Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Mais filtros










Intervalo de ano de publicação
1.
Exp Neurol ; 372: 114629, 2024 02.
Artigo em Inglês | MEDLINE | ID: mdl-38056583

RESUMO

BACKGROUND: Endoplasmic reticulum (ER) stress causes neuroinflammation and neuronal apoptosis during ischemic stroke progression. This study has investigated the role of ALKBH5 in ER stress during ischemic stroke progression. METHODS: In vivo and in vitro models of ischemic stroke were established by middle cerebral artery occlusion (MCAO) and OGD/R treatment, respectively. Cerebral infarct size was detected using triphenyltetrazolium chloride staining (TTC), and pathological changes were examined using histological staining. The levels of inflammatory factors were analyzed using Enzyme-linked immunosorbent assay. Cell counting kit-8 assay and flow cytometry were used to measure cell viability and apoptosis, respectively. The global m6A level was detected using the commercial kit, and STAT5 mRNA m6A level was determined using methylated RNA binding protein immunoprecipitation (Me-RIP). ALKBH5, YTHDF1, and STAT5 interactions were analyzed using RIP and RNA pull-down assays. RESULTS: ALKBH5 was upregulated in MCAO animals and OGD/R cell models. ALKBH5 knockdown exacerbated ER stress, neuroinflammation, and neuronal apoptosis in brain tissues and neuronal cells. ALKBH5 inhibited STAT5 mRNA stability and expression in an m6A-YTHDF1-dependent manner. STAT5 promoted ER stress by activating the PERK/eIF2/CHOP signaling pathway. Furthermore, STAT5 knockdown reversed the effects of ALKBH5 knockdown on OGD/R-induced ER stress and neuroinflammation in HT22 cells. CONCLUSION: ALKBH5 knockdown exacerbated ischemic stroke by increasing ER stress-dependent neuroinflammation and neuronal apoptosis via the STAT5/PERK/EIF2α/CHOP signaling pathway in an m6A-YTHDF1-dependent manner.


Assuntos
AVC Isquêmico , Acidente Vascular Cerebral , Animais , Doenças Neuroinflamatórias , Fator de Transcrição STAT5/genética , Fator de Transcrição STAT5/metabolismo , Fator de Transcrição STAT5/farmacologia , Acidente Vascular Cerebral/patologia , Infarto da Artéria Cerebral Média/patologia , Transdução de Sinais , Estresse do Retículo Endoplasmático , Apoptose
2.
Drug Dev Res ; 84(8): 1724-1738, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37756467

RESUMO

Signal transducer and activator of transcription 3 (STAT3) and STAT5 are the transcription factors that have been studied extensively in relevance to the development of cancers in humans. Suppression of either STAT3 or STAT5-mediated signaling events has been demonstrated to be effective in inducing cytotoxicity in cancer cells. Herein, new hybrids of triazolyl-indolo-quinoxaline are synthesized and examined for their effect on the activation of STAT3 and STAT5 pathways in gastric cancer (GC) cells. Among the newly synthesized compounds, 2,3-difluoro-6-((1-(3-fluorophenyl)-1H-1,2,3-triazol-5-yl)methyl)-6H-indolo[2,3-b]quinoxaline (DTI) displayed selective cytotoxicity against GC cells over their normal counterpart. Flow cytometric analysis, annexin-V-fluorescein isothiocyanate staining, terminal deoxynucleotidyl transferase dUTP nick end labeling assay, live and dead assay, and caspase activation experiments suggested DTI as a potent inducer of apoptosis. The mechanistic approach revealed that DTI imparts cytotoxicity via downregulating the phosphorylation of STAT3Y705 and STAT5Y694/699 . DTI significantly reduced the nuclear pool of STAT3/STAT5 and reduced the DNA interaction ability of STAT3/STAT5 as evidenced by immunofluorescence and electrophoretic mobility shift assay. Further investigation revealed that inhibitory effects towards STAT proteins were mediated through the suppression of upstream kinases such as JAK1, JAK2, and Src. Treatment of GC cells with pervanadate counteracted the DTI-driven STAT3/STAT5 inhibition suggesting the involvement of tyrosine phosphatase. Upon DTI exposure, there was a significant upregulation in the mRNA and protein expression of PTPεC, which is a negative regulator of the JAK-STAT pathway. Knockdown of PTPεC suppressed the DTI-induced STATs inhibition in GC cells. Taken together, triazolyl-indolo-quinoxaline is presented as a new inhibitor of the STAT3/STAT5 pathway in GC cells.


Assuntos
Transdução de Sinais , Neoplasias Gástricas , Humanos , Fator de Transcrição STAT5/metabolismo , Fator de Transcrição STAT5/farmacologia , Fator de Transcrição STAT3/metabolismo , Proteínas de Ligação a DNA/metabolismo , Transativadores , Regulação para Cima , Quinoxalinas/farmacologia , Janus Quinases/metabolismo , Janus Quinases/farmacologia , Fatores de Transcrição STAT/metabolismo , Fatores de Transcrição STAT/farmacologia , Fosforilação , Apoptose
3.
Oncol Rep ; 50(3)2023 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-37503758

RESUMO

Oxaliplatin (Oxa) is one of the most effective chemotherapeutic drugs used in the treatment of colorectal cancer (CRC). However, the use of this drug is associated with severe side­effects and patients eventually develop resistance to Oxa. In recent years, copper complexes have been extensively investigated as substitutes for platinum­based drugs. Therefore, a number of copper complexes have also been developed for cancer therapy, such as copper (II) complex of salicylate phenanthroline [Cu(sal)(phen)]. In the present study, the antitumor activity and the related molecular mechanisms of Cu(sal)(phen) were examined in CRC cells. As compared with the chemotherapeutic drug, Oxa, Cu(sal)(phen) was more effective in inducing apoptosis and reactive oxygen species (ROS) production, and in decreasing mitochondrial membrane potential in the CRC cell lines, HCT116 and SW480. In addition, the expression of the apoptosis­related proteins, Bcl­2 and survivin, and those of the upstream regulators, p­JAK2 and p­STAT5, were significantly decreased in the two cell lines following treatment with Cu(sal)(phen). Furthermore, the efficacy of the complex against CRC was found to be excellent in an animal model. The results of immunohistochemical analysis revealed that the expression levels of Bcl­2, survivin and Ki­67 in tumor tissues were decreased following Cu(sal)(phen) treatment. The antitumor mechanisms underlying Cu(Sal)(phen) treatment were the induction of ROS generation, the inhibition of the JAK2/STAT5 signaling pathway and the downregulation of the expression of anti­apoptotic proteins, such as Bcl­2 and survivin. On the whole, the findings of the present study indicated that Cu(sal)(phen) effectively inhibited the viability and proliferation of HCT116 and SW480 CRC cells; in the future, the authors aim to conduct further experiments in future studies to provide more evidence that supports the development of Cu(sal)(phen) as a therapeutic agent for CRC.


Assuntos
Antineoplásicos , Neoplasias Colorretais , Animais , Oxaliplatina/farmacologia , Cobre/farmacologia , Cobre/química , Cobre/metabolismo , Survivina/metabolismo , Fenantrolinas/farmacologia , Fenantrolinas/química , Fator de Transcrição STAT5/metabolismo , Fator de Transcrição STAT5/farmacologia , Salicilatos/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Apoptose , Antineoplásicos/farmacologia , Antineoplásicos/química , Proteínas Reguladoras de Apoptose/metabolismo , Neoplasias Colorretais/tratamento farmacológico , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Linhagem Celular Tumoral
4.
Cancer Biol Med ; 20(8)2023 06 27.
Artigo em Inglês | MEDLINE | ID: mdl-37381714

RESUMO

OBJECTIVE: Pancreatic ductal adenocarcinoma (PDAC) is a highly malignant gastrointestinal cancer with a 5-year survival rate of only 9%. Of PDAC patients, 15%-20% are eligible for radical surgery. Gemcitabine is an important chemotherapeutic agent for patients with PDAC; however, the efficacy of gemcitabine is limited due to resistance. Therefore, reducing gemcitabine resistance is essential for improving survival of patients with PDAC. Identifying the key target that determines gemcitabine resistance in PDAC and reversing gemcitabine resistance using target inhibitors in combination with gemcitabine are crucial steps in the quest to improve survival prognosis in patients with PDAC. METHODS: We constructed a human genome-wide CRISPRa/dCas 9 overexpression library in PDAC cell lines to screen key targets of drug resistance based on sgRNA abundance and enrichment. Then, co-IP, ChIP, ChIP-seq, transcriptome sequencing, and qPCR were used to determine the specific mechanism by which phospholipase D1 (PLD1) confers resistance to gemcitabine. RESULTS: PLD1 combines with nucleophosmin 1 (NPM1) and triggers NPM1 nuclear translocation, where NPM1 acts as a transcription factor to upregulate interleukin 7 receptor (IL7R) expression. Upon interleukin 7 (IL-7) binding, IL7R activates the JAK1/STAT5 signaling pathway to increase the expression of the anti-apoptotic protein, BCL-2, and induce gemcitabine resistance. The PLD1 inhibitor, Vu0155069, targets PLD1 to induce apoptosis in gemcitabine-resistant PDAC cells. CONCLUSIONS: PLD1 is an enzyme that has a critical role in PDAC-associated gemcitabine resistance through a non-enzymatic interaction with NPM1, further promoting the downstream JAK1/STAT5/Bcl-2 pathway. Inhibiting any of the participants of this pathway can increase gemcitabine sensitivity.


Assuntos
Adenocarcinoma , Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Humanos , Adenocarcinoma/tratamento farmacológico , Adenocarcinoma/genética , Antimetabólitos Antineoplásicos/farmacologia , Antimetabólitos Antineoplásicos/uso terapêutico , Carcinoma Ductal Pancreático/tratamento farmacológico , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/patologia , Desoxicitidina/farmacologia , Desoxicitidina/uso terapêutico , Resistencia a Medicamentos Antineoplásicos/genética , Gencitabina , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/genética , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Receptores de Interleucina-7/metabolismo , RNA Guia de Sistemas CRISPR-Cas , Fator de Transcrição STAT5/metabolismo , Fator de Transcrição STAT5/farmacologia , Neoplasias Pancreáticas
5.
Biosensors (Basel) ; 13(5)2023 May 11.
Artigo em Inglês | MEDLINE | ID: mdl-37232900

RESUMO

Increased frequency of CD4+CD25+ regulatory T-cells (Treg) has been associated with disease progression in chronic lymphocytic leukemia (CLL). Flow cytometric methods, which allow for the simultaneous analysis of their specific transcription factor Foxp3 and activated STAT proteins, together with proliferation can help to elucidate the signaling mechanisms driving Treg expansion and suppression of FOXP3- conventional CD4+T-cells (Tcon). Herein, we first report a novel approach in which STAT5 phosphorylation (pSTAT5) and proliferation (BrdU-FITC incorporation) could be analyzed specifically in FOXP3+ and FOXP3- responding cells after CD3/CD28 stimulation. The addition of magnetically purified CD4+CD25+ T-cells from healthy donors to cocultured autologous CD4+CD25- T-cells resulted in suppression of Tcon cell cycle progression accompanied by a decrease in pSTAT5. Next, a method using imaging flow cytometry is presented for the detection of cytokine-dependent pSTAT5 nuclear translocation in FOXP3-expressing cells. Finally, we discuss our experimental data obtained by combining Treg pSTAT5 analysis and antigen-specific stimulation with SARS-CoV-2 antigens. Applying these methods on samples from patients revealed Treg responses to antigen-specific stimulation and significantly higher basal pSTAT5 in CLL patients treated with immunochemotherapy. Thus, we speculate that through the use of this pharmacodynamic tool, the efficacy of immunosuppressive drugs and their possible off-target effects can be assessed.


Assuntos
COVID-19 , Leucemia Linfocítica Crônica de Células B , Humanos , Linfócitos T Reguladores/metabolismo , Leucemia Linfocítica Crônica de Células B/metabolismo , Citometria de Fluxo , SARS-CoV-2/metabolismo , Fator de Transcrição STAT5/metabolismo , Fator de Transcrição STAT5/farmacologia , Fatores de Transcrição Forkhead/metabolismo , Fatores de Transcrição Forkhead/farmacologia
6.
J Anim Sci ; 1012023 Jan 03.
Artigo em Inglês | MEDLINE | ID: mdl-36964762

RESUMO

5-Hydroxytryptamine (5-HT) is an amine produced in both the mammary gland and the central nervous system. Tryptophan hydroxylase 1 (TPH1) catalyzes the conversion of 5-hydroxytryptophan (5-HTP) into l-tryptophan, which is then converted into 5-HT by monoamine-oxidase (MAO-A). In the mammary gland, 5-HT has been shown to have a variety of paracrine-autocrine actions, including suppressing lactation, controlling the destiny of mammary epithelial cells, and maintaining calcium homeostasis throughout the transition from pregnancy to lactation. To examine the effects of 5-HT on the composition of colostrum and milk, a total of 30 transition Guan Zhong dairy goats were intramuscularly injected with 5-HTP (1.0 mg/kg) every morning before feeding from 10 d before the projected parturition date to the day of parturition. The average number of days animals received injections was 8.2 ± 3.2 d. 5-HTP treatment increased serum 5-HT concentration from days 5 to 2 relative to parturition (P < 0.05), and decreased the casein concentration of colostrum (P < 0.05). In the in vitro experiment, mammary epithelial cells isolated from three individual goats' mammary glands were separately treated with 200 µM 5-HTP, 30 µM PCPA (the specific inhibitor of TPH1), or 200 µM 5-HTP + 50 µM SB269970 (the selective antagonist of 5-HTR7). The results showed that 200 µM 5-HTP inhibited the expression of ß-casein, downregulated the activity of the JAK2/ STAT5a signaling pathway, and promoted the apoptosis of goat mammary epithelial cells (GMECs) (P < 0.05). When GMECs were treated with 30 µM Four-chloro-dl-phenylalanine (PCPA), a specific inhibitor of 5-HT synthesis, the mRNA expression of STAT5a and the phosphorylated STAT5a protein level were upregulated. The 50 µM SB269970 treatment rescued the effects of 5-HTP on GMECs (P < 0.05). Taken together, the results indicated that 5-HTP exerted an inhibitory effect on ß-casein synthesis and a proapoptotic effect in GMECs via HTR7 and the JAK2/STAT5a axis.


5-Hydroxytryptamine (5-HT), which is produced in both the mammary gland and the central nervous system, is a recognized important regulator of mammary gland homeostasis. Casein is the major protein in the milk of mammals including cows, goats, and humans, and is a crucial source of high-quality amino acids for humans. In this study, prenatal intramuscular injection of 5-hydroxytryptophan (5-HTP), the precursor of 5-HT, not only increased the level of 5-HT in the serum of goats before delivery but also decreased the concentration of casein in colostrum. Furthermore, in goat mammary epithelial cells which are responsible for milk synthesis, it was found that 5-HTP blocked genes and signal pathways related to casein synthesis, and also promoted cell apoptosis. Additional results demonstrated that the type 7 5-HT receptor (HTR7) mediated the impacts of 5-HT, which provided a potential reliable target for improving milk quality.


Assuntos
5-Hidroxitriptofano , Caseínas , Animais , Feminino , Gravidez , 5-Hidroxitriptofano/farmacologia , 5-Hidroxitriptofano/metabolismo , Apoptose , Caseínas/metabolismo , Células Epiteliais/metabolismo , Cabras/genética , Lactação , Glândulas Mamárias Animais/metabolismo , Serotonina/farmacologia , Serotonina/metabolismo , Fator de Transcrição STAT5/genética , Fator de Transcrição STAT5/metabolismo , Fator de Transcrição STAT5/farmacologia , Receptores de Serotonina/metabolismo
7.
J Transl Med ; 21(1): 115, 2023 02 11.
Artigo em Inglês | MEDLINE | ID: mdl-36774517

RESUMO

BACKGROUND: Spermatogenesis associated serine rich 2 like (SPATS2L) was highly expressed in homoharringtonine (HHT) resistant acute myeloid leukemia (AML) cell lines. However, its role is little known in AML. The present study aimed to investigate the function of SPATS2L in AML pathogenesis and elucidate the underlying molecular mechanisms. METHODS: Overall survival (OS), event-free survival (EFS), relapse-free survival (RFS) were used to evaluate the prognostic impact of SPATS2L for AML from TCGA database and ourcohort. ShRNA was used to knockdown the expression of SPATS2L. Apoptosis was assessed by flow cytometry. The changes of proteins were assessed by Western blot(WB). A xenotransplantation mice model was used to evaluate in vivo growth and survival. RNA sequencing was performed to elucidate the molecular mechanisms underlying the role of SPATS2L in AML. RESULTS: SPATS2L expression increased with increasing resistance indexes(RI) in HHT-resistant cell lines we had constructed. Higher SPATS2L expression was observed in intermediate/high-risk patients than in favorable patients. Meanwhile, decreased SPATS2L expression was observed in AML patients achieving complete remission (CR). Multivariate analysis showed high SPATS2L expression was an independent poor predictor of OS, EFS, RFS in AML. SPATS2L knock down (KD) suppressed cell growth, induced apoptosis, and suppressed key proteins of JAK/STAT pathway, such as JAK2, STAT3, STAT5 in AML cells. Inhibiting SPATS2L expression markedly enhanced the pro-apoptotic effects of traditional chemotherapeutics (Ara-c, IDA, and HHT). CONCLUSIONS: High expression of SPATS2L is a poor prognostic factor in AML, and targeting SPATS2L may be a promising therapeutic strategy for AML patients.


Assuntos
Leucemia Mieloide Aguda , Fator de Transcrição STAT5 , Animais , Camundongos , Mepesuccinato de Omacetaxina/farmacologia , Janus Quinases/metabolismo , Janus Quinases/farmacologia , Janus Quinases/uso terapêutico , Leucemia Mieloide Aguda/tratamento farmacológico , Prognóstico , Transdução de Sinais , Fatores de Transcrição STAT/metabolismo , Fatores de Transcrição STAT/farmacologia , Fatores de Transcrição STAT/uso terapêutico , Fator de Transcrição STAT5/genética , Fator de Transcrição STAT5/metabolismo , Fator de Transcrição STAT5/farmacologia , Humanos
8.
J Biomol Struct Dyn ; 41(1): 186-199, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-34842047

RESUMO

Signal Transducer and Activator of Transcription 5 (STAT5) is a transcription factor that plays a key role in neoplasia, triggered by the fusion oncogene BCR-ABL1; it is not only an essential protein for the pathogenesis of chronic myeloid leukemia (CML), but also its overexpression is associated with drug resistance developed toward various generations of Tyrosine Kinase Inhibitors (TKIs); these are still accepted as gold standard therapeutics for the treatment of CML. In this study, it was investigated whether suppression of STAT5 via a "STAT5 inhibitor" Pimozide resulted in any regain of chemosensitivity to third-generation TKI Ponatinib. Accordingly, the experimental work was designed on both parental CML cell line K562WT and its 1 nM Ponatinib-resistant counterpart, indicated as K562-Pon1. Based on the experimental results, Pimozide was more effective in resistant cells compared to wild-type cells for inducing apoptosis and block cell arrest. Combination therapy of Pimozide and Ponatinib demonstrated that STAT5 was a significant protein for regaining chemosensitivity to Ponatinib when its expression was suppressed both at mRNA and protein level. In conclusion, we consider that STAT5 inhibitor Pimozide can be a good alternative or combination therapy with TKIs for patients suffering from chemotherapeutic drug resistance. Communicated by Ramaswamy H. Sarma.


Assuntos
Leucemia Mielogênica Crônica BCR-ABL Positiva , Piridazinas , Humanos , Células K562 , Proteínas de Fusão bcr-abl , Pimozida/farmacologia , Pimozida/uso terapêutico , Fator de Transcrição STAT5/genética , Fator de Transcrição STAT5/metabolismo , Fator de Transcrição STAT5/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Inibidores de Proteínas Quinases/uso terapêutico , Resistencia a Medicamentos Antineoplásicos/genética , Piridazinas/farmacologia , Leucemia Mielogênica Crônica BCR-ABL Positiva/tratamento farmacológico , Leucemia Mielogênica Crônica BCR-ABL Positiva/genética , Proteínas/metabolismo , Apoptose , Arildialquilfosfatase/metabolismo , Arildialquilfosfatase/farmacologia , Arildialquilfosfatase/uso terapêutico
9.
Mol Biol Rep ; 50(1): 319-329, 2023 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-36331743

RESUMO

BACKGROUND: Due to deficiencies in the expression of hormone receptors, such as PR, ER and HER2, it is challenging to treat triple-negative breast cancer, which does not respond to single targeted therapy. Ruxolitinib is a Janus kinase (JAK)1/JAK2 inhibitor. MK-2206 is an allosteric AKT inhibitor. Due to the limited activities of ruxolitinib and MK-2206 for monotherapy, the need for cotreatment with other drugs has emerged. This study is the first to examine the effects of ruxolitinib and MK-2206 cotreatment on apoptosis and JAK2/STAT5 and PI3K/AKT signaling in MDA-MB-231 breast cancer cells. Additionally, this work aimed to decrease the side effects of ruxolitinib and increase its anticancer effects with MK-2206 cotreatment. METHODS AND RESULTS: Cell viability was reduced in a dose- and time-dependent manner after exposure to ruxolitinib, MK-2206 or both for 48 h, as shown by MTT assay. Ruxolitinib had a synergistic antiproliferative effect, as demonstrated by colony formation and wound healing assays. The effects of ruxolitinib, MK-2206 and their combination on apoptosis, as well as PI3K/AKT and JAK/STAT signaling, were examined by western blot analyses. Cotreatment with ruxolitinib and MK-2206 reduced proliferation with the dual inhibition of JAK2/STAT5 and PI3K/AKT signaling by decreasing PI3K, AKT, JAK2, STAT5, Caspase-9, Caspase-7, PARP, c-Myc, and Bcl-2 and increasing P53 and PTEN protein expression. CONCLUSIONS: Our results revealed the roles of P53 and PTEN in the regulation of apoptosis and the PI3K/AKT and JAK2/STAT5 signaling pathways. The dual inhibition of JAK2/STAT5 and PI3K/AKT may reduce metastasis by decreasing tumor cell survival.


Assuntos
Neoplasias da Mama , Proteínas Proto-Oncogênicas c-akt , Humanos , Feminino , Proteínas Proto-Oncogênicas c-akt/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Neoplasias da Mama/tratamento farmacológico , Fator de Transcrição STAT5/metabolismo , Fator de Transcrição STAT5/farmacologia , Proteína Supressora de Tumor p53 , Linhagem Celular Tumoral , Apoptose , Janus Quinase 2/metabolismo , Proliferação de Células
10.
Gen Comp Endocrinol ; 330: 114125, 2023 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-36116484

RESUMO

In this study, we used juvenile rainbow trout to examine the direct effects of selected environmental estrogens (EE), specifically, 17 ß-estradiol (E2), ß-sitosterol (ßS), and 4-n-nonylphenol (NP), on target tissue sensitivity to insulin-like growth factor (IGF) as assessed by expression of IGF receptor type 1 (IGFR1) mRNAs and IGF-1 binding capacity, as well as on the cell signaling pathways through which EE exert their effects. E2 and NP inhibited IGFR1A and IGFR1B mRNA expression in a time- and concentration-related manner in gill and muscle; however, ßS had no effect on expression of IGFR1 mRNAs in either tissue. NP reduced 125I-IGF binding in gill and E2 and NP reduced 125I-IGF in white muscle; ßS had no effect on 125I-IGF binding in either gill or white muscle. Treatment of gill filaments with either E2 or NP rapidly deactivated (via reduced proportion of phosphorylation) JAK2, STAT5, Akt, and ERK; ßS had no effect on the activation state of any cell signaling elements tested. The effects of EE on IGFR mRNA expression in gill were estrogen receptor (ER) dependent as the inhibitory effects were rescued by the ER antagonist, ICI 182,780. All EE tested blocked growth hormone (GH)-stimulated IGFR mRNA expression in gill filaments. GH-stimulated activation of JAK2, STAT5, Akt, and ERK were blocked by E2, ßS, and NP. Lastly, E2 and NP stimulated suppressor of cytokine signaling 2 (SOCS-2) mRNA expression, an effect that also was ER dependent. These results indicate that EE directly reduce the sensitivity of peripheral tissues to IGF by reducing mRNA and functional expression of IGFRs. Such inhibitory actions of EE are mediated, at least in part, by ER-dependent mechanisms that deactivate JAK, STAT, Akt, and ERK and enhance expression of SOCS-2. These findings together with our previous results show that EE retard growth of post-embryonic rainbow trout through widespread direct effects on the GH-IGF system, specifically, by reducing tissue sensitivity to GH, inhibiting IGF production, reducing tissue sensitivity to IGF, and by deactivating post-receptor IGF cell signaling pathways.


Assuntos
Oncorhynchus mykiss , Animais , Oncorhynchus mykiss/metabolismo , Fosforilação , Fator de Transcrição STAT5/metabolismo , Fator de Transcrição STAT5/farmacologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Fator de Crescimento Insulin-Like I/metabolismo , Estrogênios/metabolismo , Hormônio do Crescimento/metabolismo , Receptores de Somatomedina/metabolismo , Transdução de Sinais , RNA Mensageiro/genética
11.
Microb Pathog ; 173(Pt A): 105831, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-36280164

RESUMO

The Th1 cytokines production associated to signal transducer and activator of transcription 4 (STAT4) signaling amplifies the pro-inflammatory response in acute respiratory distress syndrome (ARDS). The anti-inflammatory action of commensal bacteria has been described as a secondary effect dependent on IL-10- secreting Treg cells that can act in organs far from the gut, including the lung. Despite it, no data is showing whether the previous reported anti-inflammatory action of probiotics is associated with its immunomodulatory effect dependent on Treg cells in a murine model of ARDS. Therefore, herein we focused on the short-term pretreatment effect with Lacticaseibacillus rhamnosus (Lr) in STAT4-associated Th1 cytokines as well as in population of IL-10- secreting Treg cells in a murine model of ARDS. Assays were performed in experimental groups divided into control, LPS, and Lr + LPS. Total and differential cells from bronchoalveolar lavage fluid (BALF) were counted through microscopy and the IL-10, IL-12, IL-17, IL-18, IL-22, IL-23, IL-27, IFN-γ, MMP-9, and TIMP were measured by ELISA. The peribronchial neutrophils were assessed using morphometry and for pulmonary edema was measured by Evans blue dye extravasation. The gene expression for STAT4, T-bet, STAT3, RORÉ£t, STAT5, and Foxp3 were measured by Real-Time PCR. Population of IL-10-secreting Treg cells was performed by flow cytometer. Data showed that pretreatment with Lr attenuated the number of inflammatory cells, secretion of both Th1 and Th17 cytokines, expression of STAT4/T-bet and STAT3/RORÉ£t in lung as well as alterations in lung morphometry. Otherwise, Lr was not efficient to restore mRNA expression for STAT5 and Foxp3 expression and population of IL-10-secreting Treg cells. Thus, beneficial effect of short-term pretreatment with Lr in murine model of ARDS is not dependent on an increased immunomodulatory action of IL-10-secreting Treg cells, however the anti-inflammatory effect of Lr has as target the Th1 and Th17 cytokines as well as signaling involving the STAT4/T-bet and STAT3/RORÉ£t.


Assuntos
Lacticaseibacillus rhamnosus , Pneumonia , Síndrome do Desconforto Respiratório , Camundongos , Animais , Citocinas/metabolismo , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/metabolismo , Interleucina-10/metabolismo , Fator de Transcrição STAT5/metabolismo , Fator de Transcrição STAT5/farmacologia , Lipopolissacarídeos/farmacologia , Modelos Animais de Doenças , Células Th17 , Linfócitos T Reguladores , Lacticaseibacillus rhamnosus/metabolismo , Fatores de Transcrição Forkhead/metabolismo , Anti-Inflamatórios/farmacologia , Fator de Transcrição STAT4
12.
J Neuroinflammation ; 19(1): 253, 2022 Oct 10.
Artigo em Inglês | MEDLINE | ID: mdl-36217178

RESUMO

BACKGROUND: The immune system has been implicated in synaptic plasticity, inflammation, and the progression of Alzheimer's disease (AD). However, there were few studies on improving the niche microenvironment of neural stem cells (NSCs) in the brain of AD to promote adult hippocampal neurogenesis (AHN) by regulating the function of non-parenchymal immune cells. METHODS: The lymph nodes of amyloid precursor protein/presenilin 1 (APP/PS1) and 3xTg (APP/PS1/tau) mouse models of AD were treated with photobiomodulation therapy (PBMT) for 10 J/cm2 per day for 1 month (10 min for each day), T lymphocytes isolated from these two AD models were treated with PBMT for 2 J/cm2 (5 min for each time). The NSCs isolated from hippocampus of these two AD models at E14, and the cells were co-cultivated with PBMT-treated T lymphocyte conditioned medium for NSCs differentiation. RESULTS: Our results showed that PBMT treatment could promote AHN and reverse cognitive deficits in AD mouse model. The expression of interferon-γ (IFN-γ) and interleukin-10 (IL-10) was upregulated in the brain of these two AD models after PBMT treated, which was induced by the activation of Janus kinase 2 (JAK2)-mediated signal transducer and activator of transcription 4 (STAT4)/STAT5 signaling pathway in CD4+ T cells. In addition, elevated CD4+ T cell levels and upregulated transforming growth factor-ß1 (TGFß1)/insulin-like growth factors-1 (IGF-1)/brain-derived neurotrophic factor (BDNF) protein expression levels were also detected in the brain. More importantly, co-cultivated the PBMT-treated T lymphocyte conditioned medium with NSCs derived from these two AD models was shown to promote NSCs differentiation, which was reflected in the upregulation of both neuronal class-III ß-tubulin (Tuj1) and postsynaptic density protein 95 (PSD95), but the effects of PBMT was blocked by reactive oxygen species (ROS) scavenger or JAK2 inhibitor. CONCLUSION: Our research suggests that PBMT exerts a beneficial neurogenesis modulatory effect through activating the JAK2/STAT4/STAT5 signaling pathway to promote the expression of IFN-γ/IL-10 in non-parenchymal CD4+ T cells, induction of improvement of brain microenvironmental conditions and alleviation of cognitive deficits in APP/PS1 and 3xTg-AD mouse models.


Assuntos
Doença de Alzheimer , Terapia com Luz de Baixa Intensidade , Doença de Alzheimer/complicações , Doença de Alzheimer/metabolismo , Doença de Alzheimer/terapia , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Linfócitos T CD4-Positivos/metabolismo , Cognição , Meios de Cultivo Condicionados/farmacologia , Modelos Animais de Doenças , Proteína 4 Homóloga a Disks-Large/metabolismo , Fator de Crescimento Insulin-Like I/metabolismo , Interferon gama/metabolismo , Interleucina-10/metabolismo , Janus Quinase 2/metabolismo , Camundongos , Camundongos Transgênicos , Neurogênese/fisiologia , Presenilina-1/genética , Presenilina-1/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Fator de Transcrição STAT4/metabolismo , Fator de Transcrição STAT5/metabolismo , Fator de Transcrição STAT5/farmacologia , Linfócitos T/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Tubulina (Proteína)/metabolismo
13.
Cell Tissue Res ; 389(3): 501-515, 2022 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-35748981

RESUMO

Mammary epithelial cells (MECs) are the only cells capable of synthesizing lactose. During lactation, alveolar MECs secrete lactose through the apical membrane into the alveolar lumen, whereas alveolar tight junctions (TJs) block the leakage of lactose into the basolateral sides of the MECs. However, lactose leaks from the alveolar lumen into the blood plasma in the mastitis and after weaning. This exposes the basolateral membrane of MECs to lactose. The relationship between lactose in blood plasma and milk production has been suggested. The present study determined whether lactose exposure on the basolateral membrane of mouse MECs adversely affects milk production in vitro. Restricted exposure to lactose on the basolateral side of the MECs was performed using a culture model, in which MECs on the cell culture insert exhibit milk production and less-permeable TJs. The results indicated that lactose exposure on the basolateral side inhibited casein and lipid production in the MECs. Interestingly, lactose exposure on the apical side did not show detectable effects on milk production in the MECs. Basolateral lactose exposure also caused the inactivation of STAT5, a primary transcriptional factor for milk production. Furthermore, p38 and JNK were activated by basolateral lactose exposure. The activation of p38 and JNK following anisomycin treatment reduced phosphorylated STAT5, and inhibitors of p38 blocked the reduction of phosphorylated STAT5 by basolateral lactose exposure. These findings suggest that lactose functions as a partial inhibitor for milk production but only when it directly makes contact with the basolateral membrane of MECs.


Assuntos
Glândulas Mamárias Animais , Fator de Transcrição STAT5 , Animais , Células Epiteliais/metabolismo , Feminino , Lactação/metabolismo , Lactose/metabolismo , Lactose/farmacologia , Camundongos , Leite/metabolismo , Fator de Transcrição STAT5/metabolismo , Fator de Transcrição STAT5/farmacologia
14.
Zhongguo Shi Yan Xue Ye Xue Za Zhi ; 30(3): 671-676, 2022 Jun.
Artigo em Chinês | MEDLINE | ID: mdl-35680789

RESUMO

OBJECTIVE: To investigate the effects of paclitaxel, quizartinib and their combination on proliferation, apoptosis and FLT3/STAT5 pathway of human leukemia cell line MV4-11 (FLT3-ITD+). METHODS: MV4-11 cells were treated with paclitaxel and quizartinib at different concentrations for 24 h, 48 h and 72 h, respectively, and then the two drugs were combined at 48 h to compare the inhibition of proliferation, the apoptosis rate was detected by flow cytometry, the expression of FLT3 and STAT5 mRNA was determined by fluorescence quantitative PCR, and the protein expression of FLT3, p-FLT3, STAT5 and p-STAT5 was determined by Western blot. RESULTS: Different combination groups of paclitaxel and quizartinib had synergistic inhibitory effect. The cell survival rate in the combination group was significantly lower than that in the single drug group (P<0.05). The cell apoptosis rate in the combination group was significantly higher than that in the single drug group (P<0.001). The expression of FLT3 mRNA in combination group was significantly higher than that in two single drugs (P<0.01). The expression of STAT5 mRNA in combination group was significantly higher than that in quizartinib group (P<0.001); increased compared with paclitaxel group, but there was no statistical significance. The expression level of p-FLT3、p-STAT5 protein in the combination group was significantly lower than that in the single drug group (P<0.05, P<0.05). CONCLUSION: Paclitaxel combined with quizartinib can synergistically inhibit the proliferation of MV4-11 cell line and promote the apoptosis of MV4-11 cell line by inhibiting the activity of FLT3/STAT5 pathway.


Assuntos
Leucemia Mieloide Aguda , Fator de Transcrição STAT5 , Apoptose , Benzotiazóis , Linhagem Celular Tumoral , Humanos , Leucemia Mieloide Aguda/genética , Paclitaxel/farmacologia , Paclitaxel/uso terapêutico , Compostos de Fenilureia , RNA Mensageiro , Fator de Transcrição STAT5/genética , Fator de Transcrição STAT5/metabolismo , Fator de Transcrição STAT5/farmacologia , Transdução de Sinais , Tirosina Quinase 3 Semelhante a fms
15.
Adv Exp Med Biol ; 1360: 41-54, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35505161

RESUMO

Gadd45a, Gadd45b, and Gadd45g have been implicated in cell cycle arrest, DNA repair, apoptosis, innate immunity, genomic stability, and modulation of normal blood cell development and leukemia. Each of the Gadd45 genes was shown to be regulated independently in myeloid cells in response to cytokine stimulation modulating blood cell survival and differentiation, including maintaining the quiescent stem cell pool. Gadd45a and Gadd45b were also shown to mediate the protective effects from UV in hematopoietic cells by separate signaling pathways involving either p38 activation or JNK inhibition. Furthermore, it was shown that gadd45a methylation in AML is predictive of poor survival. It was also shown that loss of Gadd45b accelerates the development of BCR-ABL driven CML in mice and leads to decreased median survival. The Gadd45b-deficient CML progenitors exhibited increased proliferation and decreased apoptosis, and this was associated with hyper-activation of c-Jun NH2-terminal kinase and Stat5. Moreover, loss of Gadd45a also accelerated the development of BCR-ABL driven CML, and this was associated with enhanced PI3K-AKT-mTOR-4E-BP1 signaling, upregulation of p30C/EBPα expression, and hyper-activation of p38 and Stat5. In human patients with chronic phase CML, gadd45a expression is up-regulated, whereas in accelerated and blast crisis phase patients, gadd45a is downregulated. Collectively, these results provide novel evidence that Gadd45a functions as a suppressor of BCR/ABL driven leukemia and may serve as a unique prognostic marker of CML progression. Thus Gadd45 proteins provide excellent targets for leukemia therapy.


Assuntos
Antígenos de Diferenciação/metabolismo , Proteínas de Ciclo Celular/metabolismo , Leucemia , Fator de Transcrição STAT5 , Animais , Apoptose , Hematopoese/genética , Humanos , Leucemia/genética , Camundongos , Fosfatidilinositol 3-Quinases/metabolismo , Fator de Transcrição STAT5/metabolismo , Fator de Transcrição STAT5/farmacologia
16.
Allergy ; 77(2): 550-558, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34716993

RESUMO

BACKGROUND: MicroRNA (miR)-146a, as an important immune regulatory factor with an anti-inflammatory effect, plays a crucial role in regulatory T-cell (Tregs) differentiation and function in allergic rhinitis (AR). The present study aimed to investigate the regulatory mechanism employed by miR-146a to control Treg differentiation and function in AR. METHODS: Expression of miR-146a and STAT5b in peripheral blood mononuclear cells (PBMCs) and nasal mucosa from patients with AR was detected by qPCR and Western blotting. Tregs were quantified by flow cytometry in miR-146a knockdown or STAT5b knockdown PBMCs. FOXP3, IL-10, and TGF-ß levels were detected by Western blotting or ELISA in miR-146a knockdown or STAT5b overexpressing PBMCs, as well as in STAT5b knockdown PBMCs overexpressing miR-146a. The effect of miR-146a on STAT5b was observed by luciferase assay and knockdown experiments. RESULTS: Levels of miR146a and STAT5b in the nasal mucosa or PBMCs were significantly lower in the AR group than in the control group. There were significantly fewer Tregs in miR-146a knockdown or STAT5b knockdown PBMCs compared to control PBMCs. Expression of FOXP3, IL-10, and TGF-ß was decreased in the miR-146a knockdown group but increased in the STAT5b overexpression group. In contrast, miR-146a overexpression increased the levels of these factors, but knockdown of STAT5b significantly inhibited this effect. Luciferase assay and knockdown experiments showed that miR-146a bound directly to STAT5b. CONCLUSIONS: miR-146a enhances Treg differentiation and function in AR by positively targeting STAT5b.


Assuntos
MicroRNAs , Rinite Alérgica , Diferenciação Celular/genética , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/metabolismo , Humanos , Interleucina-10/genética , Interleucina-10/metabolismo , Leucócitos Mononucleares/metabolismo , MicroRNAs/genética , MicroRNAs/metabolismo , Fator de Transcrição STAT5/genética , Fator de Transcrição STAT5/metabolismo , Fator de Transcrição STAT5/farmacologia , Linfócitos T Reguladores , Fator de Crescimento Transformador beta/metabolismo
17.
Biofactors ; 48(1): 67-85, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-34866251

RESUMO

Growth hormone (GH) exhibited the important biological activities in the mesenchymal stem cell (MSCs). However, the cellular behavior and properties of GH/GHR in MSCs remain unclear. A series of experiments (such as confocal laser scanning microscope [CLSM] and Western-blot) were performed to systematically investigate the cellular behavior of GH/GHR in MSCs, and the results showed that GH/GHR not only internalized into the cytoplasm, but also transported into the cell nuclei of MSCs. Furthermore, we studied the molecular mechanism by which GH/GHR internalized into cell, and the results indicated that clathrin plays more important role in the process of GHR internalization. More importantly, it can be found that nuclear-targeted GHR has the important biological functions in MSCs, which could promote MSCs proliferation. We further revealed the molecular mechanism by which nuclear-localized GHR regulates MSCs proliferation, and found that nuclear-targeted GHR enhanced the phosphorylation of STAT5, and the activated STAT5 initiates the transcription of CyclinD1, after which, the complex of CyclinD1 and CDK4 further phosphorylates Rb, and the activated Rb releases E2F1, the released E2F1 ultimately realizes the biological function of GH promoting cell proliferation. In short, in the current study,we used MSCs as a model to study the cellular behavior and properties of GH/GHR, and found that GH/GHR can internalize into cell cytoplasm and transport into the cell nuclei. Further work showed that nuclear GHR could drive cell proliferation via GHR/STAT5/CyclinD1 signaling pathway. The current research has laid an important foundation for further study on the regulatory effect of GH on MSCs.


Assuntos
Células-Tronco Mesenquimais , Fator de Transcrição STAT5 , Núcleo Celular/genética , Núcleo Celular/metabolismo , Proliferação de Células , Hormônio do Crescimento/genética , Hormônio do Crescimento/metabolismo , Hormônio do Crescimento/farmacologia , Células-Tronco Mesenquimais/metabolismo , Receptores da Somatotropina/genética , Receptores da Somatotropina/metabolismo , Fator de Transcrição STAT5/genética , Fator de Transcrição STAT5/metabolismo , Fator de Transcrição STAT5/farmacologia , Transdução de Sinais
18.
Artigo em Chinês | WPRIM (Pacífico Ocidental) | ID: wpr-939674

RESUMO

OBJECTIVE@#To investigate the effects of paclitaxel, quizartinib and their combination on proliferation, apoptosis and FLT3/STAT5 pathway of human leukemia cell line MV4-11 (FLT3-ITD+).@*METHODS@#MV4-11 cells were treated with paclitaxel and quizartinib at different concentrations for 24 h, 48 h and 72 h, respectively, and then the two drugs were combined at 48 h to compare the inhibition of proliferation, the apoptosis rate was detected by flow cytometry, the expression of FLT3 and STAT5 mRNA was determined by fluorescence quantitative PCR, and the protein expression of FLT3, p-FLT3, STAT5 and p-STAT5 was determined by Western blot.@*RESULTS@#Different combination groups of paclitaxel and quizartinib had synergistic inhibitory effect. The cell survival rate in the combination group was significantly lower than that in the single drug group (P<0.05). The cell apoptosis rate in the combination group was significantly higher than that in the single drug group (P<0.001). The expression of FLT3 mRNA in combination group was significantly higher than that in two single drugs (P<0.01). The expression of STAT5 mRNA in combination group was significantly higher than that in quizartinib group (P<0.001); increased compared with paclitaxel group, but there was no statistical significance. The expression level of p-FLT3、p-STAT5 protein in the combination group was significantly lower than that in the single drug group (P<0.05, P<0.05).@*CONCLUSION@#Paclitaxel combined with quizartinib can synergistically inhibit the proliferation of MV4-11 cell line and promote the apoptosis of MV4-11 cell line by inhibiting the activity of FLT3/STAT5 pathway.


Assuntos
Humanos , Apoptose , Benzotiazóis , Linhagem Celular Tumoral , Leucemia Mieloide Aguda/genética , Paclitaxel/uso terapêutico , Compostos de Fenilureia , RNA Mensageiro , Fator de Transcrição STAT5/farmacologia , Transdução de Sinais , Tirosina Quinase 3 Semelhante a fms
19.
Cell Death Dis ; 11(2): 149, 2020 02 24.
Artigo em Inglês | MEDLINE | ID: mdl-32094348

RESUMO

Osteosarcoma (OS) is the most common primary bone tumor that primarily affects children and adolescents. Studies suggested that dysregulation JAK/STAT signaling promotes the development of OS. Cells treated with pimozide, a STAT5 inhibitor suppressed proliferation and colony formation and induced sub G0/G1 cell cycle arrest and apoptosis. There was a reduction in cyclin D1 and CDK2 expression and Rb phosphorylation, and activation of Caspase-3 and PARP cleavage. In addition, pimozide suppressed the formation of 3-dimensional osteospheres and growth of the cells in the Tumor in a Dish lung organoid system. Furthermore, there was a reduction in expression of cancer stem cell marker proteins DCLK1, CD44, CD133, Oct-4, and ABCG2. More importantly, it was the short form of DCLK1 that was upregulated in osteospheres, which was suppressed in response to pimozide. We further confirmed by flow cytometry a reduction in DCLK1+ cells. Moreover, pimozide inhibits the phosphorylation of STAT5, STAT3, and ERK in OS cells. Molecular docking studies suggest that pimozide interacts with STAT5A and STAT5B with binding energies of -8.4 and -6.4 Kcal/mol, respectively. Binding was confirmed by cellular thermal shift assay. To further understand the role of STAT5, we knocked down the two isoforms using specific siRNAs. While knockdown of the proteins did not affect the cells, knockdown of STAT5B reduced pimozide-induced necrosis and further enhanced late apoptosis. To determine the effect of pimozide on tumor growth in vivo, we administered pimozide intraperitoneally at a dose of 10 mg/kg BW every day for 21 days in mice carrying KHOS/NP tumor xenografts. Pimozide treatment significantly suppressed xenograft growth. Western blot and immunohistochemistry analyses also demonstrated significant inhibition of stem cell marker proteins. Together, these data suggest that pimozide treatment suppresses OS growth by targeting both proliferating cells and stem cells at least in part by inhibiting the STAT5 signaling pathway.


Assuntos
Osteossarcoma/tratamento farmacológico , Pimozida/farmacologia , Fator de Transcrição STAT5/farmacologia , Proteínas Supressoras de Tumor/farmacologia , Animais , Apoptose/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Pontos de Checagem da Fase G1 do Ciclo Celular/efeitos dos fármacos , Humanos , Células-Tronco Neoplásicas/efeitos dos fármacos , Células-Tronco Neoplásicas/metabolismo , Osteossarcoma/metabolismo , Fator de Transcrição STAT5/efeitos dos fármacos , Fator de Transcrição STAT5/metabolismo , Transdução de Sinais/efeitos dos fármacos
20.
Blood Adv ; 2(17): 2199-2213, 2018 09 11.
Artigo em Inglês | MEDLINE | ID: mdl-30185437

RESUMO

T-cell acute lymphoblastic leukemia (T-ALL) constitutes an aggressive subset of ALL, the most frequent childhood malignancy. Whereas interleukin-7 (IL-7) is essential for normal T-cell development, it can also accelerate T-ALL development in vivo and leukemia cell survival and proliferation by activating phosphatidylinositol 3-kinase/protein kinase B/mechanistic target of rapamycin signaling. Here, we investigated whether STAT5 could also mediate IL-7 T-ALL-promoting effects. We show that IL-7 induces STAT pathway activation in T-ALL cells and that STAT5 inactivation prevents IL-7-mediated T-ALL cell viability, growth, and proliferation. At the molecular level, STAT5 is required for IL-7-induced downregulation of p27kip1 and upregulation of the transferrin receptor, CD71. Surprisingly, STAT5 inhibition does not significantly affect IL-7-mediated Bcl-2 upregulation, suggesting that, contrary to normal T-cells, STAT5 promotes leukemia cell survival through a Bcl-2-independent mechanism. STAT5 chromatin immunoprecipitation sequencing and RNA sequencing reveal a diverse IL-7-driven STAT5-dependent transcriptional program in T-ALL cells, which includes BCL6 inactivation by alternative transcription and upregulation of the oncogenic serine/threonine kinase PIM1 Pharmacological inhibition of PIM1 abrogates IL-7-mediated proliferation on T-ALL cells, indicating that strategies involving the use of PIM kinase small-molecule inhibitors may have therapeutic potential against a majority of leukemias that rely on IL-7 receptor (IL-7R) signaling. Overall, our results demonstrate that STAT5, in part by upregulating PIM1 activity, plays a major role in mediating the leukemia-promoting effects of IL-7/IL-7R.


Assuntos
Interleucina-7/farmacologia , Leucemia-Linfoma Linfoblástico de Células T Precursoras/patologia , Fator de Transcrição STAT5/farmacologia , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Regulação Leucêmica da Expressão Gênica , Humanos , Leucemia-Linfoma Linfoblástico de Células T Precursoras/induzido quimicamente , Leucemia-Linfoma Linfoblástico de Células T Precursoras/metabolismo , Proteínas Proto-Oncogênicas c-pim-1/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-pim-1/metabolismo , Fator de Transcrição STAT5/metabolismo , Transcrição Gênica , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...