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.
Gut Microbes ; 14(1): 2127456, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36195972

RESUMO

Severe acute pancreatitis (SAP) is a critical illness characterized by a severe systemic inflammatory response resulting in persistent multiple organ failure and sepsis. The intestinal microbiome is increasingly appreciated to play a crucial role in modulation of AP disease outcome, but limited information is available about the identity and mechanism of action for specific commensal bacteria involved in AP-associated inflammation. Here we show that Bifidobacteria, particularly B. animalis, can protect against AP by regulating pancreatic and systemic inflammation in germ-free (GF) and oral antibiotic-treated (Abx) mouse models. Colonization by B. animalis and administration of its metabolite lactate protected Abx and GF mice from AP by reducing serum amylase concentration, ameliorating pancreatic lesions and improving survival rate after retrograde injection of sodium taurocholate. B. animalis relieved macrophage-associated local and systemic inflammation of AP in a TLR4/MyD88- and NLRP3/Caspase1-dependent manner through its metabolite lactate. Supporting our findings from the mouse study, clinical AP patients exhibited a decreased fecal abundance of Bifidobacteria that was inversely correlated with the severity of systemic inflammatory responses. These results may shed light on the heterogeneity of clinical outcomes and drive the development of more efficacious therapeutic interventions for AP, and potentially for other inflammatory disorders.


Assuntos
Microbioma Gastrointestinal , Pancreatite , Doença Aguda , Amilases/metabolismo , Amilases/uso terapêutico , Animais , Antibacterianos/uso terapêutico , Bifidobacterium/metabolismo , Inflamação/tratamento farmacológico , Inflamação/metabolismo , Ácido Láctico , Camundongos , Fator 88 de Diferenciação Mieloide/metabolismo , Fator 88 de Diferenciação Mieloide/uso terapêutico , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Pancreatite/metabolismo , Ácido Taurocólico , Receptor 4 Toll-Like/metabolismo
2.
Pathol Res Pract ; 239: 154142, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-36242967

RESUMO

Excessive inflammatory response is a prominent pathogenic hallmark of acute lung injury (ALI). Long noncoding RNA (lncRNA) has been recently reported to play a key role in the pathophysiology of many inflammatory disorders, including ALI. Herein, we attempted to explore the role and underlying mechanism of lncRNA MEG3 in the inflammation in ALI. Firstly, an ALI mouse model was generated via intra-tracheal instillation of lipopolysaccharide (LPS), and then, the impact of lncRNA MEG3 on lung tissue damage, pulmonary edema, lung microvascular permeability and pulmonary inflammatory response, as well as the ALI mice survival rate was investigated. LncRNA MEG3 was upregulated in lung tissues, and knockdown of lncRNA MEG3 protected mice from LPS-induced ALI, with significantly reduced lung pathological changes, decreased lung wet/dry (W/D) ratio and lung microvascular permeability, attenuated inflammatory response, along with increased ALI mice survival. Moreover, lncRNA MEG3 could sponge miR-93, negatively regulated its expression, and lncRNA MEG3 overexpression liberated the suppression of TLR4 expression caused by miR-93. Further, functional studies demonstrated that the protective effects of lncRNA MEG3 on excessive inflammatory response may be related to miR-93-mediated modulation of TLR4/MyD88/NF-κB pathway. Collectively, lncRNA MEG3 inhibition blocked TLR4/MyD88/NF-κB pathway to repress the progression of sepsis-induced lung injury via upregulating miR-93, implying that lncRNA MEG3 might be a viable therapeutic target for ALI.


Assuntos
Lesão Pulmonar Aguda , MicroRNAs , RNA Longo não Codificante , Sepse , Animais , Camundongos , Lesão Pulmonar Aguda/genética , Lesão Pulmonar Aguda/prevenção & controle , Lesão Pulmonar Aguda/induzido quimicamente , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Lipopolissacarídeos , MicroRNAs/metabolismo , Fator 88 de Diferenciação Mieloide/metabolismo , Fator 88 de Diferenciação Mieloide/farmacologia , Fator 88 de Diferenciação Mieloide/uso terapêutico , NF-kappa B/metabolismo , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Sepse/complicações , Sepse/genética , Transdução de Sinais , Receptor 4 Toll-Like/metabolismo
3.
Clin Adv Hematol Oncol ; 20(8): 506-515, 2022 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-36125957

RESUMO

The development of high-throughput technologies has allowed us to characterize the molecular landscape of hematologic neoplasms and identify somatic mutations. As a result, we can now use these technologies to screen for and diagnose neoplastic disease, model risk factors for progression, make treatment decisions, track response to treatment, and design clinical trials. Waldenström macroglobulinemia (WM), which is a lymphoplasmacytic lymphoma, serves as a good example of how genomic data collected at the bench can be applied at the bedside. MYD88 L265P and CXCR4 nonsense and frameshift mutations are the most common recurrent variants observed in patients who have WM, with detection rates of 90% and 40%, respectively. Knowing about these mutations has made it possible to develop agents that target the underlying signaling pathways. In this review, we describe the various treatment strategies for WM and detail the genotype of the malignant WM cell.


Assuntos
Linfoma de Células B , Macroglobulinemia de Waldenstrom , Humanos , Linfoma de Células B/genética , Mutação , Fator 88 de Diferenciação Mieloide/genética , Fator 88 de Diferenciação Mieloide/metabolismo , Fator 88 de Diferenciação Mieloide/uso terapêutico , Transdução de Sinais , Macroglobulinemia de Waldenstrom/diagnóstico , Macroglobulinemia de Waldenstrom/tratamento farmacológico , Macroglobulinemia de Waldenstrom/genética
4.
Adv Sci (Weinh) ; 9(31): e2202590, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-36180407

RESUMO

Diabetes manifests as chronic inflammation and leads to the development diabetic cardiomyopathy (DCM). Targeting key proteins in inflammatory signaling may provide new therapy for DCM. In this study, the authors explore the pharmacological effects and mechanisms of Schisandrin B (Sch B), a natural compound with anti-inflammatory activity against DCM. It is shown that Sch B prevents high-level glucose (HG)-induced hypertrophic and fibrotic responses in cultured cardiomyocytes. RNA sequencing and inflammatory qPCR microarray show that Sch B mainly affects myeloid differentiation primary response 88 (MyD88)-dependent inflammatory gene expression in HG-challenged cardiomyocytes. Further studies indicate that Sch B directly binds to and inhibits MyD88 activation, but does not alter MyD88-independent Toll-like receptor signaling in vivo and in vitro. Inhibiting or silencing MyD88 is associated with reduced levels of HG-induced inflammatory cytokines and myocardial injuries in vitro. Treatment of type 1 and type 2 diabetic mice with Sch B protects heart function, reduces myocardial injuries, and decreases secretion of inflammatory cytokines. Cardiomyocyte-specific MyD88 knockout also protects mice against cardiac inflammation and injury in type 1 diabetic mice. In conclusion, these studies show that cardiomyocyte MyD88 plays an apathogenetic role in DCM and Sch B specifically targets MyD88 to reduce inflammatory DCM.


Assuntos
Diabetes Mellitus Experimental , Cardiomiopatias Diabéticas , Animais , Camundongos , Citocinas/metabolismo , Diabetes Mellitus Experimental/tratamento farmacológico , Diabetes Mellitus Experimental/induzido quimicamente , Diabetes Mellitus Experimental/complicações , Cardiomiopatias Diabéticas/tratamento farmacológico , Cardiomiopatias Diabéticas/complicações , Cardiomiopatias Diabéticas/prevenção & controle , Inflamação/tratamento farmacológico , Inflamação/metabolismo , Fator 88 de Diferenciação Mieloide/metabolismo , Fator 88 de Diferenciação Mieloide/uso terapêutico
5.
JCO Glob Oncol ; 8: e2100380, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35939775

RESUMO

PURPOSE: Waldenstrom Macroglobulinemia (WM) is a rare lymphoma with distinct clinical features, and data from Latin American patients are lacking. Therefore, we aim to investigate the clinical, therapy, and outcome patterns of WM in Latin America. METHODS: We retrospectively analyzed patients with WM diagnosed between 1991 and 2019 from 24 centers in seven Latin American countries. The study outcomes were overall survival (OS) and progression-free survival (PFS). RESULTS: We identified 159 cases (median age 67 years, male 62%). Most patients (95%) were symptomatic at diagnosis. The International Prognostic Scoring System for WM (IPSSWM) at diagnosis was available in 141 (89%) patients (high-risk 40%, intermediate-risk 37%, and low-risk 23%). Twenty-seven (17%) patients were tested for MYD88L265P, with 89% (n = 24 of 27) carrying the mutation. First-line and second-line therapies were administered to 142 (89%) and 53 (33%) patients, respectively. Chemoimmunotherapy was the most commonly used first-line (66%) and second-line (45%) approach; only 18 (11%) patients received ibrutinib. With a median follow-up of 69 months, the 5-year OS rate was 81%. In treated patients, the 5-year OS and PFS rates were 78% and 59%, respectively. High-risk IPSSWM at treatment initiation was an independent risk factor for OS (adjusted hazard ratio: 4.73, 95% CI, 1.67 to 13.41, P = .003) and PFS (adjusted hazard ratio: 2.43, 95% CI, 1.31 to 4.50, P = .005). CONCLUSION: In Latin America, the management of WM is heterogeneous, with limited access to molecular testing and novel agents. However, outcomes were similar to those reported internationally. We validated the IPSSWM score as a prognostic factor for OS and PFS. There is an unmet need to improve access to recommended diagnostic approaches and therapies in Latin America.


Assuntos
Macroglobulinemia de Waldenstrom , Idoso , Humanos , América Latina/epidemiologia , Masculino , Mutação , Fator 88 de Diferenciação Mieloide/genética , Fator 88 de Diferenciação Mieloide/uso terapêutico , Estudos Retrospectivos , Macroglobulinemia de Waldenstrom/tratamento farmacológico , Macroglobulinemia de Waldenstrom/terapia
6.
Zhonghua Fu Chan Ke Za Zhi ; 57(2): 117-124, 2022 Feb 25.
Artigo em Chinês | MEDLINE | ID: mdl-35184472

RESUMO

Objective: To explore the expression of long non-coding RNA-myeloid differentiation factor 88 (lnc-MyD88) and its relationship with the prognosis of patients with epithelial ovarian cancer (EOC). Methods: A total of 70 EOC patients who underwent initial cytoreductive surgery and platinum-based drugs combined with paclitaxel for 6 to 8 courses were selected at Sichuan Cancer Hospital from January 2016 to January 2019. The fresh cancer tissue specimens were collected. In addition, 28 fresh normal ovarian tissues from patients who underwent surgery for benign gynecological diseases during the same period were collected as control group. Reverse transcription (RT) and real-time quantitative polymerase chain reaction (qPCR) were used to detect the expression of lnc-MyD88 and myeloid differentiation factor 88 (MyD88) mRNA in EOC tissues and normal ovarian tissues. The correlation between the expression of lnc-MyD88 and MyD88 mRNA in EOC was analyzed by Pearson's correlation coefficient. The relationship between lnc-MyD88 expression and clinicopathological characteristics of patients with EOC was analyzed. Kaplan-Meier method was used to calculate the survival rate of patients. The log-rank test was used for univariate survival analysis, and Cox proportional hazard model was used for multivariate survival analysis. Results: (1) RT-qPCR showed that the relative expression level of lnc-MyD88 and MyD88 mRNA in EOC were 0.009 (0.000-0.049) and 0.001 (0.000-0.006), respectively, which were significantly higher than those of normal ovarian tissues (all P<0.01); Pearson's correlation coefficient showed that the expression of lnc-MyD88 and MyD88 mRNA in EOC was positively correlated (r2=0.610, P<0.01). (2) The high expression rate of lnc-MyD88 in EOC patients with lymph node metastasis, distant metastasis and chemotherapy resistance (71%, 64% and 70%, respectively) were significantly higher than the patients in control group (41%, 40% and 35%, respectively; all P<0.05). There were no statistically significant in the high expression rate of lnc-MyD88 in EOC patients with different ages, pathological types, pathological grades, surgical pathological stages, postoperative residual lesion size, and ascites cancer cells (all P>0.05). (3) Univariate analysis showed that surgical pathological staging, lymph node metastasis, distant metastasis, postoperative residual tumor size, and high expression of lnc-MyD88 and MyD88 mRNA significantly affected the progression-free survival (PFS) and overall survival (OS) of EOC patients (all P<0.05), ascites cancer cells were the risk factors that significantly affected PFS in EOC patients (P=0.040); multivariate analysis showed that surgical pathological staging and high expression of lnc-MyD88 and MyD88 mRNA were independent factors affecting PFS and OS in EOC patients (all P<0.05), the size of residual lesions after surgery was an independent factor affecting PFS in EOC patients (P=0.001). Conclusions: The level of lnc-MyD88 expression in ovarian cancer tissues was significantly increased. Lnc-MyD88, as a molecular marker for the poor prognosis of EOC, is related to the expression of MyD88 in EOC, and may be involved in its expression regulation, thereby affecting the survival and prognosis of EOC patients.


Assuntos
Neoplasias Epiteliais e Glandulares , Neoplasias Ovarianas , RNA Longo não Codificante , Biomarcadores Tumorais/genética , Carcinoma Epitelial do Ovário/patologia , Feminino , Humanos , Estimativa de Kaplan-Meier , Fator 88 de Diferenciação Mieloide/genética , Fator 88 de Diferenciação Mieloide/metabolismo , Fator 88 de Diferenciação Mieloide/uso terapêutico , Neoplasias Epiteliais e Glandulares/genética , Neoplasias Ovarianas/patologia , Prognóstico , RNA Longo não Codificante/genética
7.
J Drug Target ; 30(4): 430-441, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-34747270

RESUMO

Ischaemic stroke remains a leading cause of disability and mortality worldwide and ageing-associated inflammation for the aged patients specifically leads to worse post-stroke blood-brain barrier (BBB) disruption than young subjects. Accordingly, suppression of excessive inflammation can alleviate BBB injury, which provides potential therapeutic treatment for ischaemic stroke of the aged. Prior studies revealed that mesencephalic astrocyte-derived neurotrophic factor (MANF) regulated inflammatory response and alleviated liver injury in ageing. However, it is unclear whether MANF confer similar benefit to BBB of aged mice suffered from ischaemic stroke. Transient cerebral ischaemia induced by middle cerebral artery occlusion (MCAO) was conducted in aged mice (18-20 months old). MANF was injected into the right lateral ventricle 2 h after MCAO. BBB integrity, tight junctional proteins, ultrastructure of microvessels, infarct volume, neurological scores, brain water content, pro-inflammatory cytokines and neutrophil infiltration rate were determined 72 h after MCAO. H2O2-induced senescent bEnd.3 cells were applied in the in vitro study to investigate the possible mechanism. First, we confirmed that ischaemic stroke/reperfusion in senescent condition promoted the over-expression of MANF on brain endothelial cells. Then, MANF supplement could suppress the pro-inflammatory factor production, restore BBB integrity and then alleviate infarct volume, neurological scores, brain water content and neutrophil infiltration rate. In addition, MANF maintained BBB integrity after ischaemic stroke of aged condition dependent on TLR4/MyD88/NF-κB pathway via intervention of pro-inflammatory factors production. In summary, the recognition of MANF in the process of BBB breakdown at aged condition may offer novel therapeutic approaches for ischaemic stroke.


Assuntos
Isquemia Encefálica , AVC Isquêmico , Acidente Vascular Cerebral , Animais , Anti-Inflamatórios/uso terapêutico , Astrócitos , Barreira Hematoencefálica/metabolismo , Isquemia Encefálica/tratamento farmacológico , Células Endoteliais/metabolismo , Peróxido de Hidrogênio/metabolismo , Infarto da Artéria Cerebral Média/tratamento farmacológico , Inflamação/tratamento farmacológico , Inflamação/metabolismo , AVC Isquêmico/tratamento farmacológico , Camundongos , Fator 88 de Diferenciação Mieloide/metabolismo , Fator 88 de Diferenciação Mieloide/uso terapêutico , NF-kappa B/metabolismo , Fatores de Crescimento Neural/metabolismo , Fatores de Crescimento Neural/uso terapêutico , Reperfusão , Acidente Vascular Cerebral/tratamento farmacológico , Receptor 4 Toll-Like/metabolismo , Receptor 4 Toll-Like/uso terapêutico , Água/metabolismo
8.
Cancer Discov ; 7(11): 1306-1319, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-28801306

RESUMO

Adoptive immunotherapy with T cells expressing chimeric antigen receptors (CAR) has had limited success for solid tumors in early-phase clinical studies. We reasoned that introducing into CAR T cells an inducible costimulatory (iCO) molecule consisting of a chemical inducer of dimerization (CID)-binding domain and the MyD88 and CD40 signaling domains would improve and control CAR T-cell activation. In the presence of CID, T cells expressing HER2-CARζ and a MyD88/CD40-based iCO molecule (HER2ζ.iCO T cells) had superior T-cell proliferation, cytokine production, and ability to sequentially kill targets in vitro relative to HER2ζ.iCO T cells without CID and T cells expressing HER2-CAR.CD28ζ. HER2ζ.iCO T cells with CID also significantly improved survival in vivo in two xenograft models. Repeat injections of CID were able to further increase the antitumor activity of HER2ζ.iCO T cells in vivo Thus, expressing MyD88/CD40-based iCO molecules in CAR T cells has the potential to improve the efficacy of CAR T-cell therapy approaches for solid tumors.Significance: Inducible activation of MyD88 and CD40 in CAR T cells with a small-molecule drug not only enhances their effector function, resulting in potent antitumor activity in preclinical solid tumors, but also enables their remote control post infusion. Cancer Discov; 7(11); 1306-19. ©2017 AACR.This article is highlighted in the In This Issue feature, p. 1201.


Assuntos
Antígenos CD40/genética , Imunoterapia Adotiva/métodos , Fator 88 de Diferenciação Mieloide/genética , Neoplasias/terapia , Receptores de Antígenos de Linfócitos T/uso terapêutico , Animais , Antígenos CD40/imunologia , Antígenos CD40/uso terapêutico , Linhagem Celular Tumoral , Proliferação de Células/genética , Regulação Neoplásica da Expressão Gênica/imunologia , Humanos , Camundongos , Fator 88 de Diferenciação Mieloide/imunologia , Fator 88 de Diferenciação Mieloide/uso terapêutico , Neoplasias/genética , Neoplasias/imunologia , Receptor ErbB-2/genética , Receptor ErbB-2/imunologia , Receptores de Antígenos de Linfócitos T/imunologia , Linfócitos T/imunologia , Membro 7 da Superfamília de Receptores de Fatores de Necrose Tumoral/genética , Membro 7 da Superfamília de Receptores de Fatores de Necrose Tumoral/imunologia , Ensaios Antitumorais Modelo de Xenoenxerto
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...