO

ABSTRACT Objective: To explore the application methods of mitogen-activated protein kinase signal pathway inhibitors SP600125 and SB203580 in long-term in vivo experiments. Methods: A total of 55 healthy New Zealand rabbits were randomly divided into blank control group, model control group, SP low dose group, SP high dose group, SP blank group, SB low dose group, SB high dose group, SB blank group, dimethyl sulfoxide (DMSO) control group, DMSO blank group, and positive control group. Since the first day of the experiment, each group was administered the corresponding treatment for four weeks continuously. Then, the myocardial c-Jun N-terminal kinase (JNK) and the total protein of p38, protein phosphorylation and its gene expression levels were detected. Results: After intravenous treatment with adriamycin, the myocardial phosphorylate-JNK (p-JNK) and phosphorylate-p38 (p-p38) levels in all groups were increased to varying degrees, of which the model control group increased the most significantly (p < 0.05). Compared with the model control group, the myocardial p-JNK and p-p38 increased more slowly in the SP low dose group, SP high dose group, SB low dose group, SB high dose group and positive control group (p < 0.05), of which the increase in the SP high dose group and the SB high dose group was the slowest (p < 0.05). After four weeks, the total protein and messenger ribonucleic acid of the myocardial JNK and p38 in all groups had no statistically significant difference (p > 0.05). Conclusion: The continuous intravenous injection of SP600125 and SB203580 for four weeks significantly reduced the protein phosphorylation levels of JNK and p38, which provides a practical avenue for the long-term study in vivo.


RESUMEN Objetivo: Explorar los métodos de aplicación de los inhibidores SP600125 y SB203580 de la vía de señalización de la proteína quinasa activada por mitógeno en experimentos in vivo a largo plazo. Métodos: Un total de 55 conejos sanos de Nueva Zelandia fueron divididos aleatoriamente en los grupos siguientes: grupo de control en blanco, grupo de control modelo, grupo de dosis baja SP, grupo de dosis alta SP, grupo en blanco SP, grupo de dosis baja SB, grupo de dosis alta SB, grupo en blanco SB, grupo de control dimetilsulfóxido (DMSO), grupo en blanco DMSO, y grupo de control positivo. Desde el primer día del experimento, a cada grupo se le administró el tratamiento correspondiente por cuatro semanas continuas. Entonces, se detectaron la quinasa c-Jun N-terminal (JNK) miocárdica y la proteína p38 total, así como la fosforilación proteica y sus niveles de expresión génica. Resultados: Después del tratamiento intravenoso con adriamicina, los niveles de fosfo-JNK (p-JNK) y fosfo-p38 (p-p38) del miocardio aumentaron en todos los grupos en diversos grados, siendo el aumento del grupo de control modelo el más significativo (p < 0.05). En comparación con el grupo de control modelo, p-JNK y p-p38 miocárdicos aumentaron más lentamente en el grupo de dosis baja SP, el grupo de dosis alta SP, el grupo de dosis baja SB, el grupo de dosis alta SB, y el grupo de control positivo (p < 0.05). De estos, el aumento en el grupo de dosis alta SP y el grupo de dosis alta SB fue el más lento (p < 0.05). Después de cuatro semanas, la proteína total y el ácido ribonucleico mensajero de JNK y p38 miocárdicos en todos los grupos, no tuvieron diferencias significativas (p > 0.05). Conclusión: La inyección intravenosa continua de SP600125 y SB203580 durante cuatro semanas redujo significativamente los niveles de fosforilación proteica de JNK y p38, lo que proporciona una vía práctica para el estudio a largo plazo in vivo.


Assuntos
Humanos , Masculino , Coelhos , Doxorrubicina/farmacologia , Proteínas Quinases Ativadas por Mitógeno/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Fosforilação/efeitos dos fármacos , Fatores de Tempo , Transdução de Sinais/efeitos dos fármacos , Distribuição Aleatória , Expressão Gênica
6.
Braz. j. med. biol. res ; 51(2): e6520, 2018. tab, graf
Artigo em Inglês | LILACS | ID: biblio-889032

RESUMO

Multiple growth factors can be administered to mimic the natural process of bone healing in bone tissue engineering. We investigated the effects of sequential release of bone morphogenetic protein-2 (BMP-2) and vascular endothelial growth factor (VEGF) from polylactide-poly (ethylene glycol)-polylactide (PELA) microcapsule-based scaffolds on bone regeneration. To improve the double emulsion/solvent evaporation technique, VEGF was encapsulated in PELA microcapsules, to which BMP-2 was attached. The scaffold (BMP-2/PELA/VEGF) was then fused to these microcapsules using the dichloromethane vapor method. The bioactivity of the released BMP-2 and VEGF was then quantified in rat mesenchymal stem cells (rMSCs). Immunoblotting analysis showed that BMP-2/PELA/VEG promoted the differentiation of rMSCs into osteoblasts via the MAPK and Wnt pathways. Osteoblast differentiation was assessed through alkaline phosphatase expression. When compared with simple BMP-2 plus VEGF group and pure PELA group, osteoblast differentiation in BMP-2/PELA/VEGF group significantly increased. An MTT assay indicated that BMP-2-loaded PELA scaffolds had no adverse effects on cell activity. BMP-2/PELA/VEG promoted the differentiation of rMSCs into osteoblast via the ERK1/2 and Wnt pathways. Our findings indicate that the sequential release of BMP-2 and VEGF from PELA microcapsule-based scaffolds is a promising approach for the treatment of bone defects.


Assuntos
Animais , Coelhos , Ratos , Poliésteres/farmacologia , Polietilenoglicóis/farmacologia , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Fatores de Crescimento do Endotélio Vascular/metabolismo , Tecidos Suporte , Proteína Morfogenética Óssea 2/metabolismo , Células-Tronco Mesenquimais/citologia , Fatores de Tempo , Regeneração Óssea , Transdução de Sinais/fisiologia , Células Cultivadas , Modelos Animais , Proliferação de Células , beta Catenina/fisiologia , Nanopartículas , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/metabolismo , Via de Sinalização Wnt/fisiologia
7.
Clinics ; 73(supl.1): e466s, 2018. graf
Artigo em Inglês | LILACS | ID: biblio-952823

RESUMO

Protein tyrosine phosphatases have long been considered key regulators of biological processes and are therefore implicated in the origins of various human diseases. Heterozygosity, mutations, deletions, and the complete loss of some of these enzymes have been reported to cause neurodegenerative diseases, autoimmune syndromes, genetic disorders, metabolic diseases, cancers, and many other physiological imbalances. Vaccinia H1-related phosphatase, also known as dual-specificity phosphatase 3, is a protein tyrosine phosphatase enzyme that regulates the phosphorylation of the mitogen-activated protein kinase signaling pathway, a central mediator of a diversity of biological responses. It has been suggested that vaccinia H1-related phosphatase can act as a tumor suppressor or tumor-promoting phosphatase in different cancers. Furthermore, emerging evidence suggests that this enzyme has many other biological functions, such as roles in immune responses, thrombosis, hemostasis, angiogenesis, and genomic stability, and this broad spectrum of vaccinia H1-related phosphatase activity is likely the result of its diversity of substrates. Hence, fully identifying and characterizing these substrate-phosphatase interactions will facilitate the identification of pharmacological inhibitors of vaccinia H1-related phosphatase that can be evaluated in clinical trials. In this review, we describe the biological processes mediated by vaccinia H1-related phosphatase, especially those related to genomic stability. We also focus on validated substrates and signaling circuitry with clinical relevance in human diseases, particularly oncogenesis.


Assuntos
Humanos , Fosfatase 3 de Especificidade Dupla/fisiologia , Neoplasias/enzimologia , Transdução de Sinais , Análise de Sobrevida , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Neoplasias/mortalidade
8.
Braz. j. med. biol. res ; 49(7): e5313, 2016. tab, graf
Artigo em Inglês | LILACS | ID: biblio-951690

RESUMO

Ascosphaera apis is a bee pathogen that causes bee larvae infection disease, to which treatment is not yet well investigated. The aim of this study was to investigate antifungal susceptibility in vitro against A. apis and to identify a new antifungal agent for this pathogen through minimal inhibitory concentration (MIC) assay and western blot analysis. Macelignan had 1.56 and 3.125 μg/mL MIC against A. apis after 24 and 48 h, respectively, exhibiting the strongest growth inhibition against A. apis among the tested compounds (corosolic acid, dehydrocostus lactone, loganic acid, tracheloside, fangchinoline and emodin-8-O-β-D-glucopyranoside). Furthermore, macelignan showed a narrow-ranged spectrum against various fungal strains without any mammalian cell cytotoxicity. In spite of miconazole having powerful broad-ranged anti-fungal activity including A. apis, it demonstrated strong cytotoxicity. Therefore, even if macelignan alone was effective as an antifungal agent to treat A. apis, combined treatment with miconazole was more useful to overcome toxicity, drug resistance occurrence and cost effectiveness. Finally, HOG1 was revealed as a target molecule of macelignan in the anti-A. apis activity by inhibiting phosphorylation using S. cerevisiae as a model system. Based on our results, macelignan, a food-grade antimicrobial compound, would be an effective antifungal agent against A. apis infection in bees.


Assuntos
Animais , Ascomicetos/efeitos dos fármacos , Abelhas/microbiologia , Lignanas/farmacologia , Proteínas Quinases Ativadas por Mitógeno/efeitos dos fármacos , Proteínas de Saccharomyces cerevisiae/efeitos dos fármacos , Antifúngicos/farmacologia , Sais de Tetrazólio , Fatores de Tempo , Testes de Sensibilidade Microbiana , Western Blotting , Proteínas Quinases Ativadas por Mitógeno/análise , Proteínas de Saccharomyces cerevisiae/análise , Sinergismo Farmacológico , Formazans , Larva/efeitos dos fármacos , Larva/microbiologia , Larva/patogenicidade , Micoses/tratamento farmacológico
9.
Biol. Res ; 48: 1-9, 2015. ilus, graf, tab
Artigo em Inglês | LILACS | ID: biblio-950810

RESUMO

BACKGROUND: Clinical use of chemotherapeutic drug, cisplatin is limited by its toxicity and drug resistance. Therefore, efforts continue for the discovery of novel combination therapies with cisplatin, to increase efficacy and reduce its toxicity. Here, we screened 16 medicinal plant extracts from Northeast part of India and found that leaf extract of Zanthoxylum armatum DC. (ZALE) induced cytotoxicity as well as an effect on the increasing of the efficiency of chemotherapeutic drugs (cisplatin, mitomycin C and camptothecin). This work shows detail molecular mechanism of anti-cancer activity of ZALE and its potential for combined treatment regimens to enhance the apoptotic response of chemotherapeutic drugs. RESULTS: ZALE induced cytotoxicity, nuclear blebbing and DNA fragmentation in HeLA cells suggesting apoptosis induction in human cervical cell line. However, the apoptosis induced was independent of caspase 3 activation and poly ADP ribose polymerase (PARP) cleavage. Further, ZALE activated Mitogen-activated protein kinases (MAPK) pathway as revealed by increased phosphorylation of extracellular-signal-regulated kinases (ERK), p38 and c-Jun N-ter-minal kinase (JNK). Inhibition of ERK activation but not p38 or JNK completely blocked the ZALE induced apoptosis suggesting an ERK dependent apoptosis. Moreover, ZALE generated DNA double strand breaks as suggested by the induction γH2AX foci formation. Interestingly, pretreatment of certain cancer cell lines with ZALE, sensitized the cancer cells to cisplatin and other chemotherapeutic drugs. Enhanced caspase activation was observed in the synergistic interaction among chemotherapeutic drugs and ZALE. CONCLUSION: Purification and identification of the bio-active molecules from the ZALE or as a complementary treatment for a sequential treatment of ZALE with chemotherapeutic drugs might be a new challenger to open a new therapeutic window for the novel anti-cancer treatment.


Assuntos
Humanos , Extratos Vegetais/farmacologia , Cisplatino/farmacologia , Zanthoxylum/química , Antineoplásicos Fitogênicos/farmacologia , Células HeLa , Apoptose/efeitos dos fármacos , Proteínas Quinases Ativadas por Mitógeno/efeitos dos fármacos , Proteínas Quinases JNK Ativadas por Mitógeno/efeitos dos fármacos , Ativação Enzimática/efeitos dos fármacos
10.
Biol. Res ; 48: 1-8, 2015. graf
Artigo em Inglês | LILACS | ID: biblio-950773

RESUMO

BACKGROUND: Various health benefits have been attributed to Er-Miao-San (EMS), a traditional Chinese herbal formulation that contains equal amounts of cortex phellodendri (Phellodendron amurense Ruprecht) and rhizoma atractylodis (Atractylodes lancea D.C). However, its effect on the anti-inflammatory activity in human dermal fibroblasts (HDFs) and the mechanism underlying this effect are unknown. RESULTS: This study investigated the effects of EMS on TNF-α-induced MMP-1 expression in HDFs. Our data show that EMS inhibited TNF-α-induced MMP-1 expression in a concentration-dependent manner. Interestingly, EMS maintained IkB content without inhibiting the phosphorylation of MAPKs, which are well-established upstream kinases of NF-kB. Moreover, EMS reduced the level of nuclear p65 protein in HDFs. Luciferase assay revealed that EMS inhibits the transcriptional activity of NF-kBbystabilizing IkB. Our results show that EMS exerts its anti-inflammatory effect by inhibiting NF-kB-regulated genes such as IL-1ß and IL-8. Moreover, EMS effectively inhibited TNF-α-induced expression of MMP-1 via the NF-kBpathway. CONCLUSIONS: Taken together, our data suggest that EMS could potentially be used as an anti-inflammatory and anti-aging treatment.


Assuntos
Humanos , Envelhecimento/efeitos dos fármacos , Medicamentos de Ervas Chinesas/farmacologia , Extratos Vegetais/farmacologia , Derme/citologia , Metaloproteinase 1 da Matriz/biossíntese , Fibroblastos/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Indução Enzimática/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Interleucina-8/efeitos dos fármacos , Interleucina-8/metabolismo , NF-kappa B/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Proteínas Quinases Ativadas por Mitógeno/efeitos dos fármacos , Interleucina-1beta/efeitos dos fármacos , Interleucina-1beta/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Fibroblastos/enzimologia , Anti-Inflamatórios/administração & dosagem
11.
Braz. j. microbiol ; 44(4): 1241-1250, Oct.-Dec. 2013. ilus, graf, tab
Artigo em Inglês | LILACS | ID: lil-705264

RESUMO

The mitogen-activated protein (MAP) kinase pathways has been implicated in the pathogenicity of various pathogenic fungi and plays important roles in regulating pathogenicity-related morphogenesis. This work describes the isolation and characterization of MAP kinase gene, Cgl-SLT2, from Colletotrichum gloeosporioides. A DNA sequence, including 1,633 bp of Cgl-SLT2 open-reading frame and its promoter and terminator regions, was isolated via DNA walking and cloned. To analyze gene function, a gene disruption cassette containing hygromycin-resistant gene was constructed, and Cgl-SLT2 was inactivated via gene deletion. Analysis on Cgl-slt2 mutant revealed a defect in vegetative growth and sporulation as compared to the wild-type strain. When grown under nutrient-limiting conditions, hyperbranched hyphal morphology was observed in the mutant. Conidia induction for germination on rubber wax-coated hard surfaces revealed no differences in the percentage of conidial germination between the wild-type and Cgl-slt2 mutant. However, the percentage of appressorium formation in the mutant was greatly reduced. Bipolar germination in the mutant was higher than in the wild-type at 8-h post-induction. A pathogenicity assay revealed that the mutant was unable to infect either wounded or unwounded mangoes. These results suggest that the Cgl-SLT2 MAP kinase is required for C. gloeosporioides conidiation, polarized growth, appressorium formation and pathogenicity.


Assuntos
Colletotrichum/crescimento & desenvolvimento , Colletotrichum/patogenicidade , Proteínas Fúngicas/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Esporos Fúngicos/crescimento & desenvolvimento , Clonagem Molecular , DNA Fúngico/química , DNA Fúngico/genética , Proteínas Fúngicas/genética , Deleção de Genes , Hifas/crescimento & desenvolvimento , Dados de Sequência Molecular , Mutagênese Insercional , Mangifera/microbiologia , Proteínas Quinases Ativadas por Mitógeno/genética , Fases de Leitura Aberta , Regiões Promotoras Genéticas , Doenças das Plantas/microbiologia , Análise de Sequência de DNA , Virulência
12.
J. bras. nefrol ; 35(4): 332-340, out.-dez. 2013.
Artigo em Português | LILACS | ID: lil-697093

RESUMO

As drogas nefrotóxicas são responsáveis por aproximadamente 20% dos episódios de IRA em pacientes internados e ambulatoriais. A nefrotoxicidade pela cisplatina é um dos principais fatores limitantes em até 20% dos pacientes que recebem a droga, ocasionando lesões em células do epitélio tubular renal. A toxicidade da cisplatina é determinada pelo tecido-alvo e acúmulo nas células, além da interação com diversas estruturas subcelulares e com macromoléculas. A cisplatina se acumula e interfere com o funcionamento de diferentes organelas, tais como: mitocôndrias, lisossomas, retículo endoplasmático, núcleo e membrana celular, gerando inflamação e morte celular. Esta revisão tem como objetivo definir as bases fisiopatológicas e bioquímicas da nefrotoxicidade da cisplatina, revisando os principais mecanismos moleculares que levam à toxicidade tubular da cisplatina.


The nephrotoxic drugs have been responsible for about 20% of AKI episodes in inpatients and outpatients. The cisplatin nephrotoxicity is a major limiting factors in 20% of patients who have received the drug, triggering injuries in renal tubular epithelialcells. Cisplatin toxicity is determined by the target tissue and cells accumulation besides the interaction with various subcellular structures and macromolecules. Cisplatin accumulates and interferes with the functioning of different organelles such as mitochondria, lysosomes, endoplasmic reticulum, nuclei and cell membranes, causing inflammation and cell death. This review aims to define the pathophysiology and biochemistry of the cisplatin nephrotoxicity, reviewing the main molecular mechanisms that lead to tubular cisplatin toxicity.


Assuntos
Humanos , Injúria Renal Aguda/induzido quimicamente , Antineoplásicos/efeitos adversos , Cisplatino/efeitos adversos , Antineoplásicos/metabolismo , Apoptose/efeitos dos fármacos , Cisplatino/metabolismo , Proteínas Quinases Ativadas por Mitógeno/efeitos dos fármacos , Proteínas Quinases Ativadas por Mitógeno/fisiologia , Estresse Oxidativo
13.
Braz. j. infect. dis ; 17(4): 410-417, July-Aug. 2013. ilus, tab
Artigo em Inglês | LILACS | ID: lil-683127

RESUMO

BACKGROUND: Mitogen-activated protein kinase (MAPK) signaling pathway plays an important role in response to viral infection. The aim of this study was to explore the function and mechanism of MAPK signaling pathway in enterovirus 71 (EV71) infection of human rhabdomyosarcoma (RD) cells. METHODS: Apoptosis of RD cells was observed using annexin V-FITC/PI binding assay under a fluorescence microscope. Cellular RNA was extracted and transcribed to cDNA. The expressions of 56 genes of MAPK signaling pathway in EV71-infected RD cells at 8 h and 20 h after infection were analyzed by PCR array. The levels of IL-2, IL-4, IL-10, and TNF-α in the supernatant of RD cells infected with EV71 at different time points were measured by ELISA. RESULTS: The viability of RD cells decreased obviously within 48 h after EV71 infection. Compared with the control group, EV71 infection resulted in the significantly enhanced releases of IL-2, IL-4, IL-10 and TNF-α from infected RD cells (p < 0.05). At 8 h after infection, the expressions of c-Jun, c-Fos, IFN-i, MEKK1, MLK3 and NIK genes in EV71-infected RD cells were up-regulated by 2.08-6.12-fold, whereas other 19 genes (e.g. AKT1, AKT2, E2F1, IKK and NF-κB1) exhibited down-regulation. However, at 20 h after infection, those MAPK signaling molecules including MEKK1, ASK1, MLK2, MLK3, NIK, MEK1, MEK2, MEK4, MEK7, ERK1, JNK1 and JNK2 were up-regulated. In addition, the expressions of AKT2, ELK1, c-Jun, c-Fos, NF-κB p65, PI3K and STAT1 were also increased. CONCLUSION: EV71 infection induces the differential gene expressions of MAPK signaling pathway such as ERK, JNK and PI3K/AKT in RD cells, which may be associated with the secretions of inflammatory cytokines and host cell apoptosis.


Assuntos
Humanos , Enterovirus Humano A/genética , Proteínas Quinases Ativadas por Mitógeno/genética , Rabdomiossarcoma/virologia , Citocinas/genética , Ensaio de Imunoadsorção Enzimática , Enterovirus Humano A/enzimologia , Enterovirus Humano A/fisiologia , Regulação Neoplásica da Expressão Gênica , Proteínas Quinases Ativadas por Mitógeno/fisiologia , Reação em Cadeia da Polimerase , Rabdomiossarcoma/enzimologia , Rabdomiossarcoma/genética , Transdução de Sinais/genética , Transdução de Sinais/fisiologia , Fatores de Tempo , Células Tumorais Cultivadas , Regulação para Cima , Replicação Viral
14.
J. appl. oral sci ; 20(2): 128-138, Mar.-Apr. 2012. ilus, tab
Artigo em Inglês | LILACS | ID: lil-626410

RESUMO

Recently, new treatment approaches have been developed to target the host component of periodontal disease. This review aims at providing updated information on host-modulating therapies, focusing on treatment strategies for inhibiting signal transduction pathways involved in inflammation. Pharmacological inhibitors of MAPK, NFκB and JAK/STAT pathways are being developed to manage rheumatoid arthritis, periodontal disease and other inflammatory diseases. Through these agents, inflammatory mediators can be inhibited at cell signaling level, interfering on transcription factors activation and inflammatory gene expression. Although these drugs offer great potential to modulate host response, their main limitations are lack of specificity and developments of side effects. After overcoming these limitations, adjunctive host modulating drugs will provide new therapeutic strategies for periodontal treatment.


Assuntos
Humanos , Mediadores da Inflamação/antagonistas & inibidores , Peptídeos e Proteínas de Sinalização Intracelular/uso terapêutico , Terapia de Alvo Molecular/métodos , Doenças Periodontais/terapia , Transdução de Sinais/efeitos dos fármacos , Biofilmes , Mediadores da Inflamação/imunologia , Mediadores da Inflamação/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/imunologia , Janus Quinases/imunologia , Janus Quinases/metabolismo , Proteínas Quinases Ativadas por Mitógeno/imunologia , Proteínas Quinases Ativadas por Mitógeno/metabolismo , NF-kappa B/imunologia , NF-kappa B/metabolismo , Doenças Periodontais/etiologia , Doenças Periodontais/imunologia , Fatores de Transcrição STAT/imunologia , Fatores de Transcrição STAT/metabolismo
15.
Biocell ; 35(3): 71-79, Dec. 2011. tab
Artigo em Inglês | LILACS | ID: lil-653213

RESUMO

Apigenin, a nonmutagenic flavonoid, has been shown to possess free radical scavenging activities, anticarcinogenic properties, antioxidant and anti-inflammatory effects. Recently, apigenin was reported to cause gastric relaxation in murine. To assess possible effects of apigenin on migration of bladder smooth muscle (SM) cell, we isolated SM cells from peri-cancer tissue of human bladder and established a cell model that was capable to overexpress transiently MEKK1 (MEK kinase 1). Results showed that overexpression of active human MEKK1 by adenoviruses infection induced migration of human bladder smooth muscle (hBSM) cells and phosphorylation of MAPKs, ERK, JNK and p38, which are the downstream molecules of MEKK1. Then, hBSM cell overexpressing MEKK1 were exposed to apigenin (50 microM). Our data indicated that apigenin inhibited significantly activation/phosphorylation of MAPKs and migration of hBSM cells induced by MEKK1 overexpression. Besides, apigenin inhibited actin polymerization, which underlines muscle contraction and cell migration. The results suggest that apigenin inhibits activation of MAPKs and thereby the cell migration. The mechanism might be that apigenin blocks signal transmission from MEKK1 to MAPKs.


Assuntos
Humanos , Animais , Ratos , Apigenina/farmacologia , Flavonoides , MAP Quinase Quinase Quinase 1/metabolismo , Miócitos de Músculo Liso , Miócitos de Músculo Liso/metabolismo , Movimento Celular , Bexiga Urinária , Bexiga Urinária/metabolismo , Células Cultivadas , Fosforilação , Immunoblotting , MAP Quinase Quinase Quinase 1/genética , Proteínas Quinases Ativadas por Mitógeno/metabolismo
16.
Braz. j. med. biol. res ; 43(5): 500-505, May 2010. tab, ilus
Artigo em Inglês | LILACS | ID: lil-546327

RESUMO

The regulatory function of á1B-adrenoceptors in mammalian heart homeostasis is controversial. The objective of the present study was to characterize the expression/activity of key proteins implicated in cardiac calcium handling (Na+/K+-ATPase and Ca2+-ATPases) and growth (ERK1/2, JNK1/2 and p38) in mice with cardiac-selective overexpression of constitutively active mutant á1B-adrenoceptor (CAMá1B-AR), which present a mild cardiac hypertrophy phenotype. Immunoblot assays showed that myocardial plasma membrane Ca2+-ATPase (PMCA) expression was increased by 30 percent in CAMá1B-AR mice (N = 6, P < 0.05), although there was no change in sarco/endoplasmic reticulum Ca2+-ATPase (SERCA2) expression. Moreover, total Ca2+-ATPase activity was not modified, but a significant increase in the activity of the thapsigargin-resistant (PMCA) to thapsigargin-sensitive (SERCA) ratio was detected. Neither Na+/K+-ATPase activity nor the expression of á1 and á2 subunit isoforms was changed in CAMá1B-AR mouse hearts. Moreover, immunoblot assays did not provide evidence for an enhanced activation of the three mitogen-activated protein kinases studied in this stage of hypertrophy. Therefore, these findings indicate that chronic cardiac á1B-AR activation in vivo led to mild hypertrophy devoid of significant signs of adaptive modifications concerning primary intracellular calcium control and growth-related proteins, suggesting a minor pathophysiological role of this adrenergic receptor in mouse heart at this stage of development.


Assuntos
Animais , Masculino , Camundongos , Adenosina Trifosfatases/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Miocárdio/enzimologia , Receptores Adrenérgicos alfa 1/metabolismo , Sinalização do Cálcio/fisiologia , Camundongos Transgênicos , Regulação para Cima
17.
Mem. Inst. Oswaldo Cruz ; 105(3): 269-277, May 2010. ilus, graf
Artigo em Inglês | LILACS | ID: lil-547311

RESUMO

In this paper, we provide evidence that both the mRNA and protein levels of the cyclin-dependent kinase (CDK) inhibitor p21WAF1/CDK-interacting protein 1 (Cip1) increase upon infection of A431 cells with Vaccinia virus (VACV). In addition, the VACV growth factor (VGF) seems to be required for the gene expression because infection carried out with the mutant virus VACV-VGF- revealed that this strain was unable to stimulate its transcription. Our findings are also consistent with the notion that the VGF-mediated change in p21WAF1/Cip1 expression is dependent on tyrosine kinase pathway(s) and is partially dependent on mitogen-activated protein kinase/extracellular-signal regulated kinase 1/2. We believe that these pathways are biologically significant because VACV replication and dissemination was drastically affected when the infection was carried out in the presence of the relevant pharmacological inhibitors.


Assuntos
Humanos , /metabolismo , Vírus Vaccinia/fisiologia , Linhagem Celular Tumoral , /genética , Regulação Viral da Expressão Gênica/genética , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Proteínas Tirosina Quinases/genética , Proteínas Tirosina Quinases/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Transdução de Sinais/genética , Replicação Viral/genética
18.
Braz. j. med. biol. res ; 42(1): 38-43, Jan. 2009. ilus
Artigo em Inglês | LILACS | ID: lil-505416

RESUMO

Experimental and clinical evidence suggests that angiotensin II (AII) participates in renal development. Renal AII content is several-fold higher in newborn rats and mice than in adult animals. AII receptors are also expressed in higher amounts in the kidneys of newborn rats. The kidneys of fetuses whose mother received a type 1 AII receptor (AT1) antagonist during gestation present several morphological alterations. Mutations in genes that encode components of the renin-angiotensin system are associated with autosomal recessive renal tubular dysgenesis. Morphological changes were detected in the kidneys of 3-week-old angiotensin-deficient mice. Mitogen-activated protein kinases (MAPKs) are important mediators that transduce extracellular stimuli to intracellular responses. The MAPK family comprises three major subgroups, namely extracellular signal-regulated protein kinase (ERK), c-jun N-terminal kinases (JNK), and p38 MAPK (p38). Important events in renal growth during nephrogenesis such as cellular proliferation and differentiation accompanied by apoptosis on a large scale can be mediated by MAPK pathways. A decrease in glomerulus number was observed in embryos cultured for 48 and 120 h with ERK or p38 inhibitors. Many effects of AII are mediated by MAPK pathways. Treatment with losartan during lactation provoked changes in renal function and structure associated with alterations in AT1 and type 2 AII (AT2) receptors and p-JNK and p-p38 expression in the kidney. Several studies have shown that AII and MAPKs play an important role in renal development. However, the relationship between the effects of AII and MAPK activation on renal development is still unclear.


Assuntos
Animais , Camundongos , Ratos , Rim/embriologia , Sistema de Sinalização das MAP Quinases/fisiologia , Proteínas Quinases Ativadas por Mitógeno/fisiologia , Animais Recém-Nascidos , Bloqueadores do Receptor Tipo 1 de Angiotensina II/farmacologia , Rim/efeitos dos fármacos , Rim/enzimologia , Losartan/farmacologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Proteínas Quinases Ativadas por Mitógeno/efeitos dos fármacos , Cloreto de Sódio na Dieta/efeitos adversos
19.
Arq. bras. endocrinol. metab ; 52(5): 800-808, jul. 2008. ilus, graf
Artigo em Português | LILACS | ID: lil-491847

RESUMO

A síndrome de Noonan (SN) é uma síndrome genética comum que constitui importante diagnóstico diferencial em pacientes com baixa estatura, atraso puberal ou criptorquidia. A SN apresenta grande variabilidade fenotípica e é caracterizada principalmente por dismorfismo facial, cardiopatia congênita e baixa estatura. A herança é autossômica dominante com penetrância completa. O diagnóstico é clínico, com base em critérios propostos por van der Burgt, em 1994. Recentemente, diversos genes envolvidos na via de sinalização RAS-MAPK foram identificados como causadores da SN: PTPN11, KRAS, SOS1, RAF1 e MEK1. O tratamento com hormônio de crescimento (hrGH) é proposto para corrigir a baixa estatura observada nestes pacientes. Estudos recentes apontam que pacientes com SN por mutações no gene PTPN11 apresentam pior resposta ao tratamento com hrGH quando comparado com pacientes sem mutações no PTPN11. Este artigo revisará os aspectos clínicos, moleculares e do tratamento da baixa estatura de crianças com SN com hrGH.


Noonan Syndrome (NS) is one of the most common genetic syndromes and it is an important differential diagnosis in children with short stature, delayed puberty and cryptorchidism. NS is characterized by dysmorphic facial features, congenital heart defects and short stature, but there is a great variability in phenotype. NS may occur in a pattern consistent with autosomal dominant inheritance with almost complete penetrance. The diagnosis is based on a clinical score system proposed by van der Burgt e cols. in 1994. In recent years, germline mutations in the components of RAS-MAPK (mitogen activated protein kinase) pathway have been shown to be involved in the pathogenesis of NS. Mutations in PTPN11, KRAS, SOS1, RAF1 e MEK1 can explain 60-70 percent of NS molecular cause. Growth hormone therapy is proposed to correct the short stature observed in these patients. Recent studies suggest that the presence of PTPN11 mutations in patients with NS indicates a reduced growth response to short-term hrGH treatment. In this article, it is reviewed clinical and molecular aspects of NS and hrGH treatment for short stature.


Assuntos
Humanos , Insuficiência de Crescimento/genética , Hormônio do Crescimento Humano/deficiência , Síndrome de Noonan/genética , Estatura/genética , Diagnóstico Diferencial , Insuficiência de Crescimento/diagnóstico , Insuficiência de Crescimento/tratamento farmacológico , Hormônio do Crescimento Humano/uso terapêutico , Proteínas Quinases Ativadas por Mitógeno/genética , Síndrome de Noonan/diagnóstico , Síndrome de Noonan/tratamento farmacológico , Fenótipo , /genética , Estenose da Valva Pulmonar/diagnóstico
20.
Braz. j. med. biol. res ; 41(5): 344-350, May 2008. ilus
Artigo em Inglês | LILACS | ID: lil-484442

RESUMO

T-cell acute lymphoblastic leukemia (T-ALL) is a biologically heterogeneous disease with respect to phenotype, gene expression profile and activation of particular intracellular signaling pathways. Despite very significant improvements, current therapeutic regimens still fail to cure a portion of the patients and frequently implicate the use of aggressive protocols with long-term side effects. In this review, we focused on how deregulation of critical signaling pathways, in particular Notch, PI3K/Akt, MAPK, Jak/STAT and TGF-ß, may contribute to T-ALL. Identifying the alterations that affect intracellular pathways that regulate cell cycle and apoptosis is essential to understanding the biology of this malignancy, to define more effective markers for the correct stratification of patients into appropriate therapeutic regimens and to identify novel targets for the development of specific, less detrimental therapies for T-ALL.


Assuntos
Humanos , Diferenciação Celular , Leucemia-Linfoma de Células T do Adulto , Fosfotransferases/fisiologia , Transdução de Sinais/fisiologia , Linfócitos T/citologia , /fisiologia , Janus Quinases/fisiologia , Leucemia-Linfoma de Células T do Adulto/etiologia , Leucemia-Linfoma de Células T do Adulto/fisiopatologia , Leucemia-Linfoma de Células T do Adulto/terapia , Proteínas Quinases Ativadas por Mitógeno/fisiologia , Fosforilação , Proteínas Proto-Oncogênicas c-akt/fisiologia , Receptores Notch/fisiologia , Fator de Crescimento Transformador beta/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...