Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Life Sci ; 334: 122258, 2023 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-37949209

RESUMO

AIMS AND OBJECTIVES: The aim of this study is to discuss the influence of endotoxin on insulin amyloid formation, to provide guidance for therapeutic insulin preparation and storage. MATERIALS AND METHODS: The ThT and ANS binding assays were applied to characterize the dynamics curve of insulin amyloid formation with the presence or absence of endotoxin. The morphological structures of intermediate and mature insulin fibrils were observed with SEM and TEM. Secondary structural changes of insulin during fibriliation were examined with CD, FTIR and Raman spectral analysis. The cytotoxic effects of oligomeric and amyloidogenic insulin aggregates were detected using a cck-8 cell viability assay kit. The influence of endotoxin on insulin efficacy was analyzed by monitoring the activation of insulin signal transduction. KEY FINDINGS: ThT analysis showed that endotoxin, regardless of species, accelerated insulin fibrils formation in a dose-dependent manner, as observed with a shorter lag phase. ANS binding assay demonstrated endotoxin provoked the exposure of insulin hydrophobic patches. The results of SEM and TEM data displayed that endotoxin drove insulin to cluster into dense and viscous form, with thicker and stronger filaments. Based on CD, FTIR and Raman spectra, endotoxin promoted the transition of α-helix to random coil and ß-strand secondary structures during insulin aggregation. Insulins in both oligomeric and amyloidogenic forms were cytotoxic to HepG2 cells, with the former being more severe. Finally, the efficacy of endotoxin treated insulin obviously decreased. SIGNIFICANCE: Our studies revealed that endotoxin disrupts the structural integrity of insulin and promotes its amyloidosis. These findings offered theoretical guidance for insulin storage and safe utilization, as well as pointing up a new direction for insulin resistance research.


Assuntos
Amiloidose , Insulina , Humanos , Amiloide/química , Amiloidose/metabolismo , Insulina/metabolismo , Estrutura Secundária de Proteína , Transdução de Sinais , Endotoxinas
2.
Cell Death Dis ; 14(8): 499, 2023 08 04.
Artigo em Inglês | MEDLINE | ID: mdl-37542062

RESUMO

Pancreatic cancer is a highly aggressive cancer, and is primarily treated with gemcitabine, with increasing resistance. SIRT6 as a member of sirtuin family plays important roles in lifespan and diverse diseases, such as cancer, diabetes, inflammation and neurodegenerative diseases. Considering the role of SIRT6 in the cytoprotective effect, it might be a potential anticancer drug target, and is associated with resistance to anticancer therapy. However, very few SIRT6 inhibitors have been reported. Here, we reported the discovery of a pyrrole-pyridinimidazole derivative, 8a, as a new non-competitive SIRT6 inhibitor, and studied its roles and mechanisms in the antitumor activity and sensitization of pancreatic cancer to gemcitabine. Firstly, we found a potent SIRT6 inhibitor compound 8a by virtual screening and identified by molecular and cellular SIRT6 activity assays. 8a could effectively inhibit SIRT6 deacetylation activity with IC50 values of 7.46 ± 0.79 µM in FLUOR DE LYS assay, and 8a significantly increased the acetylation levels of H3 in cells. Then, we found that 8a could inhibit the cell proliferation and induce cell apoptosis in pancreatic cancer cells. We further demonstrate that 8a sensitize pancreatic cancer cells to gemcitabine via reversing the activation of PI3K/AKT/mTOR and ERK signaling pathways induced by gemcitabine and blocking the DNA damage repair pathway. Moreover, combination of 8a and gemcitabine induces cooperative antitumor activity in pancreatic cancer xenograft model in vivo. Overall, we demonstrate that 8a, a novel SIRT6 inhibitor, could be a promising potential drug candidate for pancreatic cancer treatment.


Assuntos
Neoplasias Pancreáticas , Sirtuínas , Humanos , Apoptose , Linhagem Celular Tumoral , Gencitabina , Neoplasias Pancreáticas/patologia , Fosfatidilinositol 3-Quinases/metabolismo , Pirróis/farmacologia , Pirróis/uso terapêutico , Sirtuínas/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
3.
Carbohydr Polym ; 298: 120123, 2022 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-36241295

RESUMO

Tumor surgery is often accompanied by tumor residue, tissue defects, bleeding, and bacterial infection, which can easily cause tumor recurrence, low survival rates, and delay wound healing. In this study, a multifunctional hydrogel (CA-AuAgNPs-Gel) was developed to prevent tumor recurrence and promote wound healing after tumor surgery in the absence of chemotherapeutic drugs and antibiotics. CA-AuAgNPs-Gel was prepared using iota carrageenan (CA)-capped gold­silver nanoparticles (CA-AuAgNPs) and poloxamer 407 (F127), which exhibited good biocompatibility, injectability, and near-infrared (NIR) photothermal responsiveness. CA-AuAgNPs-Gel inhibited the growth of 4T1 breast cancer in situ and the recurrence of surgically resected B16F10 melanoma. It also effectively stopped bleeding and promoted tumor postsurgical wound healing in vivo. Importantly, CA-AuAgNPs-Gel induced tumor apoptosis via photothermal-induced hyperthermia and immunogenic cell death (ICD) under NIR laser radiation. Collectively, this hydrogel shows significant clinical application prospects for inhibiting tumor recurrence and promoting wound healing for postsurgical tumor treatment.


Assuntos
Hidrogéis , Nanopartículas Metálicas , Antibacterianos/química , Carragenina/farmacologia , Ouro/farmacologia , Humanos , Hidrogéis/química , Nanopartículas Metálicas/química , Nanopartículas Metálicas/uso terapêutico , Recidiva Local de Neoplasia/tratamento farmacológico , Poloxâmero , Prata/farmacologia , Cicatrização
4.
Int J Biol Macromol ; 222(Pt A): 1078-1091, 2022 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-36183754

RESUMO

The complex tumor microenvironment (TME) makes it difficult for single chemotherapy to achieve satisfactory therapeutic effects. Here, chitosan-coated hyaluronic acid micelles (R/C/D@HAssOA) that co-delivers doxorubicin (DOX) and programmed death-ligand 1 small interfering RNA (siPD-L1) are developed to enhance anti-tumor effect by combination of immunotherapy and chemotherapy. The pH/reduction dual-responsive co-delivery micelles R/C/D@HAssOA are spherical particles about 180 nm, and have good drug loading performance, stability, biocompatibility, and TME-responsive drug release properties. The CD44 receptor targeting HA significantly enhances the cellular uptake of DOX and siPD-L1, and siPD-L1 causes the immune infiltration of CD4+/CD8+ T cells by silencing PD-L1 expression. In vivo studies show that R/C/D@HAssOA exhibits significantly stronger anti-breast cancer effect than that of free DOX and micelles loaded only DOX. Therefore, the dual-stimulus responsive micelles provide a promising strategy for combining chemotherapy and siRNA-based immunotherapy to enhance efficacy.


Assuntos
Neoplasias da Mama , Quitosana , Humanos , Feminino , Micelas , Ácido Hialurônico , Linfócitos T CD8-Positivos , Doxorrubicina/farmacologia , Oxirredução , Concentração de Íons de Hidrogênio , Linhagem Celular Tumoral , Microambiente Tumoral
5.
Nutr Cancer ; 74(5): 1780-1791, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-34601984

RESUMO

Gastric cancer is the leading cause of cancer-related death worldwide. The aim of present study was to investigate the anti-tumor effect of purified Omphalia lapidescens protein (pPeOp) in gastric cancer. Microarray analysis was performed to find out differentially expressed genes in pPeOp-treated MC-4 gastric cancer cells. The Janus kinase (JAK)/signal transducer and activator of transcription (STAT) three signaling pathway was most likely to be altered based on bioinformatics analysis. Interleukin-6 (IL-6) and NSC74859 were used as the agonist and inhibitor of the JAK/STAT3 signaling pathway, respectively. Flow cytometry and MTS assay were used for cell proliferation and viability analysis in pPeOp-treated gastric cancer cell lines with IL-6 or NSC74859. The anti-tumor effect was increased when pPeOp were co-treated with IL-6, while decreased in inhibitor treatment. The expression of the crucial members in the pathway of MC-4 cells, including glycoprotein 130 (GP130), JAK1, JAK2, STAT3, p-STAT3, suppressor of cytokine signaling SOCS1 and SOCS3, was detected by western blotting. pPeOp exhibited promising anticancer effect in the xenograft nude mice model, established by STAT3 knock down gastric cancer cells.Thus, JAK/STAT3 inhibition partially contributed to the anticancer effect of pPeOp, which may serve as a novel strategy for gastric cancer.Supplemental data for this article is available online at https://doi.org/10.1080/01635581.2021.1960385.


Assuntos
Janus Quinases , Neoplasias Gástricas , Animais , Linhagem Celular Tumoral , Humanos , Interleucina-6/genética , Interleucina-6/metabolismo , Janus Quinases/metabolismo , Janus Quinases/farmacologia , Camundongos , Camundongos Nus , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais , Neoplasias Gástricas/metabolismo
6.
Biomacromolecules ; 22(7): 2921-2934, 2021 07 12.
Artigo em Inglês | MEDLINE | ID: mdl-34180218

RESUMO

Glioblastoma (GBM) is a fatal brain tumor with poor prognosis. Blood-brain barrier (BBB) prevents the effective delivery of chemotherapeutic agents to GBM. Herein, we developed a pH/reduction-sensitive carboxymethyl chitosan nanogel (CMCSN) modified by targeting peptide angiopep-2 (ANG) and loaded with doxorubicin (DOX). The multifunctional nanogel (DOX-ANG-CMCSN) exhibited good pH and reduction sensitivity, ideal stability, and biocompatibility. Its hydrodynamic diameter was 190 nm, drug loading was 12.7%, and the cumulative release rate of 24 h was 82.3% under the simulated tumor microenvironment. More importantly, the modification of ANG significantly enhanced BBB penetration and tumor targeting ability both in vivo and in vitro. DOX-ANG-CMCSN achieved 2-3-fold higher uptake and an enhanced antitumor activity compared with nontargeted DOX-CMCSN. Therefore, the targeted nanogels with the pH/reduction dual-stimuli response may provide a promising platform for GBM-targeted chemotherapy.


Assuntos
Quitosana , Glioblastoma , Linhagem Celular Tumoral , Doxorrubicina , Glioblastoma/tratamento farmacológico , Humanos , Concentração de Íons de Hidrogênio , Nanogéis , Peptídeos , Microambiente Tumoral
7.
Chem Biodivers ; 18(6): e2100270, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-33890414

RESUMO

Amylin (hIAPP) amyloid formation plays an important role in the pathogenesis of type 2 diabetes (T2D), which makes it a promising therapeutic target for T2D. In this study, we established a screening tool for identifying chemicals affecting hIAPP amyloid formation based on a reported genetic tool, which constantly tracks protein aggregates in Saccharomyces cerevisiae. In order to obtain the hIAPP with better aggregation ability, the gene of hIAPP was tandemly ligated to create 1×, 2×, 4× or 6×-hIAPP expressing strains. By measuring the cell density and fluorescence intensity of green fluorescent protein (GFP) regulated by the aggregation status of hIAPP, it was found that four intramolecular ligated hIAPP (4×hIAPP) could form obvious amyloids with mild toxicity. The validity and reliability of the screening tool were verified by testing six reported hIAPP inhibitors, including curcumin, epigallocatechin gallate and so on. Combined with surface plasmon resonance (SPR) and the screening tool, which could be a screening system for hIAPP inhibitors, we found that crocin specifically binds to hIAPP and acts inhibit amyloid formation of hIAPP. The effect of crocin was further confirmed by Thioflavin T (ThT) fluorescence and transmission electron microscopy (TEM) analysis. Thus, a screening system for hIAPP amyloid inhibitors and a new mechanism of crocin on anti-T2D were obtained as a result of this study.


Assuntos
Carotenoides/farmacologia , Diabetes Mellitus Tipo 2/tratamento farmacológico , Hipoglicemiantes/farmacologia , Polipeptídeo Amiloide das Ilhotas Pancreáticas/antagonistas & inibidores , Agregação Patológica de Proteínas/tratamento farmacológico , Carotenoides/química , Diabetes Mellitus Tipo 2/metabolismo , Avaliação Pré-Clínica de Medicamentos , Humanos , Hipoglicemiantes/química , Polipeptídeo Amiloide das Ilhotas Pancreáticas/metabolismo , Agregação Patológica de Proteínas/metabolismo
8.
Nutr Diabetes ; 10(1): 31, 2020 08 14.
Artigo em Inglês | MEDLINE | ID: mdl-32796820

RESUMO

OBJECTIVES: The study aimed to examine the anti-diabetic effects of Gynura divaricata (GD) and the underlying mechanism. METHODS: Information about the chemical compositions of GD was obtained from extensive literature reports. Potential target genes were predicted using PharmMapper and analyzed using Kyoto Encyclopedia of Genes and Genomes (KEGG) and Gene Ontology (GO). To validate the results from bioinformatics analyses, an aqueous extract of GD was administered to type 2 diabetic rats established by feeding a high-fat and high-sugar diet followed by STZ injection. Key proteins of the PI3K/AKT signaling pathway and fatty acid metabolism signaling pathway were investigated by immunoblotting. RESULTS: The blood glucose of the rats in the GD treatment group was significantly reduced compared with the model group without treatment. GD also showed activities in reducing the levels of alanine aminotransferase (ALT), aspartate aminotransferase (AST), blood urea nitrogen (BUN), and creatinine (CREA). The levels of urine sugar (U-GLU) and urine creatinine (U-CREA) were also lowered after treatment with GD. Bioinformatics analysis showed that some pathways including metabolic pathways, insulin resistance, insulin signaling pathway, PPAR signaling pathway, bile secretion, purine metabolism, etc. may be regulated by GD. Furthermore, GD significantly increased the protein expression levels of PKM1/2, p-AKT, PI3K p85, and GLUT4 in the rat liver. In addition, the expression levels of key proteins in the fatty acid metabolism signaling pathway including AMPK, p-AMPK, PPARα, and CPT1α were significantly upregulated. The anti-apoptotic protein BCL-2/BAX expression ratio in rats was significantly upregulated after GD intervention. These results were consistent with the bioinformatics analysis results. CONCLUSIONS: Our study suggests that GD can exert hypoglycemic effects in vivo by regulating the genes at the key nodes of the PI3K/AKT signaling pathway and fatty acid metabolism signaling pathway.


Assuntos
Asteraceae/química , Diabetes Mellitus Experimental/tratamento farmacológico , Ácidos Graxos/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Extratos Vegetais/farmacologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Animais , Glicemia/análise , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 2/tratamento farmacológico , Diabetes Mellitus Tipo 2/metabolismo , Hipoglicemiantes/química , Hipoglicemiantes/farmacologia , Insulina/metabolismo , Resistência à Insulina , Fígado/metabolismo , Masculino , Extratos Vegetais/química , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos
9.
Biotechnol Lett ; 42(10): 1919-1925, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-32399660

RESUMO

OBJECTIVE: To characterize the hydrolysis product and the substrate binding in the catalytic cavity of α-agarase AgaD. RESULTS: The time course curve showed that AgaD degraded agarose by the endo-type cleavage. AgaD did not degrade agarobiose (A2) and agarotetraose (A4). The minimum-length substrate was agarohexaose (A6), which was cleaved into A2 and A4. Agarooctaose (A8) was cleaved into two molecules of A4. Consistently, TLC and NMR data identified agarotetraose (A4) as the main hydrolysate when agarose was degraded by AgaD. CONCLUSION: This study confirms AgaD is an endo-type α-agarase and A4 as the main hydrolysis product of AgaD, which suggests the catalytic cavity of AgaD accommodates eight sugar units spanning from - 4 to + 4.


Assuntos
Proteínas de Bactérias , Glicosídeo Hidrolases , Proteínas Recombinantes , Sítios de Ligação , Catálise , Gammaproteobacteria/enzimologia , Gammaproteobacteria/genética , Hidrólise , Sefarose/química , Sefarose/metabolismo
10.
Mar Drugs ; 17(1)2019 Jan 11.
Artigo em Inglês | MEDLINE | ID: mdl-30641946

RESUMO

Our previous results suggested that EPS11, a novel marine bacterial polysaccharide, might be a potential drug candidate for human non-small cell lung carcinoma treatment. In this study, we further investigate the anticancer mechanisms against liver cancer and the anti-metastatic effects in vivo of EPS11. Firstly, we found that EPS11 exerts cytotoxic effects via blocking cell adhesion and destroying filiform structure formation in Huh7.5 cells. Moreover, mass spectrometry-based proteomic analysis of EPS11-treated Huh7.5 cells revealed that expression of many adhesion-related proteins was significantly changed. It is noteworthy that the expression of CD99, a key factor related to cell adhesion, migration and cell death, is remarkably down-regulated after EPS11 treatment. Importantly, over-expression of CD99 partly rescues cell death rate, and improves cell adhesion and migration ability in Huh7.5 treated by EPS11. Thus, we propose that CD99 is a potential action target of EPS11, inhibiting cancer cell proliferation, adhesion and migration. Notably, administration of EPS11 simultaneously with tumor induction evidently reduces tumor nodule formation in the lungs, which strongly indicates that EPS11 has anti-metastatic effects in vivo. Taken together, our results suggest that EPS11 inhibits liver cancer cell growth via blocking cell adhesion and attenuating filiform structure formation, and has potential as an anti-cancer drug, targeting metastasis of cancer cells, in the future.


Assuntos
Organismos Aquáticos/química , Bacillus/química , Neoplasias Hepáticas/tratamento farmacológico , Neoplasias Pulmonares/prevenção & controle , Polissacarídeos Bacterianos/farmacologia , Antígeno 12E7/metabolismo , Animais , Adesão Celular/efeitos dos fármacos , Linhagem Celular Tumoral/transplante , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Modelos Animais de Doenças , Regulação para Baixo/efeitos dos fármacos , Ensaios de Seleção de Medicamentos Antitumorais , Humanos , Neoplasias Hepáticas/patologia , Neoplasias Pulmonares/secundário , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Polissacarídeos Bacterianos/uso terapêutico
11.
J Diabetes Res ; 2018: 8120847, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29744368

RESUMO

The aim of the present study was to identify key antidiabetic nodes in the livers of pioglitazone-treated type 2 diabetes mellitus Sprague-Dawley rats by transcriptomic and proteomic analysis. Rats were randomly divided into the control, the diabetes model, and the pioglitazone-treated groups. After treatment with pioglitazone for 11 weeks, the effects on fasting blood glucose, body weight, and blood biochemistry parameters were evaluated. Microarray and iTRAQ analysis were used to determine the differentially expressed genes/proteins in rat livers. 1.5-fold changes in gene expression and 1.2-fold changes in protein were set as the screening criteria. After treatment with pioglitazone for 11 weeks, fasting blood glucose in pioglitazone-treated rats was significantly lower than that in the model group. There was a tendency for pioglitazone to reduce TC, TG, TP, ALB, BUN, and HDL-c levels. Kyoto Encyclopedia of Genes and Genomes (KEGG) and gene ontology (GO) were applied to analyze differentially expressed genes/proteins. Furthermore, Western blotting and RT-qPCR were used to validate the results of microarray and iTRAQ. In conclusion, Cyp7a1, Cp, and RT1-EC2 are differentially expressed genes/proteins since they showed a similar trend in rats in the model group and the pioglitazone-treated group.


Assuntos
Diabetes Mellitus Tipo 2/tratamento farmacológico , Hipoglicemiantes/farmacologia , Fígado/efeitos dos fármacos , Tiazolidinedionas/farmacologia , Transcriptoma/efeitos dos fármacos , Animais , Glicemia , Diabetes Mellitus Tipo 2/metabolismo , Perfilação da Expressão Gênica , Hipoglicemiantes/uso terapêutico , Fígado/metabolismo , Masculino , Pioglitazona , Proteômica , Ratos , Ratos Sprague-Dawley , Tiazolidinedionas/uso terapêutico
12.
Oncol Lett ; 15(4): 4161-4170, 2018 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-29541181

RESUMO

Gastric cancer is one of the most common cancers globally with high rates of morbidity and mortality. Purified Omphalia lapidescens protein (pPeOp) is a protein extracted from the sclerotium of Omphalia lapidescens. The present study aimed to investigate the effects of pPeOp on the viability, migration, cell cycle progression and apoptosis of SGC-7901 cells. The expression of numerous proteins, namely matrix metallopeptidase (MMP)2, MMP9, p53, caspase-3, B-cell lymphoma (Bcl)-2, cyclin A2, cyclin B1, cyclin D1, cyclin dependent kinase (CDK)1, CDK2 and CDK4, were investigated using western blot analysis and reverse transcription-quantitative polymerase chain reaction. The results of the present study demonstrated that treating SGC-7901 cells with pPeOp markedly suppressed their migration, induced their apoptosis and arrested their progression in S phase. pPeOp also downregulated the expression of migration-associated proteins (MMP2 and MMP9) and cyclin-associated proteins (cyclin A2, cyclin B1, cyclin D1, CDK1, CDK2 and CDK4) in a dose-dependent manner. Cells treated with pPeOp significantly upregulated caspase-3 and p53 and downregulated Bcl-2. Finally, the impact of pPeOp on three key nodes of the Janus kinase (JAK)-signal transducer and activator of transcription (STAT) pathway were investigated and it was revealed that expression levels of JAK1, JAK2 and STAT3 were significantly downregulated following treatment. Together, the results of the present study suggested that pPeOp suppresses metastasis, arrests cell cycle, induces apoptosis and inhibits the JAK-STAT signaling pathway in SGC-7901 cells. Therefore, pPeOp may serve as a novel therapeutic agent for patients with gastric cancer.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...