Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 180
Filtrar
1.
Clin Transl Oncol ; 24(1): 57-65, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-34275119

RESUMO

PURPOSE: Ovarian cancer (OC) is a common malignancy, and IFN-γ, a multifunctional cytokine, is unveiled to impede the multiplication and enhance apoptosis in diverse tumor cells in previous research. Nonetheless, its function and mechanism in OC are blurred. METHODS: OC cell lines SKOV3 and OVCAR3 were dealt with different concentrations (0-40 ng/ml) of IFN-γ. CCK-8 experiment was utilized to examine cell multiplication; Flow cytometry was executed to detect apoptosis and cell cycle; Wound healing assay was utilized to detect cell migration; and Transwell experiment was implemented to examine cell invasion. qRT-PCR analysis was applied to detect STAT5, STAT3, JAK2 and JAK3 mRNA expression in OC cell lines. Western blot experiment was applied to detect the protein and phosphorylation levels of SOCS1, STAT5 and STAT3. RESULTS: IFN-γ suppressed OC cell multiplication in a concentration-dependent manner. Relative to the control group, IFN-γ restrained OC cell migration, invasion, enhanced apoptosis and prevented cell transformation from G0/G1 to S phase. Further analysis revealed that IFN-γ up-modulated SOCS1 expression and impeded STAT5 and STAT3 protein phosphorylation levels, and knockdown of SOCS1 partially counteracted the inhibitory effect of IFN-γ on STAT5 and STAT3 protein phosphorylation levels. CONCLUSION: IFN-γ represses OC progression by facilitating SOCS1 to suppress STAT3 and STAT5 protein phosphorylation.


Assuntos
Interferon gama/fisiologia , Janus Quinase 2/fisiologia , Neoplasias Ovarianas/patologia , Fator de Transcrição STAT5/fisiologia , Transdução de Sinais , Proteína 1 Supressora da Sinalização de Citocina/fisiologia , Progressão da Doença , Feminino , Humanos , Fator de Transcrição STAT3
2.
J BUON ; 26(4): 1635-1641, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34565029

RESUMO

PURPOSE: The purpose of this study was to analyze the function of curcumin to suppress the proliferative and invasive abilities of papillary thyroid carcinoma (PTC) through inhibiting the JAK2/STAT3 pathway. METHODS: After treatment of different doses of curcumin in TPC-1 and SW1736 cells, changes in viability, clonality, cell cycle, apoptosis, wound healing and invasion were determined. Western blot analyses were performed to detect protein levels of apoptosis-associated genes, JAK2 and STAT3 in TPC-1 and SW1736 cells treated with different doses of curcumin. RESULTS: Curcumin treatment dose-dependently reduced viability, clonality and metastatic ability in TPC-1 and SW1736 cells. After treatment of 10 µM or 20 µM curcumin, PTC cells were blocked in G2/M phase, and their apoptotic rate increased. Curcumin treatment downregulated Bcl-2 and upregulated Bax in PTC cells. In addition, curcumin treatment downregulated p-JAK2 and p-STAT3 in TPC-1 and SW1736 cells. CONCLUSIONS: Curcumin treatment blocks PTC cells to proliferate and invade via inhibiting the JAK2/STAT3 pathway.


Assuntos
Curcumina/farmacologia , Curcumina/uso terapêutico , Janus Quinase 2/efeitos dos fármacos , Janus Quinase 2/fisiologia , Fator de Transcrição STAT3/efeitos dos fármacos , Fator de Transcrição STAT3/fisiologia , Câncer Papilífero da Tireoide/tratamento farmacológico , Câncer Papilífero da Tireoide/patologia , Neoplasias da Glândula Tireoide/tratamento farmacológico , Neoplasias da Glândula Tireoide/patologia , Proliferação de Células/efeitos dos fármacos , Humanos , Invasividade Neoplásica , Células Tumorais Cultivadas
3.
Leukemia ; 35(10): 2875-2884, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34480104

RESUMO

Myeloproliferative neoplasms (MPN) show dysregulated JAK2 signaling. JAK2 inhibitors provide clinical benefits, but compensatory activation of MAPK pathway signaling impedes efficacy. We hypothesized that dual targeting of JAK2 and ERK1/2 could enhance clone control and therapeutic efficacy. We employed genetic and pharmacologic targeting of ERK1/2 in Jak2V617F MPN mice, cells and patient clinical isolates. Competitive transplantations of Jak2V617F vs. wild-type bone marrow (BM) showed that ERK1/2 deficiency in hematopoiesis mitigated MPN features and reduced the Jak2V617F clone in blood and hematopoietic progenitor compartments. ERK1/2 ablation combined with JAK2 inhibition suppressed MAPK transcriptional programs, normalized cytoses and promoted clone control suggesting dual JAK2/ERK1/2 targeting as enhanced corrective approach. Combined pharmacologic JAK2/ERK1/2 inhibition with ruxolitinib and ERK inhibitors reduced proliferation of Jak2V617F cells and corrected erythrocytosis and splenomegaly of Jak2V617F MPN mice. Longer-term treatment was able to induce clone reductions. BM fibrosis was significantly decreased in MPLW515L-driven MPN to an extent not seen with JAK2 inhibitor monotherapy. Colony formation from JAK2V617F patients' CD34+ blood and BM was dose-dependently inhibited by combined JAK2/ERK1/2 inhibition in PV, ET, and MF subsets. Overall, we observed that dual targeting of JAK2 and ERK1/2 was able to enhance therapeutic efficacy suggesting a novel treatment approach for MPN.


Assuntos
Regulação Leucêmica da Expressão Gênica/efeitos dos fármacos , Janus Quinase 2/antagonistas & inibidores , Proteína Quinase 1 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 3 Ativada por Mitógeno/antagonistas & inibidores , Transtornos Mieloproliferativos/tratamento farmacológico , Nitrilas/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Pirazóis/farmacologia , Pirimidinas/farmacologia , Animais , Proliferação de Células , Feminino , Humanos , Janus Quinase 2/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína Quinase 1 Ativada por Mitógeno/fisiologia , Proteína Quinase 3 Ativada por Mitógeno/fisiologia , Transtornos Mieloproliferativos/metabolismo , Transtornos Mieloproliferativos/patologia
4.
Osteoarthritis Cartilage ; 29(10): 1389-1398, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34284112

RESUMO

Osteoarthritis (OA) is a major health problem worldwide that affects the joints and causes severe disability. It is characterized by pain and low-grade inflammation. However, the exact pathogenesis remains unknown and the therapeutic options are limited. In OA articular chondrocytes undergo a phenotypic transition becoming hypertrophic, which leads to cartilage damage, aggravating the disease. Therefore, a therapeutic agent inhibiting hypertrophy would be a promising disease-modifying drug. The therapeutic use of tyrosine kinase inhibitors has been mainly focused on oncology, but the Food and Drug Administration (FDA) approval of the Janus kinase inhibitor Tofacitinib in Rheumatoid Arthritis has broadened the applicability of these compounds to other diseases. Interestingly, tyrosine kinases have been associated with chondrocyte hypertrophy. In this review, we discuss the experimental evidence that implicates specific tyrosine kinases in signaling pathways promoting chondrocyte hypertrophy, highlighting their potential as therapeutic targets for OA.


Assuntos
Condrócitos/patologia , Osteoartrite/tratamento farmacológico , Inibidores de Proteínas Quinases/farmacologia , Receptores com Domínio Discoidina/fisiologia , Receptores ErbB/fisiologia , Proteína-Tirosina Quinases de Adesão Focal/fisiologia , Humanos , Hipertrofia/tratamento farmacológico , Janus Quinase 2/fisiologia , Osteoartrite/fisiopatologia , Proteínas Tirosina Quinases/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-fyn/fisiologia , Receptores Órfãos Semelhantes a Receptor Tirosina Quinase/fisiologia , Receptor IGF Tipo 1/fisiologia , Receptor trkA/fisiologia , Receptores de Fatores de Crescimento de Fibroblastos/fisiologia , Transdução de Sinais
5.
J Zhejiang Univ Sci B ; 22(6): 492-503, 2021 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-34128372

RESUMO

Dysregulated crosstalk between different signaling pathways contributes to tumor development, including resistance to cancer therapy. In the present study, we found that the mitogen-activated extracellular signal-regulated kinase (MEK) inhibitor trametinib failed to suppress the proliferation of PANC-1 and MGC803 cells by activating the Janus kinase 2 (JAK2)/signal transducer and activator of transcription 3 (STAT3) signaling pathway, while the JAK2 inhibitor fedratinib failed to inhibit the growth of the PANC-1 cells upon stimulation of extracellular signal-regulated kinase (ERK) signaling. In particular, the most prominent enhancement of the anti-proliferative effect resulted from the concurrent blockage of the JAK2/STAT3 and ERK signaling pathways. Furthermore, the combination of the two inhibitors resulted in a reduced tumor burden in mice. Our evidence suggests novel crosstalk between JAK2/STAT3 and ERK signaling in gastric cancer (GC) and pancreatic ductal adenocarcinoma (PDAC) cells and provides a therapeutic strategy to overcome potential resistance in gastrointestinal cancer.


Assuntos
Apoptose/efeitos dos fármacos , MAP Quinases Reguladas por Sinal Extracelular/antagonistas & inibidores , Neoplasias Gastrointestinais/tratamento farmacológico , Janus Quinase 2/antagonistas & inibidores , Inibidores de Proteínas Quinases/uso terapêutico , Fator de Transcrição STAT3/antagonistas & inibidores , Animais , Linhagem Celular Tumoral , Proliferação de Células , MAP Quinases Reguladas por Sinal Extracelular/fisiologia , Feminino , Neoplasias Gastrointestinais/patologia , Humanos , Janus Quinase 2/fisiologia , Camundongos , Camundongos Endogâmicos BALB C , Piridonas/uso terapêutico , Pirimidinonas/uso terapêutico , Pirrolidinas/uso terapêutico , Fator de Transcrição STAT3/fisiologia , Transdução de Sinais/efeitos dos fármacos , Sulfonamidas/uso terapêutico , Microambiente Tumoral
6.
Front Immunol ; 12: 762989, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34975854

RESUMO

Despite recent improvements in treatment modalities, pancreatic cancer remains a highly lethal tumor with mortality rate increasing every year. Poly (ADP-ribose) polymerase (PARP) inhibitors are now used in pancreatic cancer as a breakthrough in targeted therapy. This study focused on whether PARP inhibitors (PARPis) can affect programmed death ligand-1 (PD-L1) expression in pancreatic cancer and whether immune checkpoint inhibitors of PD-L1/programmed death 1 (PD-1) can enhance the anti-tumor effects of PARPis. Here we found that PARPi, pamiparib, up-regulated PD-L1 expression on the surface of pancreatic cancer cells in vitro and in vivo. Mechanistically, pamiparib induced PD-L1 expression via JAK2/STAT3 pathway, at least partially, in pancreatic cancer. Importantly, pamiparib attenuated tumor growth; while co-administration of pamiparib with PD-L1 blockers significantly improved the therapeutic efficacy in vivo compared with monotherapy. Combination therapy resulted in an altered tumor immune microenvironment with a significant increase in windiness of CD8+ T cells, suggesting a potential role of CD8+ T cells in the combination therapy. Together, this study provides evidence for the clinical application of PARPis with anti-PD-L1/PD-1 drugs in the treatment of pancreatic cancer.


Assuntos
Antígeno B7-H1/genética , Inibidores de Checkpoint Imunológico/administração & dosagem , Neoplasias Pancreáticas/tratamento farmacológico , Inibidores de Poli(ADP-Ribose) Polimerases/farmacologia , Animais , Linfócitos T CD8-Positivos/imunologia , Linhagem Celular Tumoral , Quimioterapia Combinada , Feminino , Fluorenos/farmacologia , Humanos , Janus Quinase 2/fisiologia , Linfócitos do Interstício Tumoral/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Neoplasias Pancreáticas/imunologia , Neoplasias Pancreáticas/patologia , Inibidores de Poli(ADP-Ribose) Polimerases/administração & dosagem , Fator de Transcrição STAT3/fisiologia , Microambiente Tumoral , Regulação para Cima
7.
Oncogene ; 40(4): 746-762, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33247204

RESUMO

Leukemias are routinely sub-typed for risk/outcome prediction and therapy choice using acquired mutations and chromosomal rearrangements. Down syndrome acute lymphoblastic leukemia (DS-ALL) is characterized by high frequency of CRLF2-rearrangements, JAK2-mutations, or RAS-pathway mutations. Intriguingly, JAK2 and RAS-mutations are mutually exclusive in leukemic sub-clones, causing dichotomy in therapeutic target choices. We prove in a cell model that elevated CRLF2 in combination with constitutionally active JAK2 is sufficient to activate wtRAS. On primary clinical DS-ALL samples, we show that wtRAS-activation is an obligatory consequence of mutated/hyperphosphorylated JAK2. We further prove that CRLF2-ligand TSLP boosts the direct binding of active PTPN11 to wtRAS, providing the molecular mechanism for the wtRAS activation. Pre-inhibition of RAS or PTPN11, but not of PI3K or JAK-signaling, prevented TSLP-induced RAS-GTP boost. Cytotoxicity assays on primary clinical DS-ALL samples demonstrated that, regardless of mutation status, high-risk leukemic cells could only be killed using RAS-inhibitor or PTPN11-inhibitor, but not PI3K/JAK-inhibitors, suggesting a unified treatment target for up to 80% of DS-ALL. Importantly, protein activities-based principal-component-analysis multivariate clusters analyzed for independent outcome prediction using Cox proportional-hazards model showed that protein-activity (but not mutation-status) was independently predictive of outcome, demanding a paradigm-shift in patient-stratification strategy for precision therapy in high-risk ALL.


Assuntos
Mutação , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Proteínas ras/fisiologia , Animais , Citocinas/fisiologia , Humanos , Janus Quinase 2/genética , Janus Quinase 2/fisiologia , Camundongos , Fosfatidilinositol 3-Quinases/fisiologia , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamento farmacológico , Proteína Tirosina Fosfatase não Receptora Tipo 11/fisiologia , Receptores de Citocinas/genética , Transdução de Sinais/fisiologia , Serina-Treonina Quinases TOR/fisiologia , Proteínas ras/antagonistas & inibidores , Proteínas ras/genética
8.
Int J Mol Sci ; 21(21)2020 Nov 04.
Artigo em Inglês | MEDLINE | ID: mdl-33158194

RESUMO

Muscle cachexia is one of the most critical unmet medical needs. Identifying the molecular background of cancer-induced muscle loss revealed a promising possibility of new therapeutic targets and new drug development. In this review, we will define the signal transducer and activator of transcription 3 (STAT3) protein's role in the tumor formation process and summarize the role of STAT3 in skeletal muscle cachexia. Finally, we will discuss a vast therapeutic potential for the STAT3-inhibiting single-agent treatment innovation that, as the desired outcome, could block tumor growth and generally prevent muscle cachexia.


Assuntos
Caquexia/tratamento farmacológico , Janus Quinase 2 , Terapia de Alvo Molecular/métodos , Atrofia Muscular/tratamento farmacológico , Fator de Transcrição STAT3 , Animais , Caquexia/etiologia , Caquexia/metabolismo , Humanos , Janus Quinase 2/antagonistas & inibidores , Janus Quinase 2/metabolismo , Janus Quinase 2/fisiologia , Terapia de Alvo Molecular/tendências , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Atrofia Muscular/etiologia , Atrofia Muscular/metabolismo , Neoplasias/complicações , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Neoplasias/patologia , Fosforilação/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Fator de Transcrição STAT3/antagonistas & inibidores , Fator de Transcrição STAT3/metabolismo , Fator de Transcrição STAT3/fisiologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia
9.
Exp Hematol ; 91: 10-21, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32910996

RESUMO

Erythropoiesis is one of the most demanding processes in the body, with more than 2 million red blood cells produced every second. Multiple hereditary and acquired red blood cell disorders arise from this complex system, with existing treatments effective in managing some of these conditions but few offering a long-term cure. Finding new treatments relies on the full understanding of the cellular and molecular interactions associated with the production and maturation of red blood cells, which take place within the erythroblastic island niche. The elucidation of processes associated within the erythroblastic island niche in health and during stress erythropoiesis has relied on in vivo modeling in mice, with complexities dissected using simple in vitro systems. Recent progress using state-of-the-art stem cell technology and gene editing has enabled a more detailed study of the human niche. Here, we review these different models and describe how they have been used to identify and characterize the cellular and molecular pathways associated with red blood cell production and maturation. We speculate that these systems could be applied to modeling red blood cell diseases and finding new druggable targets, which would prove especially useful for patients resistant to existing treatments. These models could also aid in research into the manufacture of red blood cells in vitro to replace donor blood transfusions, which is the most common treatment of blood disorders.


Assuntos
Modelos Animais de Doenças , Eritroblastos/citologia , Eritropoese/fisiologia , Modelos Biológicos , Nicho de Células-Tronco/fisiologia , Estresse Fisiológico/fisiologia , Animais , Moléculas de Adesão Celular/deficiência , Comunicação Celular , Células Cultivadas , Técnicas de Cocultura , Avaliação Pré-Clínica de Medicamentos , Eritropoese/efeitos dos fármacos , Eritropoese/genética , Hematínicos/uso terapêutico , Doenças Hematológicas/tratamento farmacológico , Doenças Hematológicas/fisiopatologia , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Janus Quinase 2/genética , Janus Quinase 2/fisiologia , Macrófagos/classificação , Macrófagos/fisiologia , Camundongos , Camundongos Transgênicos , Nicho de Células-Tronco/efeitos dos fármacos , Estresse Fisiológico/genética
10.
F1000Res ; 92020.
Artigo em Inglês | MEDLINE | ID: mdl-32983414

RESUMO

More than 50 years of efforts to identify the major cytokine responsible for red blood cell (RBC) production (erythropoiesis) led to the identification of erythropoietin (EPO) in 1977 and its receptor (EPOR) in 1989, followed by three decades of rich scientific discovery. We now know that an elaborate oxygen-sensing mechanism regulates the production of EPO, which in turn promotes the maturation and survival of erythroid progenitors. Engagement of the EPOR by EPO activates three interconnected signaling pathways that drive RBC production via diverse downstream effectors and simultaneously trigger negative feedback loops to suppress signaling activity. Together, the finely tuned mechanisms that drive endogenous EPO production and facilitate its downstream activities have evolved to maintain RBC levels in a narrow physiological range and to respond rapidly to erythropoietic stresses such as hypoxia or blood loss. Examination of these pathways has elucidated the genetics of numerous inherited and acquired disorders associated with deficient or excessive RBC production and generated valuable drugs to treat anemia, including recombinant human EPO and more recently the prolyl hydroxylase inhibitors, which act partly by stimulating endogenous EPO synthesis. Ongoing structure-function studies of the EPOR and its essential partner, tyrosine kinase JAK2, suggest that it may be possible to generate new "designer" drugs that control selected subsets of cytokine receptor activities for therapeutic manipulation of hematopoiesis and treatment of blood cancers.


Assuntos
Eritrócitos/citologia , Eritropoese , Eritropoetina/fisiologia , Receptores da Eritropoetina/fisiologia , Humanos , Janus Quinase 2/fisiologia , Transdução de Sinais
11.
Biomed Pharmacother ; 131: 110750, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-32942160

RESUMO

Rhizoma Paris is a popular Chinese medicine in clinics. It contains four main saponins which are its major bioactive compounds. These saponins are Paris saponin I, II, VI and VII (PSI, PSII, PSVI and PSVII, respectively). Up to now, the research using HUVEC cells to evaluate the anti-angiogenic activity of four saponins is blank. The purpose of this study was to evaluate the anti-angiogenic properties (also known as angiotoxicity) of the four saponins in Rhizoma Paris on vascular endothelial cells-HUVEC cells, and to investigate the underlying mechanism, which has not been studied before. In this study, MTT assay, Lactate dehydrogenase (LDH) assay, wound healing experiments, transwell cell invasion assay, tubule formation experiment, DAPI staining, AV-PI double staining, and cell cycle analysis were used to determine the effects of Paris saponins. The results showed that, with increases in concentrations of PSI, PSII, PSVI and PSVII, the viability of HUVEC cells decreased significantly. In addition, four saponins dose-dependent enhanced LDH release and inhibited HUVEC cell migration, invasion, and angiogenesis. In terms of mechanism, PSI significantly inhibited protein expression in multiple signaling pathways. In particular, with the VEGF2 as the target, it activate the downstream PI3K / AKT / mTOR, SRC / eNOS, P38, PLCγ / ERK / MERK and JAK2/STAT3 signaling pathways. In conclusion, PSI, PSII, PSVI and PSVII can inhibit endothelial cell proliferation, migration and invasion, block endothelial cell cycle, induce endothelial cell apoptosis, act on protein expression in several anti-angiogenic signaling pathways, and finally inhibit angiogenesis in vitro. This study provides further data support for the clinical application of Paris saponins as antiangiogenic drugs.


Assuntos
Inibidores da Angiogênese/farmacologia , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Saponinas/farmacologia , Transdução de Sinais/efeitos dos fármacos , Apoptose/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Diosgenina/análogos & derivados , Diosgenina/farmacologia , MAP Quinases Reguladas por Sinal Extracelular/fisiologia , Células Endoteliais da Veia Umbilical Humana/fisiologia , Humanos , Janus Quinase 2/fisiologia , Óxido Nítrico Sintase Tipo III/fisiologia , Fosfatidilinositol 3-Quinases/fisiologia , Fosfolipase C gama/fisiologia , Proteínas Proto-Oncogênicas c-akt/fisiologia , Fator de Transcrição STAT3/fisiologia , Serina-Treonina Quinases TOR/fisiologia , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/fisiologia , Quinases da Família src/fisiologia
12.
J Clin Invest ; 130(5): 2630-2643, 2020 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-32045382

RESUMO

Arterial cardiovascular events are the leading cause of death in patients with JAK2V617F myeloproliferative neoplasms (MPNs). However, their mechanisms are poorly understood. The high prevalence of myocardial infarction without significant coronary stenosis or atherosclerosis in patients with MPNs suggests that vascular function is altered. The consequences of JAK2V617F mutation on vascular reactivity are unknown. We observe here increased responses to vasoconstrictors in arteries from Jak2V617F mice resulting from a disturbed endothelial NO pathway and increased endothelial oxidative stress. This response was reproduced in WT mice by circulating microvesicles isolated from patients carrying JAK2V617F and by erythrocyte-derived microvesicles from transgenic mice. Microvesicles of other cellular origins had no effect. This effect was observed ex vivo on isolated aortas, but also in vivo on femoral arteries. Proteomic analysis of microvesicles derived from JAK2V617F erythrocytes identified increased expression of myeloperoxidase as the likely mechanism accounting for their effect. Myeloperoxidase inhibition in microvesicles derived from JAK2V617F erythrocytes suppressed their effect on oxidative stress. Antioxidants such as simvastatin and N-acetyl cysteine improved arterial dysfunction in Jak2V617F mice. In conclusion, JAK2V617F MPNs are characterized by exacerbated vasoconstrictor responses resulting from increased endothelial oxidative stress caused by circulating erythrocyte-derived microvesicles. Simvastatin appears to be a promising therapeutic strategy in this setting.


Assuntos
Eritrócitos/fisiologia , Mutação com Ganho de Função , Janus Quinase 2/genética , Janus Quinase 2/fisiologia , Transtornos Mieloproliferativos/genética , Transtornos Mieloproliferativos/fisiopatologia , Animais , Antioxidantes/farmacologia , Aorta Torácica/efeitos dos fármacos , Aorta Torácica/fisiopatologia , Doenças Cardiovasculares/etiologia , Doenças Cardiovasculares/genética , Doenças Cardiovasculares/fisiopatologia , Micropartículas Derivadas de Células/fisiologia , Artéria Femoral/efeitos dos fármacos , Artéria Femoral/fisiopatologia , Humanos , Técnicas In Vitro , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Transtornos Mieloproliferativos/complicações , Estresse Oxidativo , Sinvastatina/farmacologia , Vasoconstrição/efeitos dos fármacos , Vasoconstrição/fisiologia
13.
Int J Radiat Biol ; 96(4): 434-447, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-31850822

RESUMO

Purpose: Pro-inflammatory cytokines within the tumor microenvironment, such as IL-6, contribute to the maintenance of stem cells and promote their survival following treatment. The IL-6/STAT3 pathway is a key regulator of genes involved in cancer progression. Activation of STAT3 promotes expansion of cancer stem cells in triple negative breast cancer. Radiation has also been shown to expand cancer stem cell populations and can induce stemness in nonstem cells. However, the role of IL-6/STAT3 in radiation-induced changes in cellular plasticity is unclear.Materials and methods: Expression and secretion of IL-6 from triple-negative breast cancer cell lines SUM159PT and MDA-MB-231 were determined after radiation treatment by real-time PCR and ELISA. Activation of STAT3 after radiation was determined by western blotting. Changes in cellular plasticity induced by radiation were determined by examining ALDEFLUOR activity, gene expression analysis of aldehyde dehydrogenase isoforms and mammosphere forming assays with and without the addition of STAT3 inhibitors. To determine the effect of radiation on nonstem cell populations, experiments were also carried out in ALDEFLUOR sorted cells.Results: Radiation induced an inflammatory response in both cell lines that resulted in activation of STAT3. Additionally, radiation induced a stem-like state as evidenced by an increased activity and expression of the ALDH isoforms ALDH1A1 and ALDH1A3, and increased self-renewal capabilities. Radiation increased ALDH activity and self-renewal in non-stem cell (ALDH-) populations, suggesting radiation-induced cellular reprograming. However, inhibition of STAT3 blocked the radiation-induced stem-like state in both ALDEFLUOR positive and negative populations, and enhanced radiosensitivity.Conclusions: Radiation-induced changes in cellular plasticity are STAT3 dependent and may be a potential target to reduce radioresistance in TNBC and improve treatment outcome.


Assuntos
Neoplasias da Mama/radioterapia , Plasticidade Celular/efeitos da radiação , Inflamação/fisiopatologia , Células-Tronco Neoplásicas/efeitos da radiação , Tolerância a Radiação , Fator de Transcrição STAT3/fisiologia , Aldeído Desidrogenase/metabolismo , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Feminino , Humanos , Interleucina-6/fisiologia , Janus Quinase 2/fisiologia , Fator de Transcrição STAT3/antagonistas & inibidores , Transdução de Sinais/efeitos da radiação
14.
Neuroscience ; 422: 134-145, 2019 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-31682951

RESUMO

Schwann cells (SCs) combined with acellular nerve allografts (ANAs) effectively promote the regeneration and repair of peripheral nerves, but the exact mechanism has not been fully elucidated. However, the disadvantages of SCs include their limited source and slow rate of expansion in vitro. Previous studies have found that adipose-derived stem cells have the ability to differentiate into Schwann-like cells. Therefore, we speculated that Schwann-like cells combined with ANAs could profoundly facilitate nerve regeneration and repair. The aim of the present study was to investigate the cellular and molecular mechanisms of regeneration and repair. In this study, tissue-engineered nerves were first constructed by adipose-derived Schwann-like cells and ANAs to bridge missing sciatic nerves. Then, the rats were randomly divided into five groups (n = 12 per group): a Control group; a Model group; an ADSC group; an SC-L group; and a DMEM group. Twelve weeks postsurgery, behavioral function tests and molecular biological techniques were used to evaluate the function of regenerated nerves and the relevant molecular mechanisms after sciatic nerve injury (SNI). The results showed that adipose-derived Schwann-like cells combined with ANAs markedly promoted sciatic nerve regeneration and repair. These findings also demonstrated that the expression of neurotrophic factors (NFs) was increased, and the expression of Janus activated kinase2 (JAK2)/P-JAK2, signal transducer and activator of transcription-3 (STAT3)/P-STAT3 was decreased in the spinal cord after SNI. Therefore, these results suggested that highly expressed NFs in the spinal cord could promote nerve regeneration and repair by inhibiting activation of the JAK2/STAT3 signaling pathway.


Assuntos
Aloenxertos/transplante , Janus Quinase 2/fisiologia , Regeneração Nervosa/fisiologia , Fator de Transcrição STAT3/fisiologia , Nervo Isquiático/fisiopatologia , Animais , Fator Neurotrófico Derivado do Encéfalo/biossíntese , Fator Neurotrófico Ciliar/biossíntese , Masculino , Transplante de Células-Tronco Mesenquimais/métodos , Fator de Crescimento Neural/biossíntese , Neurônios/transplante , Ratos , Recuperação de Função Fisiológica/fisiologia , Nervo Isquiático/lesões , Nervo Isquiático/cirurgia , Transdução de Sinais/fisiologia , Medula Espinal/metabolismo
15.
Cells ; 8(8)2019 08 08.
Artigo em Inglês | MEDLINE | ID: mdl-31398915

RESUMO

Clonal alterations in hematopoietic cells occur during aging and are often associated with the establishment of a subclinical inflammatory environment. Several age-related conditions and diseases may be initiated or promoted by these alterations. JAK2 mutations are among the most frequently mutated genes in blood cells during aging. The most common mutation within the JAK2 gene is JAK2-V617F that leads to constitutive activation of the kinase and thereby aberrant engagement of downstream signaling pathways. JAK2 mutations can act as central drivers of myeloproliferative neoplasia, a pre-leukemic and age-related malignancy. Likewise, hyperactive JAK-signaling is a hallmark of immune diseases and critically influences inflammation, coagulation and thrombosis. In this review we aim to summarize the current knowledge on JAK2 in clonal hematopoiesis during aging, the role of JAK-signaling in inflammation and lymphocyte biology and JAK2 function in age-related diseases and malignant transformation.


Assuntos
Envelhecimento/metabolismo , Hematopoese , Inflamação/metabolismo , Janus Quinase 2/fisiologia , Transtornos Mieloproliferativos/metabolismo , Neoplasias/metabolismo , Animais , Humanos , Camundongos
16.
Thyroid ; 29(5): 700-713, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30963819

RESUMO

Background: There is growing evidence that sphingosine-1-phosphate (S1P), a pleiotropic bioactive sphingolipid metabolite synthesized intracellularly by two closely related sphingosine kinases (SphKs), SphK1 and SphK2, is involved in inflammation. However, the role of SphKs/S1P/S1P receptors (S1PRs) in autoimmune thyroiditis (AIT) has not been studied to date. Methods: This study examined whether SphK1/S1P/S1PR1 signaling is aberrantly altered in thyroid tissues and serum of both AIT patients and a spontaneously autoimmune thyroiditis (SAT) mouse model. Murine CD4+T cells were employed to further investigate the downstream signaling of SphK1/S1P/S1PR1. Furthermore, a total of 102 NOD.H-2h4 mice, randomly divided into different groups, were used to investigate the therapeutic effect of S1PR1 blockade and its potential mechanism. Results: We found that components of the SphK1/S1P/S1PR1 pathway were abnormally expressed in patients with Hashimoto thyroiditis and in a SAT mouse model. In addition, S1P could activate signal transducer and activator of transcription 3 (STAT3) through S1PR1 and its downstream signaling pathways in CD4+T cells of NOD.H-2h4 mice. Furthermore, an in vivo study demonstrated that blocking S1PR1 by FTY720 administration could reduce the incidence and severity of thyroiditis and goiter in SAT mice in a time-dependent manner. The proportions of STAT3-related and inflammation-related cell subtypes, such as T helper 1, T helper 17, and follicular T helper cells, were elevated in the SAT group when compared to the control group, and these cell subtypes decreased after FTY720 administration. Furthermore, the downstream inflammatory cytokines of STAT3 were also downregulated after FTY720 administration. Conclusion: The present study shows that blocking Sphk1/S1P/S1PR1 signaling can ameliorate the severity of AIT, providing evidence of a promising therapeutic target for AIT.


Assuntos
Lisofosfolipídeos/farmacologia , Fosfotransferases (Aceptor do Grupo Álcool)/fisiologia , Receptores de Esfingosina-1-Fosfato/fisiologia , Esfingosina/análogos & derivados , Tireoidite Autoimune/etiologia , Animais , Citocinas/genética , Modelos Animais de Doenças , Feminino , Cloridrato de Fingolimode/farmacologia , Humanos , Janus Quinase 2/fisiologia , Camundongos , Fator de Transcrição STAT3/fisiologia , Transdução de Sinais/fisiologia , Esfingosina/farmacologia , Serina-Treonina Quinases TOR/fisiologia
17.
Indian J Dermatol Venereol Leprol ; 85(5): 455-461, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31031313

RESUMO

BACKGROUND: Alopecia areata is an autoimmune disease that occurs as a result of the loss of the inherent immune privilege of the hair follicle. It has been recently demonstrated that the interferon-γ/interleukin-15 feedback loop that signals via the Janus kinase-signal transducer and activator of transcription pathway is critical to the breakdown of this immune privilege. AIMS: To evaluate the immunological distribution of CD4+ T-cells, CD8+ T-cells, phosphorylated signal transducer and activator of transcription 1 and study its relation with the clinical and histopathological findings of the disease. MATERIALS AND METHODS: A total of 30 patients of alopecia areata were included in the study. Following a detailed history and clinical examination, a scalp biopsy was performed. Histopathology was studied and immunohistochemistry was done to demonstrate the positivity and distribution of CD4+ T-cells, CD8+ T-cells and phosphorylated signal transducer and activator of transcription 1. RESULTS: The follicular count, number of anagen and terminal hair were found to be decreased, whereas the catagen, telogen and vellus hair were found to be increased in number. A peribulbar CD4+ T-cell infiltrate was seen in 70% cases, whereas a CD8+ T-cell infiltrate was seen in 83.3% cases. An intrabulbar CD4+ T-cell infiltrate was seen in 26.7% cases, whereas a CD8+ T-cell infiltrate was seen in 70% cases. Among the 25 hair follicles dermal papilla identified, 36.8% cases were found to be positive for phospho-signal transducer and activation of transcription-1. LIMITATIONS: The drawbacks of our study included a small sample size and the use of only vertical sectioning for the scalp biopsy samples. CONCLUSION: Phosphorylated signal transducer and activator of transcription 1 positivity as an indicator of signalling via the Janus kinase-1/2 pathway was seen in 36.8% of our cases highlighting the integral role of this pathway in the pathogenesis of alopecia areata.


Assuntos
Alopecia em Áreas/imunologia , Alopecia em Áreas/patologia , Janus Quinase 1/fisiologia , Janus Quinase 2/fisiologia , Transcrição Gênica/fisiologia , Adolescente , Adulto , Linfócitos T CD4-Positivos/fisiologia , Linfócitos T CD8-Positivos/fisiologia , Criança , Pré-Escolar , Estudos Transversais , Feminino , Folículo Piloso/imunologia , Folículo Piloso/patologia , Humanos , Janus Quinases/fisiologia , Masculino , Pessoa de Meia-Idade , Adulto Jovem
18.
Mol Biol Rep ; 46(1): 207-215, 2019 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-30415442

RESUMO

Recently, much attention has been focused on the use of miRNAs in cancer treatment. The role of proto-oncogene Janus kinase-2 (JAK-2) in proliferation and survival of gastric cancer has been previously documented. The aim of this study was to evaluate the effect of a chimera consisted of nucleolin specific aptamer (NCL-Apt) and miRNA let-7d on JAK2 expression level and activity in gastric cancer cells. NCL-Apt-miRNA let-7d chimera was prepared by two methods. Gastric cancer (MKN-45) cell line and control cell line of human dermal fibroblast (HDF) were treated with the chimera and the changes in JAK2 expression and activity were determined using real-time PCR and ELISA techniques, respectively. In MKN-45 cells, the chimera caused significant decrease in JAK2 expression level and activity compared to the aptamer alone and miRNA mimic negative control. Nevertheless, transfected miRNA let-7d showed remarkable reduction in the expression level of JAK2 in comparison with control state in both MKN-45 and HDF, confirmed unspecific effect of let-7d on normal and cancerous cells. With regard to the synergic effect of this chimera on JAK2 activity, it might be viewed as a therapeutic candidate in gastric cancer. However, further studies are warranted to prove it.


Assuntos
MicroRNAs , Fosfoproteínas , Proteínas de Ligação a RNA , Neoplasias Gástricas , Humanos , Aptâmeros de Nucleotídeos/genética , Aptâmeros de Nucleotídeos/farmacologia , Linhagem Celular Tumoral , Regulação da Expressão Gênica/genética , Regulação Neoplásica da Expressão Gênica/genética , Janus Quinase 2/genética , Janus Quinase 2/fisiologia , MicroRNAs/genética , MicroRNAs/fisiologia , Fosfoproteínas/genética , Fosfoproteínas/fisiologia , Dados Preliminares , Proto-Oncogene Mas , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/fisiologia , Neoplasias Gástricas/genética , Nucleolina
19.
Reprod Sci ; 26(6): 829-838, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30526368

RESUMO

Previous studies show that methylseleninic acid (MSA), which is the most common selenium derivative used as a drug in humans, exerts specific cytotoxic effects in several cancer cell types. However, the complex mechanism of these effects has not been fully elucidated. Here, we demonstrate by Cell Counting Kit-8 in mouse breast cancer cell line 4T1 that MSA inhibits cell viability in a concentration-dependent (5, 10, 20 µmol/L) and time-dependent (6, 12, 24 hours) manner. Flow cytometry, Western blot, and Reverse Transcription-Polymerase Chain Reaction (RT-PCR) analyses indicated that MSA inhibits cancer cell invasion and induces apoptosis by the activation of caspase-3, poly ADP ribose polymerase 1 (PARP1), and BCL2-associated X. Furthermore, MSA demonstrated anticancer activity by inhibiting the Janus kinase 2/signal transducers and activators of transcription 3 (JAK2/STAT3) pathway. The MSA treatment for 24 hours decreased the phosphorylation of JAK2 and STAT3 in 4T1 cells by Western blot. We also confirmed this with the use of a JAK2 chemical inhibitor, AG490, as a positive control. In a 4T1 orthotopic allograft model, morphological and TdT-mediated dUTP nick-end labeling analyses showed that MSA treatment (1.5 mg/kg/weight) for 28 days inhibits tumor growth consistent with the clinical anticancer drug cyclophosphamide. Our observations demonstrate that MSA is a potent anticancer drug in breast cancer and uncovered a key role of the JAK2/STAT3 pathway in modulating tumor growth.


Assuntos
Antineoplásicos , Neoplasias da Mama/tratamento farmacológico , Janus Quinase 2/antagonistas & inibidores , Compostos Organosselênicos/uso terapêutico , Fator de Transcrição STAT3/antagonistas & inibidores , Transdução de Sinais/efeitos dos fármacos , Animais , Apoptose/efeitos dos fármacos , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Feminino , Janus Quinase 2/fisiologia , Camundongos , Camundongos Endogâmicos BALB C , Invasividade Neoplásica , Compostos Organosselênicos/farmacologia , Fator de Transcrição STAT3/fisiologia
20.
Cancer Cell ; 34(2): 298-314.e7, 2018 08 13.
Artigo em Inglês | MEDLINE | ID: mdl-30107178

RESUMO

Over half of colorectal cancers (CRCs) harbor TP53 missense mutations (mutp53). We show that the most common mutp53 allele R248Q (p53Q) exerts gain of function (GOF) and creates tumor dependence in mouse CRC models. mutp53 protein binds Stat3 and enhances activating Stat3 phosphorylation by displacing the phosphatase SHP2. Ablation of the p53Q allele suppressed Jak2/Stat3 signaling, growth, and invasiveness of established, mutp53-driven tumors. Treating tumor-bearing mice with an HSP90 inhibitor suppressed mutp53 levels and tumor growth. Importantly, human CRCs with stabilized mutp53 exhibit enhanced Jak2/Stat3 signaling and are associated with poorer patient survival. Cancers with TP53R248Q/W are associated with a higher patient death risk than are those having nonR248 mutp53. These findings identify GOF mutp53 as a therapeutic target in CRC.


Assuntos
Neoplasias Colorretais/terapia , Mutação , Fator de Transcrição STAT3/fisiologia , Proteína Supressora de Tumor p53/genética , Animais , Linhagem Celular Tumoral , Proliferação de Células , Neoplasias Colorretais/genética , Neoplasias Colorretais/patologia , Proteínas de Choque Térmico HSP90/antagonistas & inibidores , Humanos , Janus Quinase 2/fisiologia , Perda de Heterozigosidade , Camundongos , Invasividade Neoplásica , Proteína Supressora de Tumor p53/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA