Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Mais filtros











Intervalo de ano de publicação
1.
Sci Total Environ ; 949: 175193, 2024 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-39094643

RESUMO

Cadmium (Cd) and arsenic (As), two toxic elements to humans, are ubiquitously coexisting contaminant found in paddy fields. The accumulation of Cd and As in rice, a major food source for many people around the world, can pose a serious threat to food safety and human health. Therefore, it is crucial to be aware of these contaminants and take adequate measures to reduce the accumulation of these two elements in rice. Developing an effective method to simultaneously reduce the accumulation of Cd) and As in rice is challenging. In this study, a pot experiment was conducted to investigate the synergistic effects of selenium (Se), iron (Fe) and phosphorus (P) on the uptake, transport and accumulation of cadmium and arsenic in rice by analyzing the physical and chemical properties of the soil, the elemental concentrations and their interrelationships in the rice tissues, and the composition and morphology of the iron plaque (IP). The results showed that the combined treatments of Se, Fe and P had positive effects on reducing Cd and As accumulation in rice, reducing Cd concentrations in brown rice by 3.86-51.88 % and As concentrations by 25.37-40.81 %. The possible mechanisms for the reduction of As and Cd concentrations in rice grains were: (i) Combined application of Fe, P and Se can effectively reduce the soil available Cd and As concentration. (ii) Combined application significantly improved the formation of IP at the tillering stage and increased the crystalline iron oxides in IP, promoting the deposition of SiO2 in rice roots, thereby effectively inhibiting the uptake of Cd and As by rice roots. (iii) Interplay and interaction between elements facilitated by transporter proteins could contribute to the synergistic mitigation of Cd and As by Se, Fe and P. This study provides a valuable new approach for effective control of Cd and As concentration of rice grown in co-contaminated soil.


Assuntos
Arsênio , Cádmio , Ferro , Oryza , Fósforo , Selênio , Poluentes do Solo , Cádmio/metabolismo , Arsênio/análise , Poluentes do Solo/análise , Fósforo/análise , Solo/química
2.
Chemosphere ; 363: 142903, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39029704

RESUMO

Selenium (Se) fortification is witnessed to simultaneously inhibit absorbing Cadmium (Cd) and Arsenic (As) by rice plants, but the mechanism is unclear. Here, the effects of Se on the root morphology, iron plaque (IP) content, soil Fe2+ content, radial oxygen loss (ROL), and enzyme activities of the rice plants in the soil contaminated by Cd and As were intensively investigated through the hydroponic and soil experiments. Se effectively alleviated the toxic effects of Cd and As on the plants and the dry weight, root length, and root width were increased by 203.18%, 33.41%, and 52.81%, respectively. It also elucidated that ROL was one of the key factors to elevate IP formation by Se and the specific pathways of Se enhancing ROL were identified. ROL of the plants in the experiment group treated by Se was increased 36.76%, and correspondingly IP was magnified 50.37%, compared to the groups with Cd and As. It was owing to Se significantly increased the root porosity (62.11%), facilitating O2 transport to the roots. Additionally, Se enhanced the activities of catalase (CAT) and superoxide dismutase (SOD) to promote the catalytic degradation of ROS induced by Cd and As stress. It indirectly increased O2 release in the rhizosphere, which benefit to form more robust IP serve as stronger barrier to Cd and As. The results of our study provide a novel molecular level insight for Se promoting root IP to block Cd and As uptake by the rice plants.


Assuntos
Arsênio , Cádmio , Ferro , Oryza , Raízes de Plantas , Selênio , Poluentes do Solo , Cádmio/toxicidade , Cádmio/metabolismo , Oryza/metabolismo , Oryza/efeitos dos fármacos , Arsênio/metabolismo , Arsênio/toxicidade , Selênio/farmacologia , Poluentes do Solo/toxicidade , Poluentes do Solo/metabolismo , Ferro/metabolismo , Raízes de Plantas/metabolismo , Raízes de Plantas/efeitos dos fármacos , Superóxido Dismutase/metabolismo , Solo/química , Catalase/metabolismo , Rizosfera , Espécies Reativas de Oxigênio/metabolismo
3.
World J Gastrointest Oncol ; 16(4): 1465-1478, 2024 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-38660658

RESUMO

BACKGROUND: Colorectal cancer has a low 5-year survival rate and high mortality. Human ß-defensin-1 (hBD-1) may play an integral function in the innate immune system, contributing to the recognition and destruction of cancer cells. Long non-coding RNAs (lncRNAs) are involved in the process of cell differentiation and growth. AIM: To investigate the effect of hBD-1 on the mammalian target of rapamycin (mTOR) pathway and autophagy in human colon cancer SW620 cells. METHODS: CCK8 assay was utilized for the detection of cell proliferation and determination of the optimal drug concentration. Colony formation assay was employed to assess the effect of hBD-1 on SW620 cell proliferation. Bioinformatics was used to screen potentially biologically significant lncRNAs related to the mTOR pathway. Additionally, p-mTOR (Ser2448), Beclin1, and LC3II/I expression levels in SW620 cells were assessed through Western blot analysis. RESULTS: hBD-1 inhibited the proliferative ability of SW620 cells, as evidenced by the reduction in the colony formation capacity of SW620 cells upon exposure to hBD-1. hBD-1 decreased the expression of p-mTOR (Ser2448) protein and increased the expression of Beclin1 and LC3II/I protein. Furthermore, bioinformatics analysis identified seven lncRNAs (2 upregulated and 5 downregulated) related to the mTOR pathway. The lncRNA TCONS_00014506 was ultimately selected. Following the inhibition of the lncRNA TCONS_00014506, exposure to hBD-1 inhibited p-mTOR (Ser2448) and promoted Beclin1 and LC3II/I protein expression. CONCLUSION: hBD-1 inhibits the mTOR pathway and promotes autophagy by upregulating the expression of the lncRNA TCONS_00014506 in SW620 cells.

4.
iScience ; 27(3): 109118, 2024 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-38439955

RESUMO

Duodenogastric reflux (DGR) is closely associated with gastric inflammation and tumorigenesis; however, the precise mechanism is unclear. Hence, we aim to clarify this molecular mechanism and design an effective therapeutic strategy based on it. The present study found that DGR induced TXNIP/NLRP3 inflammasome activation and triggered pyroptosis in gastric mucosa in vitro and in vivo, in which endoplasmic reticulum (ER) stress via PERK/eIF2α/CHOP signaling was involved. Mechanistically, farnesoid X receptor (FXR) antagonized the DGR-induced PERK/eIF2α/CHOP pathway and reduced TXNIP and NLRP3 expression. Moreover, FXR suppressed NLRP3 inflammasome activation by physically interacting with NLRP3 and caspase-1. Administration of the FXR agonist OCA protected the gastric mucosa from DGR-induced barrier disruption and mucosal inflammation. In conclusion, our study demonstrates the involvement of TXNIP/NLRP3 inflammasome-mediated pyroptosis in DGR-induced gastric inflammation. FXR antagonizes gastric barrier disruption and mucosal inflammation induced by DGR. Restoration of FXR activity may be a therapeutic strategy for DGR-associated gastric tumorigenesis.

5.
Pharmacogenomics J ; 24(2): 5, 2024 Feb 20.
Artigo em Inglês | MEDLINE | ID: mdl-38378770

RESUMO

OBJECTIVE: To explore the role of p300 in the context of paclitaxel (PTX) resistance in triple-negative breast cancer (TNBC) cells, focusing on its interaction with the phosphoenolpyruvate carboxykinase 1 (PCK1)/adenosine monophosphate-activated protein kinase (AMPK) pathway. METHODS: The expression of p300 and PCK1 at the messenger ribonucleic acid (mRNA) level was detected using a quantitative polymerase chain reaction. The GeneCards and GEPIA databases were used to investigate the relationship between p300 and PCK1. The MDA-MB-231/PTX cell line, known for its PTX resistance, was chosen to understand the specific role of p300 in such cells. The Lipofectamine™ 3000 reagent was used to transfer the p300 small interfering RNA and the overexpression of PCK1 plasmid into MDA-MB-231/PTX. The expression levels of p300, PCK1, 5'AMPK and phosphorylated AMPK (p-AMPK) were determined using the western blot test. RESULTS: In TNBC cancer tissue, the expression of p300 was increased compared with TNBC paracancerous tissue (P < 0.05). In the MDA-MB-231 cell line of TNBC, the expression of p300 was lower than in the PTX-resistant TNBC cells (MDA-MB-231/PTX) (P < 0.05). The PCK1 expression was decreased in the TNBC cancer tissue compared with TNBC paracancerous tissue, and the PCK1 expression was reduced in MDA-MB-231/PTX than in MDA-MB-231 (P < 0.05) indicating that PCK1 was involved in the resistance function. Additionally, p-AMPK was decreased in MDA-MB-231/PTX compared with MDA-MB-231 (P < 0.05). The adenosine triphosphate (ATP) level was also detected and was significantly lower in MDA-MB-231/PTX than in MDA-MB-231 (P < 0.05). Additionally, cell proliferation increased significantly in MDA-MB-231/PTX at 48 and 72 h (P < 0.05) suggesting that MDA-MB-231/PTX cells obtained the resistance function which was associated with AMPK and ATP level. When p300 was inhibited, p-AMPK and ATP levels elevated in MDA-MB-231/PTX (P < 0.05). When PCK1 was suppressed, the ATP consumption rate decreased, and cell proliferation increased (P < 0.05). However, there were no changes in p300. CONCLUSIONS: In MDA-MB-231/PTX, p300 can inhibit p-AMPK and ATP levels by inhibiting PCK1 expression. Our findings suggest that targeting p300 could modulate the PCK1/AMPK axis, offering a potential therapeutic avenue for overcoming PTX resistance in TNBC.


Assuntos
Paclitaxel , Neoplasias de Mama Triplo Negativas , Humanos , Trifosfato de Adenosina/uso terapêutico , Proteínas Quinases Ativadas por AMP/genética , Proteínas Quinases Ativadas por AMP/metabolismo , Proteínas Quinases Ativadas por AMP/uso terapêutico , Linhagem Celular Tumoral , Proliferação de Células , Peptídeos e Proteínas de Sinalização Intracelular/genética , Paclitaxel/farmacologia , Paclitaxel/uso terapêutico , Fosfoenolpiruvato Carboxiquinase (GTP)/genética , Fosfoenolpiruvato Carboxiquinase (GTP)/metabolismo , Fosfoenolpiruvato Carboxiquinase (GTP)/uso terapêutico , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Neoplasias de Mama Triplo Negativas/genética , Neoplasias de Mama Triplo Negativas/metabolismo , Regulação para Cima
6.
Photodermatol Photoimmunol Photomed ; 40(1): e12931, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38009842

RESUMO

BACKGROUND: Collagen dominates the skin's extracellular matrix (ECM). Type I collagen comprises 80%-90% of the skin's collagen, followed by type III (8%-12%) and type V (5%). Reactive oxygen species, matrix metalloproteinases, and collagen degradation all increase during photoaging, which disrupts the ECM's dynamic balance and lowers the amount of total collagen in the body. In recent years, domestic and foreign researchers have conducted multidimensional and multifaceted studies on collagen and skin photoaging. Collagen and the peptides that are derivates of it are currently being used more and more in biomedicine and medical esthetics. OBJECTIVE: Offering new suggestions for both the avoidance and remedy of photoaging. METHODS: This article reviews collagen and its potential connection to skin photoaging, illustrates the effects of collagen and peptide supplementation derivatives on photoaged skin, and briefly describes other compounds that can also be used to fight photoaging by increasing collagen synthesis in the skin. RESULT: Both internal and external aging are inevitable, and as the main component of extracellular matrix, collagen plays a variety of functions in maintaining skin structure and fighting skin aging, and its role in photoaging is undeniable. Ultraviolet radiation can induce increased fragmentation and degradation of cutaneous collagen, while conversely, supplementation with collagen can effectively counteract photodamage-induced skin impairment. CONCLUSION: Collagen and its derived peptides are indispensable in photoaging skin, holding promising prospects for applications in skin aging.


Assuntos
Envelhecimento da Pele , Humanos , Raios Ultravioleta/efeitos adversos , Pele/metabolismo , Colágeno/metabolismo , Peptídeos/metabolismo
8.
Int J Pharm ; 626: 122180, 2022 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-36087627

RESUMO

Combining celecoxib with other chemopreventive drugs is a promising method of chemoprevention for cancer, especially for colorectal cancer. However, the traditional drug combination approaches are restricted with high-cost apparatus, complex and numerous unit operations. This work aims to develop an efficient spherical co-agglomeration strategy for celecoxib in combination with lovastatin, which can achieve drug combination in a single crystallization unit. The ternary solvent system was determined based on molecular simulation, and then a stable spherical agglomeration process was developed through the design of molar fraction of anti-solvent (MFA) and stirring rate to produce spherical agglomerates with high sphericity (84.2-89.9 %) and narrow size distribution. On this basis, celecoxib-benzoic acid spherical co-agglomerates were designed to form a complete spherical co-agglomeration strategy, which includes solvent system selection, spherical agglomeration and spherical co-agglomeration. Finally, celecoxib-lovastatin spherical co-agglomerates with synergistic efficacy were successfully produced by this strategy, with controllable and stable drug content (fluctuation < 2.7 %), good powder properties, and improved tabletability.


Assuntos
Ácido Benzoico , Lovastatina , Celecoxib , Combinação de Medicamentos , Tamanho da Partícula , Pós , Solventes/química
9.
Cell Death Dis ; 13(4): 388, 2022 04 21.
Artigo em Inglês | MEDLINE | ID: mdl-35449124

RESUMO

Our previous study indicated that colon cancer cells varied in sensitivity to pharmacological farnesoid X receptor (FXR) activation. Herein, we explore the regulatory mechanism of FXR in colorectal cancer (CRC) development and aim to design effective strategies of combined treatment based on the regulatory axis. We found that the expression of FXR was negatively correlated with enhancer of zeste homolog 2 (EZH2) in colon cancer tissues. EZH2 transcriptionally suppressed FXR via H3K27me3. The combination of FXR agonist OCA plus EZH2 inhibitor GSK126 acted in a synergistic manner across four colon cancer cells, efficiently inhibiting clonogenic growth and invasion in vitro, retarding tumor growth in vivo, preventing the G0/G1 to S phase transition, and inducing caspase-dependent apoptosis. Benign control cells FHC were growth-arrested without apoptosis induction, but retained long-term proliferation and invasion capacity. Mechanistically, the drug combination dramatically accelerated FXR nuclear location and cooperatively upregulated caudal-related homeobox transcription factor 2 (CDX2) expression. The depletion of CDX2 antagonized the synergistic effects of the drug combination on tumor inhibition. In conclusion, our study demonstrated histone modification-mediated FXR silencing by EZH2 in colorectal tumorigenesis, which offers useful evidence for the clinical use of FXR agonists combined with EZH2 inhibitors in combating CRC.


Assuntos
Neoplasias do Colo , Proteína Potenciadora do Homólogo 2 de Zeste , Fator de Transcrição CDX2/genética , Carcinogênese/genética , Linhagem Celular Tumoral , Proliferação de Células , Proteína Potenciadora do Homólogo 2 de Zeste/genética , Regulação Neoplásica da Expressão Gênica , Humanos
10.
Nat Commun ; 12(1): 7094, 2021 12 07.
Artigo em Inglês | MEDLINE | ID: mdl-34876574

RESUMO

Oxidative stress contributes to the pathogenesis of acute lung injury. Protein S-glutathionylation plays an important role in cellular antioxidant defense. Here we report that the expression of deglutathionylation enzyme Grx1 is decreased in the lungs of acute lung injury mice. The acute lung injury induced by hyperoxia or LPS is significantly relieved in Grx1 KO and Grx1fl/flLysMcre mice, confirming the protective role of Grx1-regulated S-glutathionylation in macrophages. Using a quantitative redox proteomics approach, we show that FABP5 is susceptible to S-glutathionylation under oxidative conditions. S-glutathionylation of Cys127 in FABP5 promotes its fatty acid binding ability and nuclear translocation. Further results indicate S-glutathionylation promotes the interaction of FABP5 and PPARß/δ, activates PPARß/δ target genes and suppresses the LPS-induced inflammation in macrophages. Our study reveals a molecular mechanism through which FABP5 S-glutathionylation regulates macrophage inflammation in the pathogenesis of acute lung injury.


Assuntos
Lesão Pulmonar Aguda/tratamento farmacológico , Proteínas de Ligação a Ácido Graxo/metabolismo , Inflamação , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Estresse Oxidativo , Substâncias Protetoras/farmacologia , Lesão Pulmonar Aguda/metabolismo , Lesão Pulmonar Aguda/patologia , Animais , Proteínas de Ligação a Ácido Graxo/genética , Regulação da Expressão Gênica , Glutarredoxinas/genética , Glutarredoxinas/metabolismo , Hiperóxia , Pulmão/patologia , Macrófagos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Oxirredução , Processamento de Proteína Pós-Traducional , Proteínas Recombinantes
11.
J BUON ; 26(4): 1365-1372, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34564993

RESUMO

PURPOSE: Human ß-defensin 1 (DEFB1) belongs to defensins family that contribute to innate immune responses and was recently found to downregulate a variety of cancers, including renal, prostatic, and oral squamous cell carcinoma, and therefore is considered as a potential tumor suppressor. However, the role of DEFB1 in hepatocellular carcinoma (HCC) still needs to be elucidated. METHODS: Quantitative PCR and Western blot were used to measure the expression levels of interested proteins. CCK-8 and colony formation assays were performed to determine the ability of cell proliferation. Tumor formation experiments in nude mice were used to examine the tumor growth. RESULTS: The expression level of DEFB1 was dramatically downregulated in human HCC. Quantitative PCR and Western blot results also showed a pronounced decrease of DEFB1 expression in the liver cancer cell lines. Rescuing the expression of DEFB1 in Huh7 cells effectively suppressed cell proliferation and reduced the colony forming ability, probably by inducing cell apoptosis and cell cycle arrest. Moreover, tumor formation experiments in nude mice also showed inhibition of tumor growth by DEFB1 expression in vivo. Furthermore, induction of DEFB1 expression induced degraded protein increase and endoplasmic reticulum (ER) stress, which subsequently activated JNK pathway. Pharmacologic inhibition of ER stress by 4-phenylbutyrate, a compound to alleviate ER stress, effectively eliminated DEFB1-induction inhibition of cell proliferation and migration. CONCLUSION: DEFB1 functions as a tumor suppressor in HCC through activating ER stress and JNK pathway, which may provide a potential strategy for HCC treatment.


Assuntos
Carcinoma Hepatocelular/patologia , Estresse do Retículo Endoplasmático/fisiologia , Neoplasias Hepáticas/patologia , Sistema de Sinalização das MAP Quinases/fisiologia , beta-Defensinas/fisiologia , Animais , Proliferação de Células , Camundongos , Camundongos Nus , Células Tumorais Cultivadas
12.
Artigo em Inglês | MEDLINE | ID: mdl-34475961

RESUMO

Sendeng-4 is a traditional Chinese medicine that has been successfully applied to anti-inflammatory diseases in clinical practice. Monomers within Sendeng-4 showed promising antitumor activity against lung cancer, colon cancer, and cutaneous cancer. However, potency of Sendeng-4 in melanoma has not been explored. This study aims to explore the potential application of Sendeng-4 in melanoma treatment. In the present study, we systemically investigate the possibility of Sendeng-4 for treatment of melanoma cancer in vitro by proliferation assay, colony formation, flow cell cytometry, RNA-seq, western blot, and fluorescence-based assay. Our data demonstrated that Sendeng-4 suppresses the proliferation and colony formation capacity of melanoma cells and induces cell cycle block at G2/M phase and eventually cell death. Mechanistically, transcriptome sequencing demonstrates that the PI3K-AKT pathway was significantly inactivated upon Sendeng-4 exposure, which was confirmed by western blot showing decreased phosphorylation of AKT. In addition, decreased BCL-2 expression and increased BAX expression were observed, suggesting programmed cell death via apoptosis. Moreover, LC3-II production as well as autophagosomes formation was observed as demonstrated by western blot and immunofluorescence, indicating elevated autophagy network by Sendeng-4 stimulation. Collectively, we concluded that Sendeng-4 might be used as an anticancer drug for melanoma.

13.
Exp Cell Res ; 407(2): 112809, 2021 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-34487729

RESUMO

Defensins are highly conserved antimicrobial peptides, which ubiquitously expressed in different species. In addition to the functions in host defense, their aberrant expression have also been documented in cancerous tissue including breast cancer, lung caner and renal carcinoma etc. Whereas, roles of Defensin Alpha 5 (DEFA5) in colon cancer has not been explored. Bioinformatic analysis was used to study the expression of DEFA5 and its correlation with clinical outcomes; Western blot, qPCR, Co-immunoprecipitation, xenograft models were used to the study the molecular mechanism. Decreased expression of DEFA5 at protein level was observed in colon tissues. Colon cancer cell lines proliferation and colony formation capacity were significantly suppressed by DEFA5 overexpression. Moreover, in vivo tumor growth in nude mice was also suppressed by DEFA5 overexpression, suggesting a tumor suppressor role of DEFA5 in colon cancer. Mechanistically, DEFA5 directly binds to the subunits of PI3K complex, thus attenuates the downstream signaling transduction, leads to delayed cell growth and metastasis. Collectively, we concluded that DEFA5 showed an inhibitory effect in colon cancer cell growth and may serve as a potential tumor suppressor in colon cancer.


Assuntos
Biomarcadores Tumorais/metabolismo , Neoplasias do Colo/prevenção & controle , Regulação Neoplásica da Expressão Gênica , Fosfatidilinositol 3-Quinases/química , alfa-Defensinas/metabolismo , Animais , Apoptose , Biomarcadores Tumorais/genética , Movimento Celular , Proliferação de Células , Neoplasias do Colo/genética , Neoplasias do Colo/metabolismo , Neoplasias do Colo/patologia , Feminino , Humanos , Camundongos , Camundongos Nus , Fosfatidilinositol 3-Quinases/metabolismo , Prognóstico , Taxa de Sobrevida , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto , alfa-Defensinas/genética
14.
Poult Sci ; 99(11): 5356-5365, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-33142452

RESUMO

Macrophages are professional phagocytic cells that play a critical role in initiating immune responses by presenting antigen and phagocytic clearance. The macrophages can be targeted for immunomodulation by beneficial microbes, such as probiotics. The aim of this study is to investigate the protective effect of Saccharomyces boulardii against Clostridium perfringens infection in avian macrophage cell line HD11. In this study, HD11 macrophages were prestimulated with S. boulardii for 6 h and then infected with C. perfringens for 3 h. Results showed that S. boulardii enhanced phagocytosis and bactericidal capacity against C. perfringens by HD11 cells. The S. boulardii effectively promoted the mRNA expression of CD80, CD83, and CD197 cell-surface molecules in C. perfringens-infected HD11 cells. Moreover, we found that prestimulation with S. boulardii reduced the mRNA expression of CD40, toll-like receptor [TLR] 4, and TLR15 induced by C. perfringens and thereby downregulated the mRNA expression of myeloid differentiation primary response 88, TNF receptor associated factor 6, nuclear factor kappa-B p65 subunit, and c-Jun N-terminal kinase genes in HD11 cells. The upregulation of cytokines (interleukin [IL]-6, tumor necrosis factor alpha, and IL-10) and inducible nitric oxide synthase mRNA expression in C. perfringens-infected HD11 cells were noticeably inhibited by S. boulardii pretreatment. Conclusively, these results might provide a new insight into the role of S. boulardii in regulating avian immune defense against C. perfringens invasion and immune escape.


Assuntos
Antibiose , Infecções por Clostridium , Clostridium perfringens , Doenças das Aves Domésticas , Saccharomyces boulardii , Animais , Antibiose/imunologia , Galinhas , Infecções por Clostridium/imunologia , Infecções por Clostridium/microbiologia , Infecções por Clostridium/veterinária , Inflamação/imunologia , Macrófagos/imunologia , Macrófagos/microbiologia , Fator 88 de Diferenciação Mieloide/imunologia , Doenças das Aves Domésticas/imunologia , Doenças das Aves Domésticas/microbiologia , Saccharomyces boulardii/imunologia , Receptor 4 Toll-Like/imunologia , Receptores Toll-Like/imunologia
15.
Cancer Sci ; 111(10): 3503-3515, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-32726482

RESUMO

Hepatocellular carcinoma (HCC) represents the majority of liver cancer and is the fourth most common cause of cancer-related death. Although advances in molecular targeted therapy have shown promise, none of these agents has yet demonstrated significant clinical benefit. Bromo- and extraterminal domain (BET) protein inhibitors have been considered potential therapeutic drugs for HCC but the biological activity remains unclear. This study found that BET protein inhibition did not effectively suppress the progression of HCC, using a transgenic HCC mouse model. Mechanistically, the BET protein inhibitor JQ1 upregulated the expression of programmed cell death-ligand 1 (PD-L1) on the plasma membrane in vivo and in vitro. Moreover, JQ1 enhanced the expression of Rab8A, which upregulated the expression of PD-L1 on the plasma membrane. This study also showed that JQ1 combined with anti-PD-L1 Ab effectively suppressed HCC progression, and this benefit was obtained by enhancing the activation and cytotoxic capabilities of CD8 T cells. These results revealed the crucial role and regulation of BET protein inhibition on the expression of PD-L1 in HCC. Thus, combining BET protein inhibition with immune checkpoint blockade offers an efficient therapeutic approach for HCC.


Assuntos
Carcinoma Hepatocelular/imunologia , Carcinoma Hepatocelular/terapia , Neoplasias Hepáticas/imunologia , Neoplasias Hepáticas/terapia , Proteínas/antagonistas & inibidores , Animais , Antineoplásicos/farmacologia , Antígeno B7-H1/metabolismo , Linfócitos T CD8-Positivos/efeitos dos fármacos , Linfócitos T CD8-Positivos/imunologia , Carcinoma Hepatocelular/tratamento farmacológico , Carcinoma Hepatocelular/metabolismo , Linhagem Celular Tumoral , Progressão da Doença , Células Hep G2 , Humanos , Imunoterapia/métodos , Neoplasias Hepáticas/tratamento farmacológico , Neoplasias Hepáticas/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Terapia de Alvo Molecular/métodos , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/imunologia
16.
Transl Cancer Res ; 9(2): 603-612, 2020 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-35117405

RESUMO

BACKGROUND: Liver cancer is the second leading cause of worldwide cancer-related death, and it has an increasing incidence rate. To investigate the role of SRC-1 and Twist1 in liver cancer and determine their expression in terms of prognosis for patients with liver cancer and in general for hepatocellular carcinoma (HCC) cell lines. METHODS: The present study included a total of 70 patients who underwent liver transplantation or hepatic resection surgeries in our hospital from May 2011 to December 2012. Demographic data and clinical variables as well as alpha-fetoprotein (AFP) and hepatitis B virus (HBV) data were collected. The expression of SRC-1 and Twist1 was determined using immunohistochemistry (IHC). The expression of SRC-1 in different HCC cell lines was determined by quantitative real-time polymerase chain reaction (qRT-PCR). Following SRC-1 silencing by sh-RNA, cell viability, invasion, migration and expression of epithelial-mesenchymal transformation (EMT)-related proteins as well as Twist levels were measured. RESULTS: The expression of SRC-1 and Twist1 was positively correlated in HCC patients. The expression of SRC-1 differed significantly based on patient tumor diameter, tumor-node-metastasis (TNM) grade, and state of liver cirrhosis, and it also differed in patients with dissimilar tumor metastasis conditions, while the expression of Twist1 in patients was significantly correlated with TNM grade and state of liver cirrhosis as well as by the conditions of tumor metastasis. Survival analysis showed that the expression of both SRC-1 and Twist1 were significantly associated with the overall survival (OS) time of HCC patients. Meanwhile, patients with both SRC-1 (+) and Twist1 (+) tissue had the lowest OS, while patients with both SRC-1 (-) and Twist1 (-) tissue had the highest OS. Cox univariate and multivariate analyzes showed that SRC-1 expression, tumor stage and liver cirrhosis were independent risk factors for OS time. SRC-1 was highly expressed in HCC cell lines, and inhibition of SRC-1 had a significantly negative impact on cell viability, invasion, migration and EMT; it also inhibited the expression of Twist. CONCLUSIONS: Expression of both Twist1 and SRC-1 were correlated with clinical outcomes and prognoses for HCC patients, and both Twist1 and SRC-1 were independent risk factors for HCC patient survival conditions. Inhibition of SRC-1 suppressed cell proliferation, invasion, migration and EMT of HCC cells, which might be the result of Twist inhibition.

17.
Autophagy ; 15(10): 1774-1786, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-30957628

RESUMO

Multiple sources contribute membrane and protein machineries to construct functional macroautophagic/autophagic structures. However, the underlying molecular mechanisms remain elusive. Here, we show that RAB2 connects the Golgi network to autophagy pathway by delivering membrane and by sequentially engaging distinct autophagy machineries. In unstressed cells, RAB2 resides primarily in the Golgi apparatus, as evidenced by its interaction and colocalization with GOLGA2/GM130. Importantly, autophagy stimuli dissociate RAB2 from GOLGA2 to interact with ULK1 complex, which facilitates the recruitment of ULK1 complex to form phagophores. Intriguingly, RAB2 appears to modulate ULK1 kinase activity to propagate signals for autophagosome formation. Subsequently, RAB2 switches to interact with autophagosomal RUBCNL/PACER and STX17 to further specify the recruitment of HOPS complex for autolysosome formation. Together, our study reveals a multivalent pathway in bulk autophagy regulation, and provides mechanistic insights into how the Golgi apparatus contributes to the formation of different autophagic structures. Abbreviations: ACTB: actin beta; ATG9: autophagy related 9A; ATG14: autophagy related 14; ATG16L1: autophagy related 16 like 1; BCAP31: B cell receptor associated protein 31; BECN1: beclin 1; Ctrl: control; CQ: chloroquine; CTSD: cathepsin D; DMSO: dimethyl sulfoxide; EBSS: Earle's balanced salt solution; EEA1: early endosome antigen 1; GDI: guanine nucleotide dissociation inhibitor; GFP: green fluorescent protein; GOLGA2: golgin A2; HOPS: homotypic fusion and protein sorting complex; IP: immunoprecipitation; KD: knockdown; KO: knockout; LAMP1: lysosomal associated membrane protein 1; LC3: microtubule-associated protein 1 light chain 3; OE: overexpression; PtdIns3K: class III phosphatidylinositol 3-kinase; SQSTM1/p62: sequestosome 1; RAB2: RAB2A, member RAS oncogene family; RAB7: RAB7A, member RAS oncogene family; RAB11: RAB11A, member RAS oncogene family; RUBCNL/PACER: rubicon like autophagy enhancer; STX17: syntaxin 17; TBC1D14: TBC1 domain family member 14; TFRC: transferrin receptor; TGOLN2: trans-golgi network protein 2; TUBB: tubulin beta class I; ULK1: unc-51 like autophagy activating kinase 1; VPS41: VPS41, HOPS complex subunit; WB: western blot; WT: wild type; YPT1: GTP-binding protein YPT1.


Assuntos
Autofagossomos/metabolismo , Autofagia/genética , Lisossomos/metabolismo , Proteína rab2 de Ligação ao GTP/fisiologia , Animais , Células Cultivadas , Células Eucarióticas/metabolismo , Células HEK293 , Células HeLa , Humanos , Lisossomos/genética , Masculino , Mamíferos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína rab2 de Ligação ao GTP/genética
18.
J Cell Physiol ; 234(11): 19406-19419, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31020664

RESUMO

The intestinal mucosal barrier is the first line to defense against luminal content penetration and performs numerous biological functions. The intestinal epithelium contains a huge surface that is lined by a monolayer of intestinal epithelial cells (IECs). IECs are dominant mediators in maintaining intestinal homeostasis that drive diverse functions including nutrient absorption, physical segregation, secretion of antibacterial peptides, and modulation of immune responses. Autophagy is a cellular self-protection mechanism in response to various stresses, and accumulating studies have revealed its importance in participating physiological processes of IECs. The regulatory effects of autophagy depend on the specific IEC types. This review aims to elucidate the myriad roles of autophagy in regulating the functions of different IECs (stem cells, enterocytes, goblet cells, and Paneth cells), and present the progress of autophagy-targeting therapy in intestinal diseases. Understanding the involved mechanisms can provide new preventive and therapeutic strategies for gastrointestinal dysfunction and diseases.


Assuntos
Autofagia/genética , Células Epiteliais/metabolismo , Gastroenteropatias/genética , Mucosa Intestinal/metabolismo , Peptídeos Catiônicos Antimicrobianos , Colite/genética , Colite/microbiologia , Colite/patologia , Células Epiteliais/microbiologia , Células Epiteliais/patologia , Gastroenteropatias/metabolismo , Gastroenteropatias/microbiologia , Células Caliciformes/metabolismo , Humanos , Mucosa Intestinal/microbiologia , Mucosa Intestinal/patologia , Intestinos/microbiologia , Intestinos/patologia , Celulas de Paneth/metabolismo , Celulas de Paneth/microbiologia , Celulas de Paneth/patologia
19.
Med Sci Monit ; 24: 4433-4439, 2018 Jun 27.
Artigo em Inglês | MEDLINE | ID: mdl-29949812

RESUMO

BACKGROUND The aim of this study was to investigate the effects of TNF-α and IL-10 on the expression of ICAM-1 and CD31 in human coronary artery endothelial cells (HCAEC). MATERIAL AND METHODS HCAEC was treated with 0, 2.5 µg/l, 5 µg/l, and 10 µg/l of TNF-α for 2 h, 6 h, and 10 h, and with 0 µg/l, 10 µg/l, 100 µg/l, and 200 µg/l of IL-10 for 5 h, 10 h and 15 h, respectively. RNA inference of TNF-αR was performed with siRNA. Real-time PCR, Western blot analysis, and ELSA were used to detect the mRNA level and protein level of ICAM-1 and CD31. RESULTS TNF-α significantly increased the mRNA and protein expression of ICAM-1 (P<0.05), and 2.5 µg/l TNF-α had the most obvious effect. RNAi of TNF-aR reduced the induction of TNF-α on the mRNA and protein expression of ICAM-1 (P<0.05). TNF-α significantly decreased the CD31 in the supernatant (P<0.05), and 2.5 µg/l TNF-a had the most obvious effect. IL-10 significantly decreased the ICAM-1 protein level. IL-10 decreased the mRNA expression and the protein expression of CD31. The effect on mRNA was not significant (P>0.05), while the effect on the protein expression was significant (P<0.05). CONCLUSIONS TNF-α and IL-10 treatment can affect the expression of ICAM-1 and CD31 in HCAEC.


Assuntos
Vasos Coronários/citologia , Células Endoteliais/metabolismo , Molécula 1 de Adesão Intercelular/metabolismo , Interleucina-10/farmacologia , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo , Fator de Necrose Tumoral alfa/farmacologia , Células Endoteliais/efeitos dos fármacos , Humanos
20.
Antiviral Res ; 150: 1-8, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29203391

RESUMO

Heptad repeat (HR) regions are highly conserved motifs located in the glycoproteins of enveloped viruses that form a six-helix bundle structure and is important in the process of virus fusion. Peptides derived from the HR regions of some viruses have also been shown to inhibit viral entry. Porcine epidemic diarrhea virus (PEDV) was predicted to have HR regions (HR1 and HR2) in the spike glycoprotein S2 subunit. Based on this analysis, six peptides derived from HR1 and HR2 were selected, expressed in Escherichia coli, purified, and characterized. Three peptides (HR2M, HR2L and HR2P) were identified as potential competitive inhibitors in PEDV in vitro infection assays, with the HR2P peptide representing the most potent inhibitor. Further study indicated that immunization of HR2P in mice elicited antibodies capable of neutralizing PEDV infection in vitro. These results demonstrate that the HR2P peptide and anti-HR2P antibody can serve as a tool for dissecting the fusion mechanism of PEDV, guiding the search for potent inhibitors with therapeutic value against PEDV infection.


Assuntos
Anticorpos Antivirais/imunologia , Infecções por Coronavirus/virologia , Peptídeos/imunologia , Vírus da Diarreia Epidêmica Suína/fisiologia , Domínios e Motivos de Interação entre Proteínas , Glicoproteína da Espícula de Coronavírus/imunologia , Glicoproteína da Espícula de Coronavírus/metabolismo , Internalização do Vírus , Sequência de Aminoácidos , Animais , Anticorpos Neutralizantes/imunologia , Antivirais/farmacologia , Chlorocebus aethiops , Humanos , Testes de Neutralização , Peptídeos/química , Peptídeos/farmacologia , Domínios e Motivos de Interação entre Proteínas/imunologia , Glicoproteína da Espícula de Coronavírus/química , Células Vero , Internalização do Vírus/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA