Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
BMC Cancer ; 24(1): 754, 2024 Jun 21.
Artigo em Inglês | MEDLINE | ID: mdl-38907215

RESUMO

PURPOSE: Diffuse midline glioma (DMG), H3 K27M-mutant is a type of diffuse high-grade glioma that occurs in the brain midline carrying an extremely poor prognosis under the best efforts of surgery, radiation, and other therapies. For better therapy, we explored the efficacy and toxicity of a novel therapy that combines apatinib and temozolomide in DMG. METHODS: A retrospective analysis of 32 patients with DMG who underwent apatinib plus temozolomide treatment was performed. Apatinib was given 500 mg in adults, 250 mg in pediatric patients once daily. Temozolomide was administered at 200 mg/m2/d according to the standard 5/28 days regimen. The main clinical data included basic information of patients, radiological and pathological characteristics of tumors, treatment, adverse reactions, prognosis. RESULTS: The objective response rate was 24.1%, and the disease control rate was 79.3%. The median PFS of all patients was 5.8 months, and median OS was 10.3 months. A total of 236 cycles of treatment were available for safety assessment and the toxicity of the combination therapy was relatively well tolerated. The most common grade 3 toxicities were myelosuppression including leukopenia (5.08%), neutropenia (4.24%), lymphopenia (2.12%), thrombocytopenia (1.69%) and anemia (1.27%). Grade 4 toxicities included neutropenia (2.12%), thrombocytopenia (2.12%) and proteinuria (1.69%). All the adverse events were relieved after symptomatic treatment or dose reduction. CONCLUSIONS: Apatinib plus temozolomide could be an effective regimen with manageable toxicities and favorable efficacy and may outperform temozolomide monotherapy, particularly in newly diagnosed adults with tumors located outside the pons. The novel therapy deserves further investigation in adult DMG patients.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica , Neoplasias Encefálicas , Glioma , Piridinas , Temozolomida , Humanos , Temozolomida/administração & dosagem , Temozolomida/uso terapêutico , Temozolomida/efeitos adversos , Feminino , Masculino , Adulto , Piridinas/administração & dosagem , Piridinas/efeitos adversos , Piridinas/uso terapêutico , Glioma/tratamento farmacológico , Glioma/patologia , Adolescente , Estudos Retrospectivos , Criança , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/patologia , Adulto Jovem , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Pré-Escolar , Pessoa de Meia-Idade , Resultado do Tratamento
2.
BMC Cancer ; 20(1): 24, 2020 Jan 08.
Artigo em Inglês | MEDLINE | ID: mdl-31914946

RESUMO

BACKGROUND: The optimal chemotherapeutics of recurrent disseminated glioblastoma has yet to be determined. We analyzed the efficacy and safety of recombinant human endostatin (rh-ES) combined with temozolomide and irinotecan in patients with recurrent disseminated glioblastoma. METHODS: We retrospectively reviewed 30 adult patients with recurrent disseminated glioblastoma treated with this combination chemotherapy at Department of Neuro-Oncology, Sanbo Brain Hospital, Capital Medical University of China from November 2009 to August 2018. Temozolomide was given orally at 200 mg/m2 daily for 5 days and rh-ES was administrated 15 mg/d daily for 14 days of each 28-day treatment cycle. Irinotecan was given intravenously every 2 weeks on a 28-day cycle at 340 mg/m2 or 125 mg/m2 depending on antiepileptic drugs. Primary endpoint was progression-free survival (PFS) at 6 months (6 m-PFS). RESULTS: The 6 m-PFS was 23.3%. The median PFS was 3.2 months. The overall survival rate (OS) at 12 months was 28.6%. The median OS was 6.9 months. Six out of 30 (20%) patients demonstrated partial radiographic response and 11 (36.7%) remained stable. The PFS of the 6 patients who got partial response was 5.8, 6.3, 6.9, 13.6, 15.8 and 16.6 months, respectively, and the median time interval of first response was 4 (range, 2.0-6.6) months. The most common adverse events were hematologic toxicities and gastrointestinal effects. The Grade ≥ 3 adverse event was hematologic toxicities. The adverse events were manageable. CONCLUSIONS: Rh-ES, in combination with cytotoxic drugs, was an alternative effective regimen with manageable toxicities in treatment of recurrent disseminated glioblastoma.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Glioblastoma/diagnóstico , Glioblastoma/terapia , Adulto , Antineoplásicos/administração & dosagem , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Endostatinas/administração & dosagem , Feminino , Glioblastoma/mortalidade , Humanos , Estimativa de Kaplan-Meier , Imageamento por Ressonância Magnética , Masculino , Pessoa de Meia-Idade , Recidiva Local de Neoplasia , Estadiamento de Neoplasias , Proteínas Recombinantes/administração & dosagem , Estudos Retrospectivos , Resultado do Tratamento , Adulto Jovem
4.
Acta Biochim Biophys Sin (Shanghai) ; 51(2): 150-158, 2019 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-30590418

RESUMO

Monocyte chemoattractant protein-induced protein 1 (MCPIP1) is a kind of zinc finger RNA binding protein, which exerts immune responses in a variety of cell types. However, the role of MCPIP1 in bovine mammary epithelial cells during mastitis has not been studied. In this study, we explored the functions of MCPIP1 in the inflammatory process induced by virulence factors of pathogens in bovine mammary alveolar cell-T (MAC-T) cell line. Our results showed that MCPIP1 was significantly highly expressed both in the mammary tissue of dairy cows with mastitis and in inflammatory MAC-T cells induced by lipopolysaccharide (LPS) or lipoteichoic acid (LTA). Furthermore, we found that overexpression of MCPIP1 in MAC-T cells abated the LPS-induced increase at the gene expression levels of inflammatory mediators tumor necrosis factor-α-α, interleukin (IL)-1ß, IL-6 and IL-8, enhanced the LPS- and LTA-induced inhibition of epithelial proliferation and promoted the LPS- and LTA-induced oxidative and DNA damage. These findings indicated that MCPIP1 has an enormous potential in regulating the inflammatory response of bovine mammary epithelial cells during infection and may provide an effective therapeutic target for bovine mastitis to reduce the damage caused by inflammatory reactions.


Assuntos
Citocinas/imunologia , Células Epiteliais/imunologia , Mediadores da Inflamação/imunologia , Mastite Bovina/imunologia , Proteínas de Ligação a RNA/metabolismo , Dedos de Zinco , Animais , Bovinos , Linhagem Celular , Proliferação de Células/genética , Citocinas/genética , Citocinas/metabolismo , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/imunologia , Mediadores da Inflamação/metabolismo , Lipopolissacarídeos , Glândulas Mamárias Animais/citologia , Mastite Bovina/genética , Mastite Bovina/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Estresse Oxidativo/imunologia , Proteínas de Ligação a RNA/genética , Ácidos Teicoicos
5.
J Cell Biochem ; 119(11): 9028-9041, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30011095

RESUMO

Bovine mastitis occurs frequently in dairy cows and is often caused by various aetiological organisms, for example, Escherichia coli. Lipopolysaccharide (LPS) is a key virulence factor of E. coli. In this study, we stimulated bovine mammary epithelial cells (BMECs) with LPS to investigate the global transcriptional response and identify specific proinflammatory factors that play important roles in blood-milk barrier damage during mastitis caused by E. coli. By performing RNA-seq, we identified a large number of significantly differentially expressed genes (DEGs) between the LPS-treated BMECs and the control cells. Among the DEGs, interleukin-1ß (IL-1ß) was selected because its messenger RNA expression was induced by LPS and its enrichment is involved in multiple inflammatory signal pathways, and its roles in blood-milk barrier damage during the process of mastitis were investigated. Exogenous IL-1ß treatment damaged the integrity of the blood-milk barrier, as indicated by the increased BMEC tight junction (TJ) permeability and confirmed by in vitro and in vivo experiments. Furthermore, the IL-1ß-induced increase in the BMEC TJ permeability was mediated by the IL-1ß-ERK1/2-MLCK axis pathway. Our data provide insights into the functions of IL-1ß in blood-milk barrier damage caused by mastitis in dairy cows.


Assuntos
Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Interleucina-1beta/farmacologia , Glândulas Mamárias Animais/citologia , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Junções Íntimas/metabolismo , Animais , Western Blotting , Bovinos , Proliferação de Células/efeitos dos fármacos , Claudina-1/metabolismo , Feminino , Camundongos , Ocludina/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Junções Íntimas/efeitos dos fármacos , Proteína da Zônula de Oclusão-1/metabolismo
6.
Mediators Inflamm ; 2018: 4093285, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29686530

RESUMO

The expression of cytochrome P4501A1 (CYP1A1) enzyme is changed in various organs during the host response to inflammation or infection, leading to alterations in the metabolism of endogenous and exogenous compounds. Results of this study showed that CYP1A1 expression was significantly downregulated in the mammary tissue of bovine with mastitis, in inflammatory epithelial cells (INEs) extracted from the tissue, and in lipopolysaccharide- (LPS-) induced INEs compared with their corresponding counterparts. Overexpression of CYP1A1 in bovine mammary epithelial cells alleviated the LPS-induced inhibition of epithelial proliferation, abated the LPS-induced increase of gene expression and protein secretion of inflammatory cytokine tumor necrosis factor-α and interleukin-6, and attenuated the LPS-induced activation of NF-κB signaling. These findings suggest that CYP1A1 has immense potential in the regulation of inflammatory responses in bovine mammary epithelial cells during mastitis and may serve as a useful therapeutic target in mitigating injuries caused by inflammatory overreaction.


Assuntos
Citocromo P-450 CYP1A1/metabolismo , Células Epiteliais/metabolismo , Inflamação/induzido quimicamente , Inflamação/metabolismo , Lipopolissacarídeos/toxicidade , Glândulas Mamárias Animais/citologia , Animais , Bovinos , Linhagem Celular , Citocromo P-450 CYP1A1/genética , Células Epiteliais/efeitos dos fármacos , Feminino , Inflamação/imunologia , Interleucina-6/metabolismo , Mastite/imunologia , Mastite/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Transdução de Sinais/efeitos dos fármacos , Fator de Necrose Tumoral alfa/metabolismo
7.
Cell Physiol Biochem ; 43(1): 82-93, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28848180

RESUMO

BACKGROUND/AIMS: Transforming growth factor-ß1 (TGF-ß1) plays a crucial role in chronic inflammation in various tissues, and is related to inflammation-caused organ fibrogenesis associated with the epithelial-mesenchymal transition (EMT) and the deposition of the extracellular matrix (ECM). However, the effect of TGF-ß1 on bovine mammary epithelial cells (BMECs) with mastitis, and its mechanism, remain unknown. METHODS: We analyzed the level of TGF-ß1 in inflamed mammary tissues and cells using western blotting. BMECs were treated with TGF-ß1, and EMT-related gene and protein expression changes were evaluated using quantitative real-time polymerase chain reaction (qPCR), western blotting, and immunofluorescence. We also inhibited the TGF/Smad signaling pathway using a receptor inhibitor, and analyzed EMT-related protein expression by western blotting. In addition, we injected TGF-ß1 into mice mammary glands to investigate whether it can cause mammary fibrosis in vivo. RESULTS: The TGF-ß1 level was up-regulated in mammary tissues with mastitis and in inducible inflammatory BMECs. TGF-ß1 treatment activated the TGF/ Smad signaling pathway in BMECs during their transition to the EMT phenotype, as indicated by morphological changes from a cobblestone-like shape to a spindle-like one. TGF-ß1 treatment also up-regulated the expression of α-smooth muscle actin, vimentin, and collagen I, albumin, and down-regulated the expression of E-cadherin both in mRNA level and protein level. Furthermore, TGF-ß1 enhanced the gene expressions of MMP2, MMP7, and fibronectin in BMECs. TGF-ß1 injection induced mice mammary infection and fibrosis. CONCLUSION: These findings suggested that aberrant up-regulation of TGF-ß1 in bovine mastitic mammary glands might play an important role in bovine mammary fibrosis caused by unresolved inflammation.


Assuntos
Transição Epitelial-Mesenquimal/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Proteínas Smad/metabolismo , Fator de Crescimento Transformador beta1/farmacologia , Actinas/metabolismo , Animais , Caderinas/metabolismo , Bovinos , Células Cultivadas , Células Epiteliais/citologia , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Feminino , Glândulas Mamárias Animais/citologia , Metaloproteinase 2 da Matriz/genética , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 7 da Matriz/genética , Metaloproteinase 7 da Matriz/metabolismo , Camundongos , Fosforilação/efeitos dos fármacos , Pirazóis/farmacologia , Pirróis/farmacologia , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Fator de Crescimento Transformador beta1/genética , Fator de Crescimento Transformador beta1/metabolismo , Regulação para Cima/efeitos dos fármacos
8.
Front Oncol ; 10: 601175, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33634023

RESUMO

BACKGROUND: Treatment for recurrent glioblastoma is poor, and there is a need for better therapies. Here we retrospectively assessed the efficacy and toxicity of temozolomide plus apatinib, an oral small-molecule tyrosine kinase inhibitor targeting vascular endothelial growth factor receptor 2 in recurrent glioblastoma. MATERIALS AND METHODS: A retrospective analysis of patients with recurrent glioblastoma who underwent apatinib plus temozolomide treatment was performed. Apatinib was given at 500 mg once daily. Temozolomide was administered at 200 mg/m2/d on days 1-5 or 50 mg/m2/d continuous daily according to whether they had experienced temozolomide maintenance treatment before. The main clinical data collected included tumor characteristics, status of MGMT promoter, and IDH mutation, number of relapse, response, survival, adverse reactions, and salvage therapies. RESULTS: From April 2016 to August 2019, thirty-one patients were identified. The objective response rate was 26.3%, and the disease control rate was 84.2%. The progression-free survival (PFS) at 6 months and overall survival (OS) at 12 months were 44.6 and 30.2%. The median PFS and OS were 4.9 and 8.2 months, respectively. Two patients achieved long PFS of 30.9 and 38.7+ months. The median survival time after progression of the patients with or without salvage bevacizumab was 5.1 versus 1.2 months. The most common grade 3 or 4 toxicities were hypertension (5.8%), decreased appetite (5.8%), and thrombocytopenia (4.3%), most of which were resolved after symptomatic treatment or dose reduction. CONCLUSION: Apatinib plus temozolomide is an effective salvage regimen with manageable toxicities for recurrent glioblastoma and could not reduce the sensitivity to bevacizumab.

9.
PeerJ ; 7: e6715, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30984483

RESUMO

The function of long non-coding RNA H19 (H19) on cell proliferation has been observed in various cell types, and the increased expression of H19 was also found in the lipopolysaccharide (LPS)-induced inflammatory bovine mammary epithelial cells (MAC-T). However, the roles of H19 in the inflammatory response and physiological functions of bovine mammary epithelial cell are not clear. In the present study, we found that overexpression of H19 in MAC-T cells significantly promoted cell proliferation, increased the protein and mRNA level of ß-casein, and enhanced the expression of tight junction (TJ)-related proteins while inhibited staphylococcus aureus adhesion to cells. In addition, results demonstrated that overexpression of H19 affected the LPS-induced immune response of MAC-T cells by promoting expressions of inflammatory factors, including TNF-α, IL-6, CXCL2 and CCL5, and activating the NF-κB signal pathway. Our findings indicate that H19 is likely to play an important role in maintaining normal functions and regulating immune response of bovine mammary epithelial cells.

10.
PeerJ ; 5: e3950, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29062612

RESUMO

Increased levels of long noncoding RNA H19 (H19) have been observed in many inflammatory and organ fibrosis diseases including ulcerative colitis, osteoarthritis, liver fibrosis, renal fibrosis and pulmonary fibrosis. However, the role of H19 in bovine mastitis and mastitis-caused fibrosis is still unclear. In our study, H19 was characterized as a novel regulator of EMT induced by transforming growth factor-ß1 (TGF-ß1) in bovine mammary alveolar cell-T (MAC-T) cell line. We found that H19 was highly expressed in bovine mastitis tissues and inflammatory MAC-T cells induced by virulence factors of pathogens. TGF-ß1 was also highly expressed in inflammatory MAC-T cells, and exogenous TGF-ß1 could induce EMT, enhance extracellular matrix protein expression, and upregulate H19 expression in epithelial cells. Stable expression of H19 significantly promotes EMT progression and expression of ECM protein induced by TGF-ß1 in MAC-T cells. Furthermore, by using a specific inhibitor of the PI3K/AKT pathway, we demonstrated that TGF-ß1 upregulated H19 expression through PI3K/AKT pathway. All these observations imply that the lncRNA H19 modulated TGF-ß1-induced epithelial to mesenchymal transition in bovine epithelial cells through PI3K/AKT signaling pathway, which suggests that mammary epithelial cells might be one source for myofibroblasts in vivo in the mammary glands under an inflammatory condition, thereby contributing to mammary gland fibrosis.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA