Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 81
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Med Sci Monit ; 28: e934914, 2022 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-35279667

RESUMEN

BACKGROUND Curcumol is a hydrogenated austenitic compound with hemiketal. In this study we evaluated the effects of curcumol on local inflammatory response, cell proliferation, and metastasis in endometriosis, and elucidated the underlying mechanisms. MATERIAL AND METHODS Ectopic endometrial stromal cells were treated with increasing doses of curcumol. The MTT assay was used to assess cell viability. FITC-labeled annexin-V/PI double-staining method and flow cytometry were used to determine cell apoptosis. Cell migration was evaluated using a wound healing assay. ELISA kits were used to detect the levels of TNF-alpha, IL-6, and IL-1ß. Western blot assay was used to examine the phosphorylation degree of JAK2 and STAT3 and the expression of Bax, Bcl2, and caspase-3 proteins. Autologous endometrial transplantation was used to establish a rat model to assess the anti-EMS effect of curcumol in vivo. RESULTS Curcumol can inhibit the proliferation of ectopic endometrial stromal cells, promote cell apoptosis, and weaken cell migration ability. Curcumol can reduce the expression of Bax and caspase-3 protein and increase the expression of Bcl2 protein. Curcumol also can inhibit the secretion of inflammatory cytokines, including tumor necrosis cytokines (TNF)-alpha, interleukin (IL)-6, and IL-1ß, by ectopic endometrial stromal cells. In addition, curcumol can also inhibit the phosphorylation of JAK2 and STAT3. In vivo experiments also proved that curcumol could inhibit the growth of ectopic lesions in EMS model rats. CONCLUSIONS Curcumol can inhibit the JAK2/STAT3 pathway, reduce the inflammatory cytokines secreted by ectopic endometrial stromal cells, inhibit cell proliferation and migration, and reduce the volume of ectopic lesions.


Asunto(s)
Apoptosis , ADN/genética , Endometriosis/genética , Janus Quinasa 2/genética , Factor de Transcripción STAT3/genética , Sesquiterpenos/farmacología , Útero/metabolismo , Adulto , Proliferación Celular , Supervivencia Celular , Medicamentos Herbarios Chinos/farmacología , Endometriosis/tratamiento farmacológico , Endometriosis/metabolismo , Femenino , Humanos , Janus Quinasa 2/biosíntesis , Estudios Retrospectivos , Factor de Transcripción STAT3/biosíntesis , Transducción de Señal , Útero/patología , Adulto Joven
2.
Int Immunopharmacol ; 99: 107974, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34358862

RESUMEN

Gestational diabetes mellitus (GDM) is a common complication of pregnancy characterized by intrauterine hyperglycemia, which is often associated with a high risk of obesity and diabetes in the offspring. In this study, we established a GDM mouse model by intraperitoneal injection of streptozotocin to investigate the immuno-inflammatory responses in the liver of adult offspring. Glucose tolerance test (GTT) and insulin tolerance test (ITT) were employed to evaluate the glucose tolerance status. Hematoxylin-eosin staining was used to examine the histological changes in the liver. Quantitative real-timePCR (qRT-PCR) was applied to examine the mRNA expression of immune factors. Western blot and immunofluorescence analyses were used to examine the expression of target protein. Additionally, cell experiments were performed to validate the in vivo results. Compared to the control group, the area of fat vacuoles and the number of lymphocyte cells were significantly higher in the 20 weeks-old offspring of GDM mice. The elevated mRNA level of the pro-inflammatory cytokines IL-1ß, IL-6, IL-33 and immune receptors CD3 and CD36 were found in the liver of F1-GDM. The protein level of IL-6r and the phosphorylation of JAK2 and STAT3 were significantly up-regulated. Moreover, the mRNA level of IL-6, IL-1ß and IL-33 and the phosphorylation of JAK2 and STAT3 were also up-regulated in the hepatocyte treated with high concentration of glucose. Our results suggest that intrauterine hyperglycemia is associated with increased inflammation in the liver of adult male offspring.


Asunto(s)
Diabetes Gestacional/patología , Hepatitis/patología , Hiperglucemia/patología , Hígado/patología , Animales , Citocinas/análisis , Citocinas/biosíntesis , Diabetes Mellitus Experimental/patología , Femenino , Intolerancia a la Glucosa/complicaciones , Hepatitis/congénito , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Resistencia a la Insulina , Interleucinas/biosíntesis , Interleucinas/sangre , Janus Quinasa 2/biosíntesis , Janus Quinasa 2/efectos de los fármacos , Janus Quinasa 2/genética , Recuento de Linfocitos , Masculino , Ratones , Ratones Endogámicos ICR , Embarazo , Cultivo Primario de Células , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Factor de Transcripción STAT3/biosíntesis , Factor de Transcripción STAT3/efectos de los fármacos , Factor de Transcripción STAT3/genética , Vacuolas/patología
3.
Inflammopharmacology ; 29(4): 1101-1109, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-34218389

RESUMEN

There are accumulating reports regarding poor response to common antidepressant therapy. Antidepressant resistance is often linked to inflammatory system activation and patients displaying inflammation prior to the treatment are less responsive to antidepressants. We hypothesized that the inefficacy of antidepressant therapy in some patients may be attributable to the drugs' inflammatory mode of action, which has been overlooked because of their substantial therapeutic benefit. Bupropion is a commonly prescribed antidepressant that is often used to treat seasonal affective disorders as well. Nevertheless, research suggests that bupropion causes inflammation and worsens depressive symptoms. Therefore, we investigated the impact of bupropion on cytokines of innate and adaptive immunity, as well as immune signaling pathways. We treated lipopolysaccharide (LPS)-stimulated human peripheral blood mononuclear cells (PBMCs) with different doses of bupropion. Pro-/anti-inflammatory cytokines [tumor necrosis factor alpha (TNFα), interleukin-1ß (IL-1ß), IL-17, and IL-10] were assessed at both transcriptional and translational levels as well as the involvement of JAK2 /STAT3, TLR2, and TLR4 signaling in this process. Bupropion reduced IL-17A, TNFα, and IL-1ß protein levels in the cultures. Nonetheless, bupropion increased IL-1ß (P < 0.0001), TNFα (P < 0.0001), and IL-17A (P < 0.05) mRNA levels. Treatment enhanced both IL-10 concentration (P < 0.0001) and gene expression (P < 0.0001). TLR2 (P < 0.0001), TLR4 (P < 0.0001), JAK2 (P < 0.0001), and STAT3 (P < 0.0001) gene expression also rose in response to bupropion. The findings imply that bupropion, particularly 50 µM and 100 µM, has pro-inflammatory effects and should be co-administered with anti-inflammatory medications, at least in patients with inflammatory conditions.


Asunto(s)
Antiinflamatorios/farmacología , Bupropión/farmacología , Janus Quinasa 2/biosíntesis , Factor de Transcripción STAT3/biosíntesis , Receptor Toll-Like 2/biosíntesis , Receptor Toll-Like 4/biosíntesis , Adulto , Células Cultivadas , Relación Dosis-Respuesta a Droga , Humanos , Janus Quinasa 2/inmunología , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/inmunología , Leucocitos Mononucleares/metabolismo , Masculino , Factor de Transcripción STAT3/agonistas , Factor de Transcripción STAT3/inmunología , Receptor Toll-Like 2/agonistas , Receptor Toll-Like 2/inmunología , Receptor Toll-Like 4/agonistas , Receptor Toll-Like 4/inmunología , Adulto Joven
4.
J Neuroinflammation ; 18(1): 150, 2021 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-34225736

RESUMEN

BACKGROUND: Our recent studies have identified that the red nucleus (RN) dual-directionally modulates the development and maintenance of mononeuropathic pain through secreting proinflammatory and anti-inflammatory cytokines. Here, we further explored the action of red nucleus IL-33 in the early development of mononeuropathic pain. METHODS: In this study, male rats with spared nerve injury (SNI) were used as mononeuropathic pain model. Immunohistochemistry, Western blotting, and behavioral testing were used to assess the expressions, cellular distributions, and actions of red nucleus IL-33 and its related downstream signaling molecules. RESULTS: IL-33 and its receptor ST2 were constitutively expressed in the RN in naive rats. After SNI, both IL-33 and ST2 were upregulated significantly at 3 days and peaked at 1 week post-injury, especially in RN neurons, oligodendrocytes, and microglia. Blockade of red nucleus IL-33 with anti-IL-33 neutralizing antibody attenuated SNI-induced mononeuropathic pain, while intrarubral administration of exogenous IL-33 evoked mechanical hypersensitivity in naive rats. Red nucleus IL-33 generated an algesic effect in the early development of SNI-induced mononeuropathic pain through activating NF-κB, ERK, p38 MAPK, and JAK2/STAT3, suppression of NF-κB, ERK, p38 MAPK, and JAK2/STAT3 with corresponding inhibitors markedly attenuated SNI-induced mononeuropathic pain or IL-33-evoked mechanical hypersensitivity in naive rats. Red nucleus IL-33 contributed to SNI-induced mononeuropathic pain by stimulating TNF-α expression, which could be abolished by administration of inhibitors against ERK, p38 MAPK, and JAK2/STAT3, but not NF-κB. CONCLUSIONS: These results suggest that red nucleus IL-33 facilitates the early development of mononeuropathic pain through activating NF-κB, ERK, p38 MAPK, and JAK2/STAT3. IL-33 mediates algesic effect partly by inducing TNF-α through activating ERK, p38 MAPK and JAK2/STAT3.


Asunto(s)
Interleucina-33/biosíntesis , Janus Quinasa 2/biosíntesis , Mononeuropatías/metabolismo , Neuralgia/metabolismo , Núcleo Rojo/metabolismo , Factor de Transcripción STAT3/biosíntesis , Animales , Sistema de Señalización de MAP Quinasas/fisiología , Masculino , Mononeuropatías/patología , Neuralgia/patología , Ratas , Ratas Sprague-Dawley , Núcleo Rojo/patología , Factor de Necrosis Tumoral alfa/biosíntesis , Proteínas Quinasas p38 Activadas por Mitógenos/biosíntesis
5.
Drug Deliv ; 28(1): 1166-1178, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34121564

RESUMEN

CD123 targeting molecules have been widely applied in acute myelocytic leukemia (AML) therapeutics. Although antibodies have been more widely used as targeting molecules, aptamer have unique advantages for CD123 targeting therapy. In this study, we constructed an aptamer hydrogel termed as SSFH which could be precisely cut by Cas9/sgRNA for programmed SS30 release. To construct hydrogel, rolling-circle amplification (RCA) was used to generate hydrogel containing CD123 aptamer SS30 and sgRNA-targeting sequence. After incubation with Cas9/sgRNA, SSFH could lose its gel property and liberated the SS30 aptamer sequence, and released SS30 has been confirmed by gel electrophoresis. In addition, SS30 released from SSFH could inhibit cell proliferation and induce cell apoptosis in vitro. Moreover, SSFH could prolong survival rate and inhibit tumor growth via JAK2/STAT5 signaling pathway in vivo. Additionally, molecular imaging revealed SSFH co-injected with Cas9/sgRNA remained at the injection site longer than free aptamer. Furthermore, once the levels of cytokines were increasing, the complementary sequences of aptamers injection could neutralize SS30 and relieve side effect immediately. This study suggested that CD123 aptamer hydrogel SSFH and Cas9/sgRNA system has strong potential for CD123-positive AML anticancer therapy.


Asunto(s)
Aptámeros de Nucleótidos/administración & dosificación , Aptámeros de Nucleótidos/farmacología , Sistemas CRISPR-Cas , Hidrogeles/química , Subunidad alfa del Receptor de Interleucina-3/administración & dosificación , Subunidad alfa del Receptor de Interleucina-3/metabolismo , Leucemia Mieloide Aguda/tratamiento farmacológico , Animales , Apoptosis , Línea Celular Tumoral , Proliferación Celular , Supervivencia Celular , Química Farmacéutica , Citocinas/efectos de los fármacos , Portadores de Fármacos , Humanos , Janus Quinasa 2/biosíntesis , Ratones , Transducción de Señal/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
6.
J Cutan Med Surg ; 25(2): 157-162, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33174479

RESUMEN

BACKGROUND: The Janus kinase-signal transducer and activator of transcription signaling pathway has been suggested as a promising therapeutic target in vitiligo. However, limited data is available on the cutaneous expression of JAK in vitiligo. AIM: This study is designed to analyze the cutaneous expression patterns of JAK1, 2, and 3 in vitiligo and investigate their relation to the disease clinical parameters. METHODS: This case-control study recruited 24 patients having active vitiligo and 20 age, sex, and skin type-matched healthy volunteers. Skin biopsies were obtained from patients (lesional, perilesional and nonlesional) and controls for assessment of JAK1, 2, and 3 expression using RT-PCR. RESULTS: JAK1 and JAK3 were overexpressed in patients' skin compared to control skin and showed a stepwise pattern of upregulation from control to nonlesional, perilesional and lesional skin. However, JAK3 showed much stronger expression. In contrast JAK2 expression showed no significant difference in any of lesional, perilesional or nonlesional skin compared to control skin. JAK1 and JAK3 expression levels showed no correlation with neither the disease activity nor severity. CONCLUSION: JAK1 and more prominently JAK3 are upregulated in vitiliginous skin and possibly contribute to the pathogenesis of the disease. Accordingly, selective JAK3/1 inhibition may provide a favorable therapeutic opportunity for vitiligo patients.This study is registered on the ClinicalTrials.gov Identifier: NCT03185312.


Asunto(s)
Janus Quinasa 1/biosíntesis , Janus Quinasa 2/biosíntesis , Janus Quinasa 3/biosíntesis , Piel/metabolismo , Vitíligo/metabolismo , Adulto , Estudios de Casos y Controles , Femenino , Humanos , Masculino , Estudios Prospectivos , Vitíligo/diagnóstico , Adulto Joven
7.
Inflammation ; 44(1): 206-216, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-32772240

RESUMEN

Sjögren's syndrome (SS) is a chronic autoimmune disease targeting salivary and lacrimal glands. C-X-C motif chemokine ligand 10 (CXCL10) expression is upregulated in lip salivary glands (LSGs) of primary SS (pSS) patients, and CXCL10 involved in SS pathogenesis via immune-cell accumulation. Moreover, interferon (IFN)-γ enhances CXCL10 production via the Janus kinase (JAK)/signal transducer and activator of transcription (STAT) pathway. We investigated the effects of baricitinib, a selective JAK1/2 inhibitor, on both IFN-γ-induced CXCL10 production and immune-cell chemotaxis. We used immunohistochemical staining to determine the expression levels and localization of JAK1 and JAK2 in LSGs of SS patients (n = 12) and healthy controls (n = 3). We then evaluated the effect of baricitinib in an immortalized normal human salivary gland ductal (NS-SV-DC) cell line. Immunohistochemical analysis of LSGs from pSS patients revealed strong JAK1 and JAK2 expression in ductal and acinar cells, respectively. Baricitinib significantly inhibited IFN-γ-induced CXCL10 expression as well as the protein levels in an immortalized human salivary gland ductal-cell clone in a dose-dependent manner. Additionally, western blot analysis showed that baricitinib suppressed the IFN-γ-induced phosphorylation of STAT1 and STAT3, with a stronger effect observed in the case of STAT1. It also inhibited IFN-γ-mediated chemotaxis of Jurkat T cells. These results suggested that baricitinib suppressed IFN-γ-induced CXCL10 expression and attenuated immune-cell chemotaxis by inhibiting JAK/STAT signaling, suggesting its potential as a therapeutic strategy for pSS.


Asunto(s)
Azetidinas/farmacología , Quimiocina CXCL10/antagonistas & inhibidores , Interferón gamma/farmacología , Janus Quinasa 1/antagonistas & inhibidores , Janus Quinasa 2/antagonistas & inhibidores , Purinas/farmacología , Pirazoles/farmacología , Factor de Transcripción STAT1/antagonistas & inhibidores , Conductos Salivales/metabolismo , Sulfonamidas/farmacología , Azetidinas/uso terapéutico , Línea Celular Transformada , Quimiocina CXCL10/biosíntesis , Femenino , Humanos , Janus Quinasa 1/biosíntesis , Janus Quinasa 2/biosíntesis , Células Jurkat , Purinas/uso terapéutico , Pirazoles/uso terapéutico , Factor de Transcripción STAT1/biosíntesis , Conductos Salivales/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Síndrome de Sjögren/tratamiento farmacológico , Síndrome de Sjögren/metabolismo , Sulfonamidas/uso terapéutico
8.
Can J Cardiol ; 35(11): 1546-1556, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31679624

RESUMEN

BACKGROUND: Endothelial progenitor cell (EPC) therapy has been suggested as a major breakthrough in the treatment of ischemic diseases. However, the molecular mechanism that underlies EPC functional regulation is still unclear. METHODS: We examined the angiogenic capacity of EPCs in a hindlimb ischemia model of wild-type and ClC-3 knockout mice. RESULTS: Mice lacking of ClC-3 exhibited reduced blood flow recovery and neovascularization in ischemic muscles 7 and 14 days after hind limb ischemia. Moreover, compared with wild-type EPCs, the hindlimb blood reperfusion in mice receiving ClC-3 knockout EPCs was significantly impaired, accompanied by reduced EPC homing and retention. In vitro, EPCs derived from ClC-3 knockout mice displayed impaired migratory, adhesive, and angiogenic activity. CXC chemokine receptor 4 (CXCR4) expression was significantly reduced in EPC from ClC-3 knockout mice compared with wild-type. Moreover, the expression and phosphorylation of Janus kinase 2 (JAK-2), a downstream signalling of CXCR4, was also reduced in ClC-3 knockout EPC, indicating that CXCR4/JAK-2 signalling is dysregulated by ClC-3 deficiency. Consistent with this assumption, the migratory capacity of wild-type EPCs was attenuated by either CXCR4 antagonist AMD3100 or JAK-2 inhibitor AG490. More importantly, the impaired migratory capacity of ClC-3 knockout EPCs was rescued by overexpression of CXCR4. CONCLUSIONS: ClC-3 plays a critical role in the angiogenic capacity of EPCs and EPC-mediated neovascularization of ischemic tissues. Disturbance of CXCR4/JAK-2 signalling may contribute to the functional impairment of ClC-3 deficient EPCs. Thus, ClC-3 may be a potential therapeutic target for modulating neovascularization in ischemic diseases.


Asunto(s)
Canales de Cloruro/genética , Regulación de la Expresión Génica , Isquemia/metabolismo , Janus Quinasa 2/genética , Neovascularización Patológica/metabolismo , Receptores CXCR4/genética , Trasplante de Células Madre/métodos , Animales , Western Blotting , Células Cultivadas , Canales de Cloruro/biosíntesis , Canales de Cloruro/deficiencia , Modelos Animales de Enfermedad , Células Progenitoras Endoteliales/citología , Células Progenitoras Endoteliales/metabolismo , Miembro Posterior/irrigación sanguínea , Isquemia/patología , Isquemia/terapia , Janus Quinasa 2/biosíntesis , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Musculares , Neovascularización Patológica/patología , Neovascularización Patológica/terapia , Receptores CXCR4/biosíntesis , Transducción de Señal
9.
Eur Rev Med Pharmacol Sci ; 23(21): 9238-9250, 2019 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-31773675

RESUMEN

OBJECTIVE: We attempted to clarify the regulatory mechanism of UCA1/miR-331-3p/IL6R on cell progression in multiple myeloma (MM). PATIENTS AND METHODS: The expression of UCA1, miR-331-3p, and IL6R in tumor tissues and cells was measured by qRT-PCR. Cell Counting Kit-8 (CCK-8) was conducted to detect cell proliferation, and flow cytometry assay was applied to examine cell apoptosis. Protein expression of L6R, p-JAK2, p-STAT3, c-Myc, CyclinD1, Bcl-2, and Bax was detected by Western blot assay. The interaction among miR-331-3p, UCA1, and IL6R was determined by Luciferase reporter system. Murine xenograft assay was performed to confirm the biological function of UCA1 in vivo. RESULTS: The expression of UCA1 and IL6R was up-regulated, while miR-331-3p was down-regulated in MM tumors and cell lines compared with normal tissues and cells. By calculation, miR-331-3p was correlated with UCA1 or IL6R inversely. In addition, UCA1 knockdown suppressed cell proliferation and promoted apoptosis in vitro and in vivo. Luciferase reporter system confirmed the interaction between miR-331-3p and UCA1 or IL6R. More importantly, UCA1 restored miR-331-3p mediated inhibition of proliferation and promotion on apoptosis of MM cells. Consistently, IL6R rescued UCA1 knockdown caused repression on MM cell growth and elevation on apoptosis. Besides, UCA1 facilitated the activation of the JAK2/STAT3 signaling pathway by enhancing IL6R expression via targeting miR-331-3p. CONCLUSIONS: UCA1 accelerates proliferation and suppresses apoptosis in MM by targeting miR-331-3p/IL6R axis to activate JAK2/STAT3 pathway, providing potential targets for the diagnosis and therapy of MM.


Asunto(s)
Apoptosis/fisiología , Proliferación Celular/fisiología , Janus Quinasa 2/biosíntesis , MicroARNs/biosíntesis , Mieloma Múltiple/fisiopatología , ARN Largo no Codificante/fisiología , Receptores de Interleucina-6/biosíntesis , Factor de Transcripción STAT3/biosíntesis , Línea Celular Tumoral , Ciclina D1/biosíntesis , Regulación hacia Abajo/genética , Regulación Neoplásica de la Expresión Génica/fisiología , Humanos , Mieloma Múltiple/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/biosíntesis , Proteínas Proto-Oncogénicas c-myc/biosíntesis , ARN Largo no Codificante/biosíntesis , Transducción de Señal/fisiología , Transfección , Regulación hacia Arriba/genética , Ensayos Antitumor por Modelo de Xenoinjerto , Proteína X Asociada a bcl-2/biosíntesis
10.
Eur Rev Med Pharmacol Sci ; 23(21): 9286-9294, 2019 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-31773696

RESUMEN

OBJECTIVE: Cytokine signal transduction inhibitor 3 (SOCS3) negatively regulates Janus kinases (JAK) - signal transducer and activator of transcription (STAT) pathway. Bioinformatics analysis revealed a targeted relationship between miR-203 and SOCS3 mRNA. This study investigated the role of miR-203 in ovarian cancer cell proliferation and apoptosis. PATIENTS AND METHODS: Ovarian cancer tissues and adjacent tissues were collected to detect the expression of miR-203 and SOCS3. Ovarian cancer HO8910 cells were divided into miR-NC group, miR-203 inhibitor group, and miR-203 mimic group followed by the analysis of the expression of miR-203 and SOCS3 mRNA by quantitative Reverse Transcription-PCR (qRT-PCR), protein expression of p-JAK2 and p-STAT3 by Western blot, cell apoptosis by flow cytometry, and proliferation by 5-Ethynyl-2'-deoxyuridine (EdU) staining chronologically. RESULTS: Compared with adjacent tissues, miR-203 expression was significantly increased in tumor tissues and SCOS3 mRNA expression was decreased. Compared with those with lower miR-203 expression, the prognosis of patients with higher expression of miR-203 was significantly worse. There was a targeted regulatory relationship between miR-203 and SOCS3 mRNA. Compared with IOSE80 cells, miR-203 expression in HO8910 and SKOV3 cells was increased, and its expressions of SOCS3 mRNA and protein were decreased. Compared with miR-NC group, the transfection of miR-203 inhibitor significantly increased SOCS3 expression, and decreased the expression of p-JAK2 and p-STAT3 protein. We draw the conclusion that miR-203 increased cell apoptosis and decreased cell proliferation. However, opposite results were observed after the transfection of miR-203 mimic. CONCLUSIONS: Abnormal miR-203 and SOCS3 expression are related to the pathogenesis of ovarian cancer. MiR-203 affects the proliferation of JAK-STAT pathway and regulates the proliferation and apoptosis of ovarian cancer cells by targeting the inhibition of SOCS3 expression.


Asunto(s)
Apoptosis/fisiología , Carcinoma Epitelial de Ovario/fisiopatología , Proliferación Celular/fisiología , MicroARNs/fisiología , Neoplasias Ováricas/fisiopatología , Proteína 3 Supresora de la Señalización de Citocinas/fisiología , Carcinoma Epitelial de Ovario/metabolismo , Línea Celular Tumoral , Femenino , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Janus Quinasa 2/biosíntesis , MicroARNs/agonistas , MicroARNs/antagonistas & inhibidores , MicroARNs/biosíntesis , Imitación Molecular/fisiología , Neoplasias Ováricas/metabolismo , Factor de Transcripción STAT3/biosíntesis , Proteína 3 Supresora de la Señalización de Citocinas/biosíntesis , Transfección
11.
PLoS One ; 14(10): e0221635, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31600213

RESUMEN

Aberrant activation of the JAK/STAT pathway is thought to be the critical event in the pathogenesis of the chronic myeloproliferative neoplasms, polycythemia vera, essential thrombocythemia and primary myelofibrosis. The most frequent genetic alteration in these pathologies is the activating JAK2V617F mutation, and expression of the mutant gene in mouse models was shown to cause a phenotype resembling the human diseases. Given the body of genetic evidence, it has come as a sobering finding that JAK inhibitor therapy only modestly suppresses the JAK2V617F allele burden, despite showing clear benefits in terms of reducing splenomegaly and constitutional symptoms in patients. To gain a better understanding if JAK2V617F is required for maintenance of myeloproliferative disease once it has evolved, we generated a conditional inducible transgenic JAK2V617F mouse model using the SCL-tTA-2S tet-off system. Our model corroborates that expression of JAK2V617F in hematopoietic stem and progenitor cells recapitulates key hallmarks of human myeloproliferative neoplasms, and exhibits gender differences in disease manifestation. The disease was found to be transplantable, and importantly, reversible when transgenic JAK2V617F expression was switched off. Our results indicate that mutant JAK2V617F-specific inhibitors should result in profound disease modification by disabling the myeloproliferative clone bearing mutant JAK2.


Asunto(s)
Regulación de la Expresión Génica , Células Madre Hematopoyéticas , Janus Quinasa 2 , Trastornos Mieloproliferativos , Transgenes , Sustitución de Aminoácidos , Animales , Modelos Animales de Enfermedad , Células Madre Hematopoyéticas/metabolismo , Células Madre Hematopoyéticas/patología , Humanos , Janus Quinasa 2/biosíntesis , Janus Quinasa 2/genética , Ratones , Ratones Transgénicos , Mutación Missense , Trastornos Mieloproliferativos/genética , Trastornos Mieloproliferativos/metabolismo , Trastornos Mieloproliferativos/patología
13.
Mol Carcinog ; 58(8): 1512-1525, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31069881

RESUMEN

Lung cancer is a leading cause of cancer-related death worldwide. Cyanopyridines and aminocyanopyridines with carbon-nitrogen bonds have been proved to exert significant anticancer, antibacterial, and anti-inflammatory effects. In this study, we showed that aminocyanopyridine 3o and 3k displaying potent antitumor activity via inhibiting the signal transducer and activator of transcription 3 (STAT3) pathway. They blocked the constitutive STAT3 phosphorylation in a dose- and time-dependent manner and regulated the transcription of STAT3 target genes encoding apoptosis factors. Most importantly, 3o also inhibited interleukin-6-induced STAT3 activation and nuclear localization. Furthermore, 3o significantly inhibited the tumor growth of H460-derived xenografts. Taken together, these findings suggest that 3o and 3k are promising therapeutic drug candidates for lung cancer by inhibiting persistent STAT3 signaling.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias Pulmonares/tratamiento farmacológico , Piridinas/farmacología , Factor de Transcripción STAT3/antagonistas & inhibidores , Células A549 , Animales , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Femenino , Humanos , Interleucina-6/metabolismo , Janus Quinasa 2/biosíntesis , Janus Quinasa 3/biosíntesis , Neoplasias Pulmonares/patología , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Simulación del Acoplamiento Molecular , Fosforilación/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
14.
Acta Haematol ; 141(4): 261-267, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30965317

RESUMEN

BCR-ABL1-negative myeloproliferative disorders and chronic myeloid leukaemia are haematologic malignancies characterised by single and mutually exclusive genetic alterations. Nevertheless, several patients co-expressing the JAK2V617F mutation and the BCR-ABL1 transcript have been described in the literature. We report the case of a 61-year-old male who presented with an essential thrombocythaemia phenotype and had a subsequent diagnosis of chronic phase chronic myeloid leukaemia. Colony-forming assays demonstrated the coexistence of 2 different haematopoietic clones: one was positive for the JAK2V617F mutation and the other co-expressed both JAK2V617F and the BCR-ABL1 fusion gene. No colonies displayed the BCR-ABL1 transcript alone. These findings indicate that the JAK2V617F mutation was the founding genetic alteration of the disease, followed by the acquisition of the BCR-ABL1 chimeric oncogene. Our data support the hypothesis that a heterozygous JAK2V617F clone may have favoured the bi-clonal nature of this myeloproliferative disorder, generating clones harbouring a second transforming genetic event.


Asunto(s)
Proteínas de Fusión bcr-abl , Regulación Enzimológica de la Expresión Génica , Regulación Leucémica de la Expresión Génica , Janus Quinasa 2 , Leucemia Mielógena Crónica BCR-ABL Positiva , Mutación Missense , Trombocitemia Esencial , Sustitución de Aminoácidos , Ensayo de Unidades Formadoras de Colonias , Proteínas de Fusión bcr-abl/biosíntesis , Proteínas de Fusión bcr-abl/genética , Humanos , Janus Quinasa 2/biosíntesis , Janus Quinasa 2/genética , Leucemia Mielógena Crónica BCR-ABL Positiva/enzimología , Leucemia Mielógena Crónica BCR-ABL Positiva/genética , Leucemia Mielógena Crónica BCR-ABL Positiva/patología , Masculino , Persona de Mediana Edad , Trombocitemia Esencial/enzimología , Trombocitemia Esencial/genética , Trombocitemia Esencial/patología
15.
Haematologica ; 104(1): 70-81, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30171023

RESUMEN

Thrombosis is the main cause of morbidity and mortality in patients with JAK2V617F myeloproliferative neoplasms. Recent studies have reported the presence of JAK2V617F in endothelial cells of some patients with myeloproliferative neoplasms. We investigated the role of endothelial cells that express JAK2V617F in thrombus formation using an in vitro model of human endothelial cells overexpressing JAK2V617F and an in vivo model of mice with endothelial-specific JAK2V617F expression. Interestingly, these mice displayed a higher propensity for thrombus. When deciphering the mechanisms by which JAK2V617F-expressing endothelial cells promote thrombosis, we observed that they have a pro-adhesive phenotype associated with increased endothelial P-selectin exposure, secondary to degranulation of Weibel-Palade bodies. We demonstrated that P-selectin blockade was sufficient to reduce the increased propensity of thrombosis. Moreover, treatment with hydroxyurea also reduced thrombosis and decreased the pathological interaction between leukocytes and JAK2V617F-expressing endothelial cells through direct reduction of endothelial P-selectin expression. Taken together, our data provide evidence that JAK2V617F-expressing endothelial cells promote thrombosis through induction of endothelial P-selectin expression, which can be reversed by hydroxyurea. Our findings increase our understanding of thrombosis in patients with myeloproliferative neoplasms, at least those with JAK2V617F-positive endothelial cells, and highlight a new role for hydroxyurea. This novel finding provides the proof of concept that an acquired genetic mutation can affect the pro-thrombotic nature of endothelial cells, suggesting that other mutations in endothelial cells could be causal in thrombotic disorders of unknown cause, which account for 50% of recurrent venous thromboses.


Asunto(s)
Células Endoteliales/metabolismo , Janus Quinasa 2/biosíntesis , Selectina-P/biosíntesis , Trombosis/metabolismo , Animales , Modelos Animales de Enfermedad , Células Endoteliales/patología , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Humanos , Hidroxiurea/farmacología , Janus Quinasa 2/genética , Ratones , Ratones Transgénicos , Selectina-P/genética , Trombosis/tratamiento farmacológico , Trombosis/genética , Trombosis/patología
16.
Eur Rev Med Pharmacol Sci ; 22(22): 7614-7620, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30536300

RESUMEN

OBJECTIVE: The aim of this study was to explore the role of lncRNA ZNF667-AS1 in the recovery of spinal cord injury (SCI), and to investigate its underlying mechanism. MATERIALS AND METHODS: Mice were randomly assigned to the SCI group, the sham group and the lncRNA ZNF667-AS1 group, with 10 mice in each group. With Infinite Horizon device at a dose of 80 Kdyn, mice in the SCI group and the lncRNA ZNF667-AS1 group experienced SCI by an acute hit on the C5 spinous process. Before animal procedures, mice in the lncRNA ZNF667-AS1 group were additionally injected with overexpression lentivirus of lncRNA ZNF667-AS1. On the contrary, mice in the sham group only received laminectomy. After successful construction of the SCI model in mice, grip strength was accessed. LncRNA ZNF667-AS1 expression in spinal cord tissues before and after SCI was detected by quantitative Real Time-Polymerase Chain Reaction (qRT-PCR), respectively. Meanwhile, the protein expression levels of relative genes in Janus Kinase-signal transducer and activator of transcription (JAK-STAT) pathway were detected by Western blot. RESULTS: Grip strength of forelimb in the SCI group recovered significantly slower than that of the sham group. With the prolongation of SCI, the expression of lncRNA ZNF667-AS1 was gradually decreased. However, the expression levels of JAK2, STAT3 and iNOS were upregulated in a time-dependent manner. In addition, mice in the lncRNA ZNF667-AS1 group presented remarkable grip strength recovery of forelimb after SCI. CONCLUSIONS: LncRNA ZNF667-AS expression is gradually downregulated after SCI. Meanwhile, it inhibits the inflammatory response and promotes SCI recovery via suppressing the JAK-STAT pathway.


Asunto(s)
Inflamación/tratamiento farmacológico , Quinasas Janus/efectos de los fármacos , ARN Largo no Codificante/uso terapéutico , Factores de Transcripción STAT/efectos de los fármacos , Traumatismos de la Médula Espinal/tratamiento farmacológico , Animales , Femenino , Vectores Genéticos , Fuerza de la Mano , Inflamación/etiología , Janus Quinasa 2/biosíntesis , Janus Quinasa 2/genética , Lentivirus/genética , Ratones , Ratones Endogámicos C57BL , Óxido Nítrico Sintasa de Tipo II/biosíntesis , Óxido Nítrico Sintasa de Tipo II/genética , ARN Largo no Codificante/genética , Recuperación de la Función , Factor de Transcripción STAT3/biosíntesis , Factor de Transcripción STAT3/genética , Transducción de Señal/efectos de los fármacos , Traumatismos de la Médula Espinal/complicaciones , Traumatismos de la Médula Espinal/fisiopatología
17.
Arthritis Res Ther ; 20(1): 115, 2018 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-29880011

RESUMEN

BACKGROUND: IL-17A has recently emerged as a potential target that regulates the extensive inflammation and abnormal bone formation observed in ankylosing spondylitis (AS). Blocking IL-17A is expected to inhibit bony ankylosis. Here, we investigated the effects of anti IL-17A agents in AS. METHODS: TNFα, IL-17A, and IL-12/23 p40 levels in serum and synovial fluid from patients with ankylosing spondylitis (AS), rheumatoid arthritis (RA), osteoarthritis (OA), or healthy controls (HC) were measured by ELISA. Bone tissue samples were obtained at surgery from the facet joints of ten patients with AS and ten control (Ct) patients with noninflammatory spinal disease. The functional relevance of IL-17A, biological blockades, Janus kinase 2 (JAK2), and non-receptor tyrosine kinase was assessed in vitro with primary bone-derived cells (BdCs) and serum from patients with AS. RESULTS: Basal levels of IL-17A and IL-12/23 p40 in body fluids were elevated in patients with AS. JAK2 was also highly expressed in bone tissue and primary BdCs from patients with AS. Furthermore, addition of exogenous IL-17A to primary Ct-BdCs promoted the osteogenic stimulus-induced increase in ALP activity and mineralization. Intriguingly, blocking IL-17A with serum from patients with AS attenuated ALP activity and mineralization in both Ct and AS-BdCs by inhibiting JAK2 phosphorylation and downregulating osteoblast-involved genes. Moreover, JAK2 inhibitors effectively reduced JAK2-driven ALP activity and JAK2-mediated events. CONCLUSIONS: Our findings indicate that IL-17A regulates osteoblast activity and differentiation via JAK2/STAT3 signaling. They shed light on AS pathogenesis and suggest new rational therapies for clinical AS ankylosis.


Asunto(s)
Diferenciación Celular/fisiología , Interleucina-17/metabolismo , Janus Quinasa 2/biosíntesis , Osteoblastos/metabolismo , Factor de Transcripción STAT3/metabolismo , Espondilitis Anquilosante/metabolismo , Adulto , Anciano , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales/uso terapéutico , Anticuerpos Monoclonales Humanizados , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Femenino , Humanos , Interleucina-17/antagonistas & inhibidores , Janus Quinasa 2/antagonistas & inhibidores , Masculino , Persona de Mediana Edad , Osteoblastos/efectos de los fármacos , Espondilitis Anquilosante/tratamiento farmacológico , Espondilitis Anquilosante/patología
18.
Biomed Res Int ; 2018: 3628121, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29789785

RESUMEN

Nonnutritive sweetener use is a common practice worldwide. Although considered safe for human consumption, accumulating evidence suggests these compounds may affect metabolic homeostasis; however, there is no consensus on the role of frequent sweetener intake in appetite and weight loss. We sought to determine whether frequent intake of commercial sweeteners induces changes in the JAK2/STAT3 signaling pathway in the brain of mice, as it is involved in the regulation of appetite and body composition. We supplemented adult BALB/c mice with sucrose, steviol glycosides (SG), or sucralose, daily, for 6 weeks. After supplementation, we evaluated body composition and expression of total and phosphorylated JAK2, STAT3, and Akt, as well as SOCS3 and ObRb, in brain tissue. Our results show that frequent intake of commercial SG decreases energy intake, adiposity, and weight gain in male animals, while increasing the expression of pJAK2 and pSTAT3 in the brain, whereas sucralose increases weight gain and pJAK2 expression in females. Our results suggest that chronic intake of commercial sweeteners elicits changes in signaling pathways that have been related to the control of appetite and energy balance in vivo, which may have relevant consequences for the nutritional state and long term health of the organism.


Asunto(s)
Encéfalo/metabolismo , Conducta Alimentaria/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Edulcorantes/farmacología , Animales , Femenino , Janus Quinasa 2/biosíntesis , Masculino , Ratones , Ratones Endogámicos BALB C , Proteínas Proto-Oncogénicas c-akt/biosíntesis , Receptores de Leptina/biosíntesis , Factor de Transcripción STAT3/biosíntesis , Proteína 3 Supresora de la Señalización de Citocinas/biosíntesis
19.
J Neurosurg ; 130(3): 977-988, 2018 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-29521586

RESUMEN

OBJECTIVE: Ischemic stroke remains a significant cause of death and disability in industrialized nations. Janus tyrosine kinase (JAK) and signal transducer and activator of transcription (STAT) of the JAK2/STAT3 pathway play important roles in the downstream signal pathway regulation of ischemic stroke-related inflammatory neuronal damage. Recently, microRNAs (miRNAs) have emerged as major regulators in cerebral ischemic injury; therefore, the authors aimed to investigate the underlying molecular mechanism between miRNAs and ischemic stroke, which may provide potential therapeutic targets for ischemic stroke. METHODS: The JAK2- and JAK3-related miRNA (miR-135, miR-216a, and miR-433) expression levels were detected by real-time quantitative reverse-transcriptase polymerase chain reaction (qRT-PCR) and Western blot analysis in both oxygen-glucose deprivation (OGD)-treated primary cultured neuronal cells and mouse brain with middle cerebral artery occlusion (MCAO)-induced ischemic stroke. The miR-135, miR-216a, and miR-433 were determined by bioinformatics analysis that may target JAK2, and miR-216a was further confirmed by 3' untranslated region (3'UTR) dual-luciferase assay. The study further detected cell apoptosis, the level of lactate dehydrogenase, and inflammatory mediators (inducible nitric oxide synthase [iNOS], matrix metalloproteinase-9 [MMP-9], tumor necrosis factor-α [TNF-α], and interleukin-1ß [IL-1ß]) after cells were transfected with miR-NC (miRNA negative control) or miR-216a mimics and subjected to oxygen-glucose deprivation/reoxygenation (OGD/R) damage with 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay, annexin V-FITC/PI, Western blots, and enzyme-linked immunosorbent assay detection. Furthermore, neurological deficit detection and neurological behavior grading were performed to determine the infarction area and neurological deficits. RESULTS: JAK2 showed its highest level while miR-216a showed its lowest level at day 1 after ischemic reperfusion. However, miR-135 and miR-433 had no obvious change during the process. The luciferase assay data further confirmed that miR-216a can directly target the 3'UTR of JAK2, and overexpression of miR-216a repressed JAK2 protein levels in OGD/R-treated neuronal cells as well as in the MCAO model ischemic region. In addition, overexpression of miR-216a mitigated cell apoptosis both in vitro and in vivo, which was consistent with the effect of knockdown of JAK2. Furthermore, the study found that miR-216a obviously inhibited the inflammatory mediators after OGD/R, including inflammatory enzymes (iNOS and MMP-9) and cytokines (TNF-α and IL-1ß). Upregulating miR-216a levels reduced ischemic infarction and improved neurological deficit. CONCLUSIONS: These findings suggest that upregulation of miR-216a, which targets JAK2, could induce neuroprotection against ischemic injury in vitro and in vivo, which provides a potential therapeutic target for ischemic stroke.


Asunto(s)
Apoptosis/genética , Isquemia Encefálica/genética , Regulación de la Expresión Génica/genética , Inflamación/genética , Janus Quinasa 2/biosíntesis , Janus Quinasa 2/genética , MicroARNs/genética , Factor de Transcripción STAT3/genética , Transducción de Señal/genética , Regiones no Traducidas 3'/genética , Animales , Infarto Encefálico/patología , Masculino , Ratones , Enfermedades del Sistema Nervioso/etiología , Enfermedades del Sistema Nervioso/genética , Cultivo Primario de Células , Accidente Cerebrovascular/genética , Regulación hacia Arriba
20.
Histopathology ; 72(2): 259-269, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-28795418

RESUMEN

AIMS: CD274 (PDL1) and JAK2 (9p24.1) gene amplifications have been recently described in pulmonary carcinomas in association with programmed death-ligand 1 (PD-L1) expression. Furthermore, PTEN loss has been explored preclinically in relation to PD-L1 expression. Our aim was to determine whether these genomic alterations affect PD-L1 expression levels in non-small-cell lung cancer. METHODS AND RESULTS: PD-L1 and PTEN expression determined by immunohistochemistry (IHC), and CD274, JAK2 and PTEN copy number alterations (CNAs) determined by fluorescence in-situ hybridisation, were studied in 171 pulmonary carcinoma specimens. PD-L1 expression was positive in 40 cases (23.3%), and CD274 amplification was present in 14 tumours (8.8%). Concordance between both events was found in 12 of 14 amplified cases (P = 0.0001). We found nine JAK2-amplified cases (5.7%), seven with PD-L1 expression (P = 0.0006). Moreover, six of the seven cases had JAK2 and CD274 coamplification (9p24.1 genomic amplification). Remarkably, the average PD-L1 IHC score was higher in these amplified cases (230 versus 80; P = 0.001). Non-statistical associations were observed between PD-L1 expression and PTEN loss and PTEN deletions. CONCLUSIONS: We describe a subset of patients (8.2%) who had 9p24.1 amplifications resulting in high expression of PD-L1. Our results provide evidence for genomic up-regulation of PD-L1 expression in non-small-cell lung cancer.


Asunto(s)
Antígeno B7-H1/genética , Carcinoma de Pulmón de Células no Pequeñas/genética , Regulación Neoplásica de la Expresión Génica/genética , Janus Quinasa 2/genética , Neoplasias Pulmonares/genética , Adulto , Anciano , Anciano de 80 o más Años , Femenino , Amplificación de Genes , Humanos , Janus Quinasa 2/biosíntesis , Masculino , Persona de Mediana Edad , Estudios Retrospectivos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...