Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
BMC Musculoskelet Disord ; 24(1): 711, 2023 Sep 06.
Artigo em Inglês | MEDLINE | ID: mdl-37674188

RESUMO

BACKGROUND: The purpose of this study was to measure the femoral prosthesis flexion angle (FPFA) in total knee arthroplasty (TKA) using three-dimensional reconstruction, and to assess the differences in early clinical efficacy between patients with different degrees of flexion. METHODS: We conducted a prospective cohort study. From June 2019 to May 2021, 113 patients admitted for TKA due to osteoarthritis of the knee were selected. The patients' postoperative knee joints were reconstructed in three dimensions according to postoperative three-dimensional computed tomography (CT) scans. The FPFA was measured, and the patients were divided into 4 groups: anterior extension group (FPFA < 0°), mildly flexed group (0° ≤ FPFA < 3°), moderately flexed group (3° ≤ FPFA < 6°) and excessively flexed group (6° ≤ FPFA). The differences in the Knee Society Score (KSS), knee Range of Motion (ROM), and visual analogue scale (VAS) scores were measured and compared between the four groups at each postoperative time point. RESULTS: Postoperative KSS, ROM, and VAS were significantly improved in all groups compared to the preoperative period. At 1 year postoperatively, the ROM was significantly greater in the mildly flexed group (123.46 ± 6.51°) than in the anterior extension group (116.93 ± 8.05°) and the excessively flexed group (118.76 ± 8.20°) (P < 0.05). The KSS was significantly higher in the mildly flexed group (162.68 ± 12.79) than in the other groups at 6 months postoperatively (P < 0.05). The higher KSS (174.17 ± 11.84) in the mildly flexed group was maintained until 1 year postoperatively, with a statistically significant difference (P < 0.05). No significant difference in VAS scores was observed between groups at each time point. CONCLUSIONS: A femoral prosthesis flexion angle of 0-3° significantly improved postoperative knee mobility, and patients could obtain better Knee Society Scores after surgery, which facilitated the postoperative recovery of knee function. TRIAL REGISTRATION: ChiCTR2100051502, 2021/09/24.


Assuntos
Artroplastia do Joelho , Membros Artificiais , Humanos , Artroplastia do Joelho/efeitos adversos , Estudos Prospectivos , Fêmur/diagnóstico por imagem , Fêmur/cirurgia , Articulação do Joelho/diagnóstico por imagem , Articulação do Joelho/cirurgia
2.
Acta Pharmacol Sin ; 43(9): 2386-2396, 2022 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-35082393

RESUMO

ACT001, derived from traditional herbal medicine, is a novel compound with effective anticancer activity in clinical trials. However, little is known regarding its role in pituitary adenomas. Here, we demonstrated that ACT001 suppressed cell proliferation and induced cell death of pituitary tumor cells in vitro and in vivo. ACT001 was also effective in suppressing the growth of different subtypes of human pituitary adenomas. The cytotoxic mechanism ACT001 employed was mainly related to autophagic cell death (ACD), indicated by autophagosome formation and LC3-II accumulation. In addition, ACT001-mediated inhibitory effect decreased when either ATG7 was downregulated or cells were cotreated with autophagy inhibitor 3-methyladenine (3-MA). RNA-seq analysis showed that mitogen-activated protein kinase (MAPK) pathway was a putative target of ACT001. Specifically, ACT001 treatment promoted the phosphorylation of JNK and P38 by binding to mitogen-activated protein kinase kinase 4 (MEK4). Our study indicated that ACT001-induced ACD of pituitary tumor cells via activating JNK and P38 phosphorylation by binding with MEK4, and it might be a novel and effective anticancer drug for pituitary adenomas.


Assuntos
Antineoplásicos , Morte Celular Autofágica , Neoplasias Hipofisárias , Antineoplásicos/química , Antineoplásicos/farmacologia , Apoptose , Autofagia , Linhagem Celular Tumoral , Furanos , Humanos , MAP Quinase Quinase 4/metabolismo , Sistema de Sinalização das MAP Quinases , Proteínas Quinases Ativadas por Mitógeno/farmacologia , Neoplasias Hipofisárias/tratamento farmacológico
3.
J Clin Endocrinol Metab ; 104(12): 6345-6356, 2019 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-31369093

RESUMO

CONTEXT: Our previous study demonstrated that the expression of long noncoding RNA (lncRNA) H19 was frequently downregulated in human primary pituitary adenomas and negatively correlated with tumor progression. However, the role of exosomal lncRNA H19 in the inhibition of pituitary tumor growth remains unclear. OBJECTIVE: To investigate whether exosomal H19 could be transported across the cell membrane to exert its inhibitory effect on pituitary tumor growth. DESIGN: Empty lentivirus GH3 cells with or without H19 overexpression were used to establish a xenograft model. Isolated exosomes were identified by transmission electron microscopy, nanoparticle tracking, and Western blotting. The expression levels of serum exosomal H19 from 200 healthy subjects and 206 patients with various subtypes of pituitary tumors were detected by ultracentrifugation and quantitative real-time PCR. RESULTS: The growth of distal tumor cells was inhibited by transferring exosomal H19, which could be transported through cell membrane and exert its inhibitory effect. Cabergoline increased H19 expression and played a synergic therapeutic effect with exosomal H19. Exosomal H19 inhibited phosphorylation of the mTORC1 substrate 4E-BP1. Of note, the expression level of exosomal H19 in the patients with all subtypes of pituitary tumors was significantly lower than that in the healthy subjects. The change of plasma exosomal H19 level may be correlated with the prognosis or drug response of the patients. CONCLUSION: Exosomal H19 inhibits the growth of distal pituitary tumors through inhibiting 4E-BP1 phosphorylation. Plasma exosomal H19 may serve as an important biomarker for predicting medical responses of patients with prolactinomas.


Assuntos
Adenoma/prevenção & controle , Biomarcadores Tumorais/genética , Exossomos/metabolismo , Neoplasias Hipofisárias/prevenção & controle , RNA Longo não Codificante/genética , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Adenoma/genética , Adenoma/metabolismo , Adenoma/patologia , Animais , Apoptose , Biomarcadores Tumorais/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Proliferação de Células , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Alvo Mecanístico do Complexo 1 de Rapamicina/genética , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Camundongos , Camundongos Nus , Fosforilação , Neoplasias Hipofisárias/genética , Neoplasias Hipofisárias/metabolismo , Neoplasias Hipofisárias/patologia , Prognóstico , RNA Longo não Codificante/metabolismo , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
4.
Nat Commun ; 9(1): 4624, 2018 11 05.
Artigo em Inglês | MEDLINE | ID: mdl-30397197

RESUMO

Aberrant expression of long noncoding RNA H19 has been associated with tumour progression, but the underlying molecular tumourigenesis mechanisms remain largely unknown. Here, we report that H19 expression is frequently downregulated in human primary pituitary adenomas and is negatively correlated with tumour progression. Consistently, upregulation of H19 expression inhibits pituitary tumour cell proliferation in vitro and tumour growth in vivo. Importantly, we uncover a function of H19, which controls cell/tumour growth through inhibiting function of mTORC1 but not mTORC2. Mechanistically, we show that H19 could block mTORC1-mediated 4E-BP1 phosphorylation without affecting S6K1 activation. At the molecular level, H19 interacted with 4E-BP1 at the TOS motif and competitively inhibited 4E-BP1 binding to Raptor. Finally, we demonstrate that H19 is more effective than cabergoline treatment in the suppression of pituitary tumours. Together, our study uncovered the role of H19-mTOR-4E-BP1 axis in pituitary tumour growth regulation that may be a potential therapeutic target for human pituitary tumours.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Alvo Mecanístico do Complexo 1 de Rapamicina/efeitos dos fármacos , Fosfoproteínas/metabolismo , Neoplasias Hipofisárias/metabolismo , RNA Longo não Codificante/antagonistas & inibidores , RNA Longo não Codificante/metabolismo , Animais , Cabergolina/farmacologia , Carcinogênese , Proteínas de Transporte , Proteínas de Ciclo Celular , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Progressão da Doença , Regulação para Baixo , Feminino , Células HEK293 , Xenoenxertos , Humanos , Alvo Mecanístico do Complexo 2 de Rapamicina/metabolismo , Camundongos , Fosforilação/efeitos dos fármacos , Neoplasias Hipofisárias/tratamento farmacológico , RNA Longo não Codificante/genética , Proteínas Quinases S6 Ribossômicas 70-kDa/metabolismo , Regulação para Cima
5.
J Clin Endocrinol Metab ; 102(10): 3692-3703, 2017 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-28973192

RESUMO

Context: The dopamine agonist cabergoline (CAB) has been used widely in the treatment of prolactinomas and other types of pituitary adenomas, but its clinical use is hampered by intolerance in some patients with prolactinoma and lack of effectiveness in other pituitary tumor types. Chloroquine (CQ) is an old drug widely used to treat malaria. Recent studies, including our own, have revealed that CAB and CQ are involved in induction of autophagy and activation of autophagic cell death. Objective: To test whether CAB and CQ can function cooperatively to suppress growth of pituitary adenomas as well as other cancers. Results: In vitro studies using the rat pituitary tumor cell lines MMQ and GH3, human pituitary tumor cell primary cultures, and several human cancer cell lines showed that CQ enhanced suppression of cell proliferation by CAB. These results were confirmed in in vivo xenograft models in nude mice and estrogen-induced rat prolactinomas. To understand the mechanism of combined CAB and CQ action, we established a low-CAB-dose condition in which CAB was able to induce autophagy but failed to suppress cell growth. Addition of CQ to low-dose CAB blocked normal autophagic cycles and induced apoptosis, evidenced by the further accumulation of p62/caspase-8/LC3-II. Conclusion: The data suggest that combined use of CAB and CQ may increase clinical effectiveness in treatment of human pituitary adenomas, as well as other cancers, making it an attractive option in tumor and cancer therapies.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Cloroquina/administração & dosagem , Ergolinas/administração & dosagem , Neoplasias Hipofisárias/tratamento farmacológico , Prolactinoma/tratamento farmacológico , Animais , Cabergolina , Morte Celular/efeitos dos fármacos , Células Cultivadas , Feminino , Células Hep G2 , Humanos , Camundongos , Camundongos Nus , Neoplasias Hipofisárias/patologia , Prolactinoma/patologia , Ratos , Ratos Endogâmicos F344 , Ensaios Antitumorais Modelo de Xenoenxerto
6.
Autophagy ; 13(8): 1404-1419, 2017 Aug 03.
Artigo em Inglês | MEDLINE | ID: mdl-28613975

RESUMO

Dopamine agonists such as bromocriptine and cabergoline have been successfully used in the treatment of pituitary prolactinomas and other neuroendocrine tumors. However, their therapeutic mechanisms are not fully understood. In this study we demonstrated that DRD5 (dopamine receptor D5) agonists were potent inhibitors of pituitary tumor growth. We further found that DRD5 activation increased production of reactive oxygen species (ROS), inhibited the MTOR pathway, induced macroautophagy/autophagy, and led to autophagic cell death (ACD) in vitro and in vivo. In addition, DRD5 protein was highly expressed in the majority of human pituitary adenomas, and treatment of different human pituitary tumor cell cultures with the DRD5 agonist SKF83959 resulted in growth suppression, and the efficacy was correlated with the expression levels of DRD5 in the tumors. Furthermore, we found that DRD5 was expressed in other human cancer cells such as glioblastomas, colon cancer, and gastric cancer. DRD5 activation in these cell lines suppressed their growth, inhibited MTOR activity, and induced autophagy. Finally, in vivo SKF83959 also inhibited human gastric cancer cell growth in nude mice. Our studies revealed novel mechanisms for the tumor suppressive effects of DRD5 agonists, and suggested a potential use of DRD5 agonists as a novel therapeutic approach in the treatment of different human tumors and cancers.


Assuntos
Autofagia , Neoplasias Hipofisárias/metabolismo , Neoplasias Hipofisárias/patologia , Receptores de Dopamina D5/metabolismo , 2,3,4,5-Tetra-Hidro-7,8-Di-Hidroxi-1-Fenil-1H-3-Benzazepina/análogos & derivados , 2,3,4,5-Tetra-Hidro-7,8-Di-Hidroxi-1-Fenil-1H-3-Benzazepina/farmacologia , Animais , Autofagossomos/efeitos dos fármacos , Autofagossomos/metabolismo , Autofagossomos/ultraestrutura , Autofagia/efeitos dos fármacos , Cabergolina , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Ergolinas/farmacologia , Ergolinas/uso terapêutico , Humanos , Camundongos Nus , Neoplasias Hipofisárias/tratamento farmacológico , Neoplasias Hipofisárias/ultraestrutura , Ratos , Espécies Reativas de Oxigênio/metabolismo , Receptores de Dopamina D5/genética , Superóxido Dismutase/metabolismo , Serina-Treonina Quinases TOR/metabolismo
7.
Int J Endocrinol ; 2016: 8580750, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27999593

RESUMO

Objective. The aim of this study is to observe clinical outcomes after more than ten years of followup in a group of patients with invasive giant prolactinomas (IGPs) treated with dopamine agonists (DAs). Methods. Twenty-five patients met the criteria of IGPs, among which 16 patients primarily received bromocriptine (BRC) and the other nine had undergone unsuccessful microsurgery prior to BRC treatment. Results. After a mean follow-up period of 135.5 ± 4.7 months, the clinical symptoms in all patients improved by different degrees. Tumor volume was decreased by a mean of 98.6%, and the tumors of 19 patients had almost completely disappeared. The mean duration of treatment at maximal doses of BRC was 48.5 months. At the last follow-up visit, nineteen patients had normal PRL levels, and 14 of these patients had received the low-dose BRC treatment (at an average of 2.9 ± 0.3 mg/d). Younger patients < 25 years had a significantly higher rate of persistent hyperprolactinemia after long-term BRC treatment (p = 0.043). Conclusion. DAs are a first-line therapy for IGPs because they can effectively achieve long-term control in both shrinking tumor volume and normalizing the PRL level, and majority of patients need low-dose DA maintenance. Younger patients are prone to persistent hyperprolactinemia despite long-term DA treatment.

8.
Endocrine ; 52(3): 641-51, 2016 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-26662185

RESUMO

Dopamine agonists (DAs) are the first-line treatment of prolactinomas. They function through the dopamine 2 receptor (D2R) in the tumor cells. Endocan, also called endothelial cell-specific molecule-1 (ESM1), has been described as a marker of neoangiogenesis. However, whether ESM1 promotes the resistance of prolactinomas to DA therapy is largely unknown. In our study, 25 patients with prolactinomas were divided into resistant- and sensitive- groups according to the clinical response to bromocriptine. We found that ESM1-microvessel density of resistant prolactinomas was significantly higher than that of sensitive prolactinomas (47.9 ± 11.6, n = 8, vs 13.1 ± 2.8, n = 17, p = 0.0006), indicating that ESM1 was a DA resistance-related gene. Immunostaining showed that ESM1 was expressed in tumor vessels and sporadic tumor cells, and ESM1 was overlapped with the Smooth Muscle Actin (SMA) and von Willebrand Factor (VWF) in the tumor vessels. Silencing of ESM1 markedly suppressed the viability of GH3 and MMQ cells in vitro, and furthermore, significantly increased the sensitivity of GH3 and MMQ cells to DA treatment. Additionally, silencing of ESM1 down-regulated the angiogenesis-associated genes, such as VEGFR2, FGF2, CD34, CD31, VWF, and EGFR. Knockdown of ESM1 decreased endothelial tube formation of HUVECs, and significantly increased the sensitivity of HUVECs to Avastin treatment. Therefore, we first demonstrate that DA resistance-related ESM1 promotes the angiogenesis and tumor cells growth of prolactinomas, suggesting that ESM1 may be a novel therapeutic target for prolactinomas.


Assuntos
Agonistas de Dopamina/uso terapêutico , Resistencia a Medicamentos Antineoplásicos/genética , Proteínas de Neoplasias/fisiologia , Neovascularização Patológica/genética , Neoplasias Hipofisárias/patologia , Prolactinoma/patologia , Proteoglicanas/fisiologia , Adolescente , Adulto , Idoso , Animais , Bromocriptina/farmacologia , Bromocriptina/uso terapêutico , Estudos de Casos e Controles , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/genética , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Células Cultivadas , Agonistas de Dopamina/farmacologia , Feminino , Técnicas de Silenciamento de Genes , Células Endoteliais da Veia Umbilical Humana , Humanos , Masculino , Pessoa de Meia-Idade , Proteínas de Neoplasias/antagonistas & inibidores , Neoplasias Hipofisárias/irrigação sanguínea , Neoplasias Hipofisárias/genética , Prolactinoma/irrigação sanguínea , Prolactinoma/genética , Proteoglicanas/antagonistas & inibidores , Ratos , Adulto Jovem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...