Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Anticancer Drugs ; 35(2): 163-176, 2024 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-37948318

RESUMEN

Chemotherapy is the main treatment option for acute myeloid leukemia (AML), but acquired resistance of leukemic cells to chemotherapeutic agents often leads to difficulties in AML treatment and disease relapse. High calcitonin receptor-like (CALCRL) expression is closely associated with poorer prognosis in AML patients. Therefore, this study was performed by performing CALCRL overexpression constructs in AML cell lines HL-60 and Molm-13 with low CALCRL expression. The results showed that overexpression of CALCRL in HL-60 and Molm-13 could confer resistance properties to AML cells and reduce the DNA damage and cell cycle G0/G1 phase blocking effects caused by daunorubicin (DNR) and others. Overexpression of CALCRL also reduced DNR-induced apoptosis. Mechanistically, the Cancer Clinical Research Database analyzed a significant positive correlation between XRCC5 and CALCRL in AML patients. Therefore, the combination of RT-PCR and Western blot studies further confirmed that the expression levels of XRCC5 and PDK1 genes and proteins were significantly upregulated after overexpression of CALCRL. In contrast, the phosphorylation levels of AKT/PKCε protein, a downstream pathway of XRCC5/PDK1, were significantly upregulated. In the response study, transfection of overexpressed CALCRL cells with XRCC5 siRNA significantly upregulated the drug sensitivity of AML to DNR. The expression levels of PDK1 protein and AKT/PKCε phosphorylated protein in the downstream pathway were inhibited considerably, and the expression of apoptosis-related proteins Bax and cleaved caspase-3 were upregulated. Animal experiments showed that the inhibitory effect of DNR on the growth of HL-60 cells and the number of bone marrow invasions were significantly reversed after overexpression of CALCRL in nude mice. However, infection of XCRR5 shRNA lentivirus in HL-60 cells with CALCRL overexpression attenuated the effect of CALCRL overexpression and upregulated the expression of apoptosis-related proteins induced by DNR. This study provides a preliminary explanation for the relationship between high CALCRL expression and poor prognosis of chemotherapy in AML patients. It offers a more experimental basis for DNR combined with molecular targets for precise treatment in subsequent studies.


Asunto(s)
Daunorrubicina , Leucemia Mieloide Aguda , Animales , Ratones , Humanos , Daunorrubicina/farmacología , Regulación hacia Arriba , Ratones Desnudos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Células HL-60 , Apoptosis , Autoantígeno Ku/genética , Autoantígeno Ku/metabolismo , Autoantígeno Ku/farmacología , TYK2 Quinasa/genética , TYK2 Quinasa/metabolismo , TYK2 Quinasa/farmacología , Janus Quinasa 1/genética , Janus Quinasa 1/metabolismo , Janus Quinasa 1/farmacología , Proteína Similar al Receptor de Calcitonina/genética , Proteína Similar al Receptor de Calcitonina/metabolismo
2.
Clinics (Sao Paulo) ; 78: 100265, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37562217

RESUMEN

OBJECTIVES: The authors determined the level of Expression of Leptin (LEP) in Polycystic Ovary Syndrome (PCOS) patients with or without obesity and in GCs treated with insulin. METHODS: LEP expression was first assessed in ovary cortex specimens collected from women with PCOS with or without obesity as well as from healthy controls. Ovarian Granulosa Cells (OGCs) induced by insulin extracted from a mouse model were used in further functional research. RESULTS: Real-time PCR and western blotting indicated that LEP expression was upregulated in GCs induced by insulin, in comparison with that in GCs not induced by insulin. Furthermore, the knockdown of LEP resulted in a reduction in growth and multiplication and an increase in apoptosis and inflammation in GCs induced by insulin. Next, the authors evaluated the effect of LEP on three key pathways of inflammation (MAPK, NF-kB, and JAK1/STAT3); results showed that the JAK1/STAT3 pathway was induced by LEP knockdown, as evidenced by the upregulation of phosphor-JAK1, phosphor-STAT3, and nuclear STAT3 expression. Administration of curcumin, a specific inhibitor of STAT3, counteracted the effect of LEP knockdown on cell inflammation and apoptosis. CONCLUSION: The present data suggest that upregulation of LEP expression in the PCOS granulosa cell model is essential for reducing apoptosis and inflammation by modulating the JAK1/STAT3 pathway axis.


Asunto(s)
Síndrome del Ovario Poliquístico , Humanos , Ratones , Animales , Femenino , Síndrome del Ovario Poliquístico/metabolismo , Leptina/efectos adversos , Leptina/metabolismo , Células de la Granulosa/metabolismo , Insulina , Obesidad , Apoptosis , Janus Quinasa 1/metabolismo , Janus Quinasa 1/farmacología , Factor de Transcripción STAT3/metabolismo , Factor de Transcripción STAT3/farmacología
3.
Biochem Genet ; 61(1): 372-389, 2023 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-35931919

RESUMEN

Atherosclerosis (AS) is the typical cardiovascular disease, which is the main underlying inducement of cardiovascular diseases. Aberrant expression of long noncoding RNA HLA complex group 11 (HCG11) was engaged with atherosclerosis. The objective of the present research was to explore the role and the potential mechanism of HCG11 in AS. Human umbilical vein endothelial cells (HUVECs) were stimulated with oxidized low-density lipoprotein (ox-LDL) to induce the AS model in vitro. The cell viability was detected by MTT assay. Flow cytometry was performed to determine cell pyroptosis. Gene and protein levels were detected by qPCR or Western blot assay. The interaction between HCG11, miR-224-3p, and Janus kinase 1 (JAK1) was validated by dual-luciferase reporter assays. Ox-LDL treatment aggravated cell pyroptosis and inflammation in HUVECs. And the levels of HCG11 and JAK1 was enhanced in ox-LDL-induced HUVECs, while miR-224-3p expression was reduced. Additionally, knockdown of HCG11 or miR-224-3p overexpression reversed the ox-LDL-induced cell viability decline and the increase of cell pyroptosis and inflammation-related proteins, including gasdermin D N-terminal (GSDMD-N), Caspase-1, NOD-like receptor family pyrin domain-containing 3 (NLRP3), interleukin 18 (IL-18), and interleukin 1beta (IL-1ß). Moreover, HCG11 could modulate the JAK1 expression via targeting miR-224-3p. The inhibitory effect of HCG11 silencing on cell pyroptosis and inflammation was reversed by miR-224-3p knockdown. Furthermore, overexpression of miR-224-3p could repress the ox-LDL-induced cell pyroptosis and inflammation via regulating JAK1 expression. Knockdown of HCG11 alleviated cell pyroptosis and inflammation induced by ox-LDL via targeting the miR-224-3p/JAK1 axis, indicating that HCG11 could be the latent target of diagnosis or treatment for AS.


Asunto(s)
Aterosclerosis , MicroARNs , ARN Largo no Codificante , Humanos , MicroARNs/genética , MicroARNs/metabolismo , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Janus Quinasa 1/genética , Janus Quinasa 1/farmacología , Aterosclerosis/genética , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Inflamación/genética , Lipoproteínas LDL/farmacología , Apoptosis , Proliferación Celular/fisiología
4.
Am J Physiol Endocrinol Metab ; 323(5): E405-E417, 2022 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-36103628

RESUMEN

Polycystic ovary syndrome (PCOS) is an extremely common endocrine-metabolic disorder and the main cause of infertility in premenopausal women, thus targeted treatments are sorely needed. Accumulative evidence showed that exogenous supplementation of IL-22 in PCOS mice may be of significant positive effect on insulin resistance (IR), a root causative factor for this condition, but much remained unknown about its mechanism. According to our previous study, troxerutin, a common anticoagulant and thrombolytic agent in clinic, alleviated various PCOS-like phenotypes in dihydrotestosterone (DHT)-treated rat model with unclear mechanism. Here, glucose tolerance tests (GTTs), insulin tolerance tests (ITTs), and homeostatic model assessment of insulin resistance (HOMA-IR) analyses revealed that troxerutin treatment in DHT-treated rats also significantly improved insulin resistance and enhanced serum IL-22 levels, which thereby activated IL-22R1/Janus kinase 1 (JAK1)/signal transducer and activator of transcription-3 (STAT3) signaling pathway in pancreatic islet. This protective effect of troxerutin on insulin resistance improvement was blocked by an inhibitor of p-STAT3, S3I-201. Troxerutin administration to DHT rats decreased the relative abundance of Bifidobacterium and enhanced secondary bile acid profiles, which were positively correlated with serum IL-22 concentration. Conclusively, the present study reported that troxerutin is an endogenous enhancer of IL-22 and the effect of troxerutin on insulin resistance improvement was via IL-22R1/JAK1/STAT3 signaling activation in a DHT-induced PCOS rat model. These insights may be translated into a primary therapeutic agent for PCOS with insulin resistance and hyperandrogenism.NEW & NOTEWORTHY Troxerutin decreased the relative abundance of Bifidobacterium, along with enhancement of secondary bile acids/IL-22 system, which thereby activated its downstream IL-22R1/JAK1/STAT3 signaling pathway in pancreatic ß cells, subsequently attenuated insulin resistance (IR), hyperandrogenism and PCOS-like phenotypes in DHT-induced PCOS rat models. Troxerutin is an endogenous IL-22 enhancer and may be of therapeutic value for PCOS with insulin resistance.


Asunto(s)
Hiperandrogenismo , Resistencia a la Insulina , Síndrome del Ovario Poliquístico , Animales , Femenino , Humanos , Ratones , Ratas , Anticoagulantes , Ácidos y Sales Biliares/farmacología , Dihidrotestosterona/farmacología , Fibrinolíticos , Insulina/metabolismo , Resistencia a la Insulina/fisiología , Janus Quinasa 1/metabolismo , Janus Quinasa 1/farmacología , Síndrome del Ovario Poliquístico/inducido químicamente , Síndrome del Ovario Poliquístico/tratamiento farmacológico , Transducción de Señal , Factor de Transcripción STAT3/metabolismo , Interleucina-22
5.
Expert Opin Drug Metab Toxicol ; 18(5): 347-355, 2022 May.
Artículo en Inglés | MEDLINE | ID: mdl-35796377

RESUMEN

INTRODUCTION: Atopic dermatitis (AD) is the most common inflammatory skin disorder. Despite the high disease burden, the therapeutic options are limited and their efficacy in controlling AD might be partially satisfactory. AREAS COVERED: Most of the key mediators in AD pathogenesis act through the JAK/STAT signaling pathway, which represents a valid therapeutic target. The first generation of JAK inhibitors, namely tofacitinib and ruxolitinib, inhibit multiple JAKs, whereas newer JAK inhibitors show more selective inhibitory effects for specific JAKs. The aim of this review was to discuss the role of the JAK/STAT pathway in AD and its inhibition, with a special focus on pharmacodynamic properties. EXPERT OPINION: JAK inhibitors have different selectivity for various JAK molecules, which influences their pharmacodynamics, efficacy, and safety profile. Since many key cytokines in AD signal through JAK1, the selective JAK1 inhibition may be effective, avoiding the concomitant inhibition of JAK2- and JAK3-dependent pathways could be associated with additional safety issues. Therefore, selective JAK1 inhibitors may represent promising therapeutic agents for AD, as they might prevent off-target effects of JAK inhibitors, especially related to the hematologic profile.


Asunto(s)
Dermatitis Atópica , Inhibidores de las Cinasas Janus , Dermatitis Atópica/tratamiento farmacológico , Humanos , Janus Quinasa 1/metabolismo , Janus Quinasa 1/farmacología , Inhibidores de las Cinasas Janus/efectos adversos , Quinasas Janus/metabolismo , Quinasas Janus/farmacología , Inhibidores de Proteínas Quinasas/efectos adversos , Factores de Transcripción STAT/metabolismo , Factores de Transcripción STAT/farmacología , Factores de Transcripción STAT/uso terapéutico , Transducción de Señal
6.
Cell Metab ; 34(3): 487-501.e8, 2022 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-35235776

RESUMEN

The Krebs cycle-derived metabolite itaconate and its derivatives suppress the inflammatory response in pro-inflammatory "M1" macrophages. However, alternatively activated "M2" macrophages can take up itaconate. We therefore examined the effect of itaconate and 4-octyl itaconate (OI) on M2 macrophage activation. We demonstrate that itaconate and OI inhibit M2 polarization and metabolic remodeling. Examination of IL-4 signaling revealed inhibition of JAK1 and STAT6 phosphorylation by both itaconate and OI. JAK1 activation was also inhibited by OI in response to IL-13, interferon-ß, and interferon-γ in macrophages and in T helper 2 (Th2) cells. Importantly, JAK1 was directly modified by itaconate derivatives at multiple residues, including cysteines 715, 816, 943, and 1130. Itaconate and OI also inhibited JAK1 kinase activity. Finally, OI treatment suppressed M2 macrophage polarization and JAK1 phosphorylation in vivo. We therefore identify itaconate and OI as JAK1 inhibitors, suggesting a new strategy to inhibit JAK1 in M2 macrophage-driven diseases.


Asunto(s)
Activación de Macrófagos , Macrófagos , Janus Quinasa 1/metabolismo , Janus Quinasa 1/farmacología , Macrófagos/metabolismo , Transducción de Señal , Succinatos
7.
Bioengineered ; 13(3): 5724-5736, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35184688

RESUMEN

Elevated level of glucolipotoxicity induces the loss of pancreatic ß-cells functions and plays an important role in the development of type 2 diabetes (T2DM). Previous studies have indicated the importance of developing therapies against T2DM, while circular RNA (circRNA) has gained attraction as a modulator of pancreatic ß-cell function. In the present study role of circPIP5K1A in dysfunctional ß cells and mouse pancreas was comprehensively analyzed. INS-1E, as it has close similarity with naïve pancreatic ß-cells, and clinical samples of T2DM patients were used to investigate the effect of circPIP5K1A, miR-552-3p, and Janus kinase 1 (JAK1). While, INS-1E cells were exposed to PAHG conditions (0.5 mM palmitic acid and 28 mM glucose) as studies have suggested that increased level of fatty acid and glucose resulted in autophagy activation of pancreatic ß-cells that leads to T2DM. Key player of JAK1-STAT3 pathway and the level of Reactive Oxygen Species, inflammatory factors, and insulin secretion was detected to analyze the of the active association of circPIP5K1A, miR-552-3p with JAK1pathway. Our study has revealed the elevated level ofcircPIP5K1A and JAK1, but reduced level of miR-552-3pin the serum of T2DM patients. Furthermore, we also found that reduced expression ofcircPIP5K1A leads to decreased rate of inflammation, oxidative damage and apoptosisinINS-1E cells induced by glucolipotoxicity. CircPIP5K1A was available to competitively combine with miR-552-3p, while whose direct target was JAK1. In conclusion, our study suggested a novel involvement of circPIP5K1A in a cross talk between miR5523p/JAK1/STAT3 pathways in ß-cells as a new therapeutic target for T2DM.


Asunto(s)
Diabetes Mellitus Tipo 2 , MicroARNs , Animales , Apoptosis/genética , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Glucosa/metabolismo , Humanos , Inflamación/genética , Janus Quinasa 1/genética , Janus Quinasa 1/metabolismo , Janus Quinasa 1/farmacología , Ratones , MicroARNs/metabolismo , Estrés Oxidativo/genética , ARN Circular/genética
8.
Sci Rep ; 3: 1095, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23393614

RESUMEN

Tumors exhibit complex organization and contain a variety of cell populations. The realization that the regenerative properties of a tumor may be largely confined to a cell subpopulation (cancer stem cell) is driving a new era of anti-cancer research. Cancer stem cells from Glioblastoma Multiforme tumors express markers that are also expressed in non-cancerous neural stem cells, including nestin and Sox2. We previously showed that the transcription factor Hes3 is a marker of neural stem cells, and that its expression is inhibited by JAK activity. Here we show that Hes3 is also expressed in cultures from glioblastoma multiforme which express neural stem cell markers, can differentiate into neurons and glia, and can recapitulate the tumor of origin when transplanted into immunocompromised mice. Similar to observations in neural stem cells, JAK inhibits Hes3 expression. Hes3 RNA interference reduces the number of cultured glioblastoma cells suggesting a novel therapeutic strategy.


Asunto(s)
Neoplasias del Sistema Nervioso Central/patología , Proteínas de Unión al ADN/metabolismo , Glioblastoma/patología , Células Madre Neoplásicas/metabolismo , Factores de Transcripción/metabolismo , Angiopoyetina 2/metabolismo , Animales , Biomarcadores/metabolismo , Neoplasias del Sistema Nervioso Central/tratamiento farmacológico , Neoplasias del Sistema Nervioso Central/metabolismo , Proteínas de Unión al ADN/genética , Células Madre Embrionarias/metabolismo , Factor de Crecimiento Epidérmico/farmacología , Factor 2 de Crecimiento de Fibroblastos/farmacología , Glioblastoma/tratamiento farmacológico , Glioblastoma/metabolismo , Janus Quinasa 1/metabolismo , Janus Quinasa 1/farmacología , Ratones , Células Madre Neoplásicas/patología , Fosforilación , ARN Interferente Pequeño , Proteínas Represoras , Factor de Transcripción STAT3/metabolismo , Factores de Transcripción/genética , Células Tumorales Cultivadas
9.
Drugs ; 72(16): 2117-27, 2012 Nov 12.
Artículo en Inglés | MEDLINE | ID: mdl-23061804

RESUMEN

Ruxolitinib is a selective inhibitor of Janus kinases (JAK) 1 and 2, which are involved in the signalling pathway of various cytokines and growth factors essential to haematopoiesis. JAK 1 and 2 are implicated in the development of myelofibrosis, as well as other haematological malignancies. Ruxolitinib is the first agent approved for the treatment of myelofibrosis. In a randomized, double-blind, placebo-controlled, multicentre trial (COMFORT-I) in patients with myelofibrosis, significantly more ruxolitinib than placebo recipients achieved a ≥ 35% reduction in spleen volume (primary endpoint) at 24 weeks. In a randomized, open-label, multicentre trial (COMFORT-II) in patients with myelofibrosis, significantly more ruxolitinib than best available therapy recipients achieved the same primary endpoint at 48 weeks. Significantly more ruxolitinib than placebo recipients achieved a ≥ 50% reduction in Total Symptom Score at 24 weeks in COMFORT-I. Ruxolitinib generally improved health-related quality-of-life scores, while best available therapy was generally associated with worsened scores at 48 weeks in COMFORT-II. In COMFORT-I, overall survival data appeared to favour ruxolitinib over placebo; of note, most placebo recipients had crossed over to receive ruxolitinib. In COMFORT-II, a significant difference in overall survival between ruxolitinib and best available therapy was not shown; this trial was not powered to detect such a difference. In clinical trials in patients with myelofibrosis, ruxolitinib was generally associated with an acceptable tolerability profile. In the placebo-controlled trial, the most commonly reported grade 3 or 4 adverse events in ruxolitinib recipients were thrombocytopenia, anaemia and neutropenia. These haematological adverse events were mainly managed with dosage interruptions/reductions and/or transfusions, and rarely resulted in discontinuation.


Asunto(s)
Janus Quinasa 1/antagonistas & inhibidores , Janus Quinasa 2/antagonistas & inhibidores , Mielofibrosis Primaria/tratamiento farmacológico , Pirazoles/farmacología , Humanos , Janus Quinasa 1/farmacocinética , Janus Quinasa 1/farmacología , Janus Quinasa 2/farmacocinética , Janus Quinasa 2/farmacología , Nitrilos , Pirazoles/farmacocinética , Pirimidinas , Ensayos Clínicos Controlados Aleatorios como Asunto , Resultado del Tratamiento
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA