Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
EBioMedicine ; 100: 104967, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38241975

RESUMO

The incidence of cancer has shown a great increase during the past decades and poses tough challenges to cancer treatment. Anti-tumour immunotherapy, represented by immune checkpoint inhibitors (ICIs), possesses favorable remission in unrestricted spectrum of cancer types. However, its efficacy seems to be heterogeneous among accumulating studies. Emerging evidences suggest that gut microbiota can modulate anti-tumour immuno-response and predict clinical prognosis. Therefore, remodeling microbiota characteristics with fecal microbiota transplantation (FMT) may be capable of reinforcing host ICIs performance by regulating immune-tumour cell interactions and altering microbial metabolites, thereby imperceptibly shifting the tumour microenvironment. However, the long-term safety of FMT is under concern, which calls for more rigorous screening. In this review, we examine current experimental and clinical evidences supporting the FMT efficacy in boosting anti-tumour immuno-response and lessening tumour-related complications. Moreover, we discuss the challenges in FMT and propose feasible resolutions, which may offer crucial guidance for future clinical operations.


Assuntos
Microbioma Gastrointestinal , Microbiota , Neoplasias , Humanos , Transplante de Microbiota Fecal , Neoplasias/terapia , Imunoterapia , Microambiente Tumoral
2.
J Adv Res ; 52: 219-232, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37586642

RESUMO

INTRODUCTION: The perturbations of gut microbiota could interact with excessively activated immune responses and play key roles in the etiopathogenesis of ulcerative colitis (UC). Desulfovibrio, the most predominant sulfate reducing bacteria (SRB) resided in the human gut, was observed to overgrow in patients with UC. The interactions between specific gut microbiota and drugs and their impacts on UC treatment have not been demonstrated well. OBJECTIVES: This study aimed to elucidate whether Desulfovibrio vulgaris (D. vulgaris, DSV) and its flagellin could activate nucleotide-binding oligomerization domain-like receptors (NLR) family of apoptosis inhibitory proteins (NAIP) / NLR family caspase activation and recruitment domain-containing protein 4 (NLRC4) inflammasome and promote colitis, and further evaluate the efficacy of eugeniin targeting the interaction interface of D. vulgaris flagellin (DVF) and NAIP to attenuate UC. METHODS: The abundance of DSV and the occurrence of macrophage pyroptosis in human UC tissues were investigated. Colitis in mice was established by dextran sulfate sodium (DSS) and gavaged with DSV or its purified flagellin. NAIP/NLRC4 inflammasome activation and macrophage pyroptosis were evaluated in vivo and in vitro. The effects of eugeniin on blocking the interaction of DVF and NAIP/NLRC4 and relieving colitis were also assessed. RESULTS: The abundance of DSV increased in the feces of patients with UC and was found to be associated with disease activity. DSV and its flagellin facilitated DSS-induced colitis in mice. Mechanistically, RNA sequencing showed that gene expression associated with inflammasome complex and pyroptosis was upregulated after DVF treatment in macrophages. DVF was further demonstrated to induce significant macrophage pyroptosis in vitro, depending on NAIP/NLRC4 inflammasome activation. Furthermore, eugeniin was screened as an inhibitor of the interface between DVF and NAIP and successfully alleviated the proinflammatory effect of DVF in colitis. CONCLUSION: Targeting DVF-induced NAIP/NLRC4 inflammasome activation and macrophage pyroptosis ameliorates UC. This finding is of great significance for exploring the gut microbiota-host interactions in UC development and providing new insights for precise treatment.


Assuntos
Colite Ulcerativa , Desulfovibrio vulgaris , Humanos , Camundongos , Animais , Inflamassomos/metabolismo , Flagelina/metabolismo , Desulfovibrio vulgaris/metabolismo , Colite Ulcerativa/induzido quimicamente , Colite Ulcerativa/tratamento farmacológico , Macrófagos/metabolismo , Proteínas de Ligação ao Cálcio/metabolismo , Proteínas Adaptadoras de Sinalização CARD/metabolismo , Proteína Inibidora de Apoptose Neuronal/metabolismo
3.
Cancer Sci ; 113(2): 459-477, 2022 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-34811848

RESUMO

A high-fat diet (HFD) leads to long-term exposure to gut microbial metabolite secondary bile acids, such as deoxycholic acid (DCA), in the intestine, which is closely linked to colorectal cancer (CRC). Evidence reveals that vasculogenic mimicry (VM) is a critical event for the malignant transformation of cancer. Therefore, this study investigated the crucial roles of DCA in the regulation of VM and the progression of intestinal carcinogenesis. The effects of an HFD on VM formation and epithelial-mesenchymal transition (EMT) in human CRC tissues were investigated. The fecal DCA level was detected in HFD-treated Apcmin/+ mice. Then the effects of DCA on VM formation, EMT, and vascular endothelial growth factor receptor 2 (VEGFR2) signaling were evaluated in vitro and in vivo. Here we demonstrated that compared with a normal diet, an HFD exacerbated VM formation and EMT in CRC patients. An HFD could alter the composition of the gut microbiota and significantly increase the fecal DCA level in Apcmin/+ mice. More importantly, DCA promoted tumor cell proliferation, induced EMT, increased VM formation, and activated VEGFR2, which led to intestinal carcinogenesis. In addition, DCA enhanced the proliferation and migration of HCT-116 cells, and induced EMT process and vitro tube formation. Furthermore, the silence of VEGFR2 reduced DCA-induced EMT, VM formation, and migration. Collectively, our results indicated that microbial metabolite DCA promoted VM formation and EMT through VEGFR2 activation, which further exacerbated intestinal carcinogenesis.


Assuntos
Carcinogênese/patologia , Ácido Desoxicólico/metabolismo , Mucosa Intestinal/patologia , Neovascularização Patológica/patologia , Adulto , Idoso , Animais , Apoptose , Ácidos e Sais Biliares/análise , Movimento Celular , Proliferação de Células , Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/patologia , Dieta Hiperlipídica/efeitos adversos , Transição Epitelial-Mesenquimal , Fezes/química , Fezes/microbiologia , Feminino , Microbioma Gastrointestinal , Células HCT116 , Humanos , Mucosa Intestinal/microbiologia , Masculino , Camundongos , Pessoa de Meia-Idade , Neovascularização Patológica/etiologia , Neovascularização Patológica/microbiologia , Transdução de Sinais , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/genética , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
4.
Front Oncol ; 11: 739648, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34733783

RESUMO

Accumulating evidence from studies in humans and animal models has elucidated that gut microbiota, acting as a complex ecosystem, contributes critically to colorectal cancer (CRC). The potential mechanisms often reported emphasize the vital role of carcinogenic activities of specific pathogens, but in fact, a series of metabolites produced from exogenous dietary substrates or endogenous host compounds occupy a decisive position similarly. Detrimental gut microbiota-derived metabolites such as trimethylamine-N-oxide, secondary bile acids, hydrogen sulfide and N-nitroso compounds could reconstruct the ecological composition and metabolic activity of intestinal microorganisms and formulate a microenvironment that opens susceptibility to carcinogenic stimuli. They are implicated in the occurrence, progression and metastasis of CRC through different mechanisms, including inducing inflammation and DNA damage, activating tumorigenic signaling pathways and regulating tumor immunity. In this review, we mainly summarized the intimate relationship between detrimental gut microbiota-derived metabolites and CRC, and updated the current knowledge about detrimental metabolites in CRC pathogenesis. Then, multiple interventions targeting these metabolites for CRC management were critically reviewed, including diet modulation, probiotics/prebiotics, fecal microbiota transplantation, as well as more precise measures such as engineered bacteria, phage therapy and chemopreventive drugs. A better understanding of the interplay between detrimental microbial metabolites and CRC would hold great promise against CRC.

5.
Oxid Med Cell Longev ; 2021: 3328505, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34804363

RESUMO

Inflammaging refers to chronic, low-grade inflammation during aging, which contributes to the pathogenesis of age-related diseases. Studies have shown that probiotic intervention in the aging stage could delay aging-related disorders. However, whether the application of probiotics in early life could have antiaging effects on offspring was unknown. Here, we investigated the effects of Lactobacillus rhamnosus GG (LGG) colonization in early life on inflammaging of offspring. Pregnant mice with the same conception time were given LGG live bacteria (LC group) or LGG fixed bacteria (NC group) from the 18th day after pregnancy until natural birth. The progeny mice were treated with 107 cfu of live or fixed LGG for 0-5 days after birth, respectively. LGG colonization could be detected in the feces of 3-week offspring. The 16S rRNA sequencing analysis of 3-week-old offspring showed that colonization of LGG in early life could alter the composition and diversity of gut microbiota. Interestingly, the beneficial effects of LGG colonization in early life on the microbiota lasted to 8 months old. The abundance of longevity-related bacteria (Lactobacillus, Bifidobacterium, and Akkermansia muciniphila) increased significantly in the LGG colonization group. In addition, LGG colonization increased the abundance of short-chain fatty acid- (SCFA-) producing bacteria and the production of cecal SCFAs. LGG colonization in early life protected the intestinal barrier, enhanced antioxidant defense, attenuated epithelial cell DNA damage, and inhibited intestinal low-grade inflammation in 8-month-old progeny mice. Mechanically, LGG could upregulate Sirtuin1 (SIRT1)/Adenosine 5'-monophosphate-activated protein kinase (AMPK)/Peroxisome proliferator-activated receptor γ coactivator 1-α (PGC-1α) pathway and repress activation of nuclear factor-kappa B (NF-κB), while the protective effect of LGG was blunted after SIRT1 gene silencing. Together, LGG colonization in early life could ameliorate inflammaging of offspring, which would provide a new strategy for the prevention of age-related diseases.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Inflamação/prevenção & controle , Lacticaseibacillus rhamnosus/fisiologia , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/metabolismo , Probióticos/administração & dosagem , Sirtuína 1/metabolismo , Proteínas Quinases Ativadas por AMP/genética , Animais , Animais Recém-Nascidos , Feminino , Microbioma Gastrointestinal , Inflamação/microbiologia , Inflamação/patologia , Intestinos/efeitos dos fármacos , Intestinos/microbiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , NF-kappa B/genética , NF-kappa B/metabolismo , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/genética , Gravidez , Sirtuína 1/genética
6.
Biochim Biophys Acta Rev Cancer ; 1876(2): 188626, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34520804

RESUMO

The human body harbors a vast array of microbiota that modulates host pathophysiological processes and modifies the risk of diseases including cancer. With the advent of metagenomic sequencing studies, the intratumoural microbiota has been found as a component of the tumor microenvironment, imperceptibly affecting the tumor progression and response to current antitumor treatments. The underlying carcinogenic mechanisms of intratumoural microbiota, mainly including inducing DNA damages, activating oncogenic signaling pathways and suppressing the immune response, differ significantly in varied organs and are not fully understood. Some native or genetically engineered microbial species can specifically accumulate and replicate within tumors to initiate antitumor immunity, which will be conducive to pursue precise cancer therapies. In this review, we summarized the community characteristics and therapeutic potential of intratumoural microbiota across diverse tumor types. It may provide new insights for a better understanding of tumor biology and hint at the significance of manipulating intratumoural microbiota.


Assuntos
Microbiota/imunologia , Neoplasias/imunologia , Microambiente Tumoral/imunologia , Humanos
7.
J Chin Med Assoc ; 83(1): 25-31, 2020 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-31809304

RESUMO

BACKGROUND: We previously confirmed the targeting of high-mobility group box 1 (HMGB1) by miR-25. This project aims to further investigate whether miR-25 improves myocardial ischemia-reperfusion injury (IRI) in vivo by targeting HMGB1. METHODS: A rat model of myocardial IRI was established by the ligation of the left anterior descending coronary artery for 45 minutes followed by 2, 4, or 6 hours reperfusion. The expression of miR-25, HMGB1, and apoptosis-related proteins in the myocardium was determined by quantitative real-time polymerase chain reaction (PCR) and western blotting. The activities of myocardial enzymes and the release of inflammatory cytokines were evaluated by enzyme-linked immunosorbent assay. Evans blue/triphenyltetrazolium chloride double staining was performed to assess infarct size. Myocardial apoptosis was detected by terminal deoxynucleotidyl transferase dUTP nick end labeling staining. RESULTS: MiR-25 expression was significantly downregulated, while HMGB1 was highly expressed at the mRNA and protein levels in myocardial tissues after induction of the IRI model. MiR-25 agomir administration suppressed the expression of HMGB1 in myocardial tissues. Furthermore, administration of both miR-25 agomir and lentivirus-mediated short hairpin RNA (shRNA) interference targeting HMGB1 sh-HMGB1 resulted in reduced serum myocardial enzyme activities, cytokine secretion, and myocardial apoptosis during myocardial IRI. CONCLUSION: MiR-25 mitigated myocardial IRI-induced damage by targeting HMGB1.


Assuntos
Proteína HMGB1/genética , MicroRNAs/fisiologia , Traumatismo por Reperfusão Miocárdica/prevenção & controle , Animais , Apoptose , Citocinas/biossíntese , Modelos Animais de Doenças , Masculino , Miocárdio/patologia , Ratos Sprague-Dawley
8.
World J Clin Cases ; 7(3): 396-404, 2019 Feb 06.
Artigo em Inglês | MEDLINE | ID: mdl-30746382

RESUMO

BACKGROUND: Cardiac resynchronization therapy (CRT) can be used as an escalated therapy to improve heart function in patients with cardiac dysfunction due to long-term right ventricular pacing. However, guidelines are only targeted at adults. CRT is rarely used in children. CASE SUMMARY: This case aimed to implement biventricular pacing in one child with heart failure who had a left ventricular ejection fraction < 35% at 4 years after implantation of an atrioventricular sequential pacemaker due to atrioventricular block. Postoperatively, echocardiography showed atrial sensing ventricular pacing and QRS wave duration of 120-130 ms, and cardiac function significantly improved after upgrading pacemaker. CONCLUSION: Patients whose cardiac function is deteriorated to a level to upgrade to CRT should be upgraded to reverse myocardial remodeling as soon as possible.

10.
J Mater Chem B ; 3(16): 3242-3253, 2015 Apr 28.
Artigo em Inglês | MEDLINE | ID: mdl-32262318

RESUMO

In this work, biodegradable nanoparticles (NPs) were assembled with sodium carboxymethyl cellulose (CMC) and zein to produce zein-CMC NPs. Paclitaxel (PTX) was 95.5% encapsulated at a zein-CMC weight ratio of 1 : 3 and the NPs were spherical with an average particle size of approximately 159.4 nm, with the PTX concentration maintained at 80 µg mL-1. The NPs demonstrated good stability over a broad range of pH ranging from 3.7 to 11.0. The zein-CMC NPs were seen to provide a sustained release of PTX for up to 72 h, which led to an 80% release of the total loaded PTX in vitro. Confocal laser scanning microscopy (CLSM) and flow cytometry studies showed that the zein-CMC NPs could effectively transport encapsulated molecules into both drug-sensitive (HepG2 cells) and drug-resistant cancer cells (MCF-7 cells). Moreover, in vitro viability studies revealed that the PTX-loaded zein-CMC NPs had greater potency than free PTX in the PTX resistant MCF-7 cells at higher concentration. Furthermore, PTX-loaded NPs displayed obvious efficiency in the apoptosis of HepG2 cells. Zein-CMC NPs have shown significant potential as a highly versatile and potent platform for cancer therapy.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA