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.
Front Pharmacol ; 15: 1397116, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38831880

RESUMO

Background: Alternol is a small molecular compound isolated from the fermentation of a mutant fungus obtained from Taxus brevifolia bark. Our previous studies showed that Alternol treatment induced reactive oxygen species (ROS)-dependent immunogenic cell death. This study conducted a comprehensive investigation to explore the mechanisms involved in Alternol-induced immunogenic cell death. Methods: Prostate cancer PC-3, C4-2, and 22RV1 were used in this study. Alternol interaction with heat shock proteins (HSP) was determined using CETSA assay. Alternol-regulated ER stress proteins were assessed with Western blot assay. Extracellular adenosine triphosphate (ATP) was measured using ATPlite Luminescence Assay System. Results: Our results showed that Alternol interacted with multiple cellular chaperone proteins and increased their expression levels, including endoplasmic reticulum (ER) chaperone hypoxia up-regulated 1 (HYOU1) and heat shock protein 90 alpha family class B member 1 (HSP90AB1), as well as cytosolic chaperone heat shock protein family A member 8 (HSPA8). These data represented a potential cause of unfolded protein response (UPR) after Alternol treatment. Further investigation revealed that Alternol treatment triggered ROS-dependent (ER) stress responses via R-like ER kinase (PERK), inositol-requiring enzyme 1α (IRE1α). The double-stranded RNA-dependent protein kinase (PKR) but not activating transcription factor 6 (ATF6) cascades, leading to ATF-3/ATF-4 activation, C/EBP-homologous protein (CHOP) overexpression, and X-box binding protein XBP1 splicing induction. In addition, inhibition of these ER stress responses cascades blunted Alternol-induced extracellular adenosine triphosphate (ATP) release, one of the classical hallmarks of immunogenic cell death. Conclusion: Taken together, our data demonstrate that Alternol treatment triggered multiple ER stress cascades, leading to immunogenic cell death.

2.
Cancer Med ; 13(3): e6736, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38204220

RESUMO

BACKGROUND: The mechanism of decreased T cells infiltrating tumor tissues in hepatocellular carcinoma is poorly understood. METHODS: Cells were separated from the single-cell RNA-sequence dataset of hepatocellular carcinoma patients (GSE149614) for cell-cell communication. Flow cytometry, EDU staining, H3-Ser28 staining, confocal immunofluorescence staining, western blotting and naked microsubcutaneous tumors were performed for the mechanism of NGF-NGFR promoting proliferation. RESULTS: The present study has revealed that during the process of T-cell infiltration from adjacent tissues to tumor tissues, an inefficiency in NGF-NGFR communication occurs in the tumor tissues. Importantly, NGF secreted by tumor cells interacts with NGFR present on the membranes of the infiltrated T cells, thereby promoting the proliferation through the activation of mitotic spindle signals. Mechanistically, the mediation of mitotic spindle signal activation promoting proliferation is executed by HDAC1-mediated inhibition of unclear trans-localization of PREX1. Furthermore, PD-1 mAb acts synergistically with the NGF-NGFR communication to suppress tumor progression in both mouse models and HCC patients. Additionally, NGF-NGFR communication was positively correlates with the PD-1/PDL-1 expression. However, expressions of NGF and NGFR are low in tumor tissues, which is responsible for the invasive clinicopathological features and the disappointing prognosis in HCC patients. CONCLUSION: Inefficiency in NGF-NGFR communication impairs PD-1 mAb immunotherapy and could thus be utilized as a novel therapeutic target in the treatment of HCC patients in clinical practice.


Assuntos
Carcinoma Hepatocelular , Neoplasias Hepáticas , Animais , Camundongos , Humanos , Carcinoma Hepatocelular/terapia , Receptor de Morte Celular Programada 1 , Neoplasias Hepáticas/terapia , Linfócitos T , Imunoterapia , Fatores de Troca do Nucleotídeo Guanina , Proteínas do Tecido Nervoso , Receptores de Fator de Crescimento Neural
3.
Biochem Biophys Res Commun ; 656: 1-9, 2023 05 14.
Artigo em Inglês | MEDLINE | ID: mdl-36940637

RESUMO

Sorafenib has been used to enhance the survival outcome of hepatocellular carcinoma (HCC) patients. But, occurrence resistance to sorafenib subtracts from its therapeutic benefits. Herein, we identified that FOXM1 was markedly upregulated in both tumor samples and sorafenib-resistant HCC tissues. We also demonstrated that patients with decreased FOXM1 expression had longer overall survival (OS) and progression-free survival (PFS) in the cohort of sorafenib-treated patients. For HCC cells resistant to sorafenib, the IC50 value of sorafenib and the expression of FOXM1 were increased. In addition, Downregulation of FOXM1 expression alleviated the occurrence of resistance to sorafenib and reduced the proliferative potential and viability of HCC cells. Mechanically, the suppression of the FOXM1 gene resulted in the downregulation of KIF23 levels. Moreover, downregulation of FOXM1 expression reduced the levels of RNA polymerase II (RNA pol II) and histone H3 lysine 27 acetylation (H3K27ac) on the KIF23 promoter, further epigenetically silencing the production of KIF23. More intriguingly, our results similarly revealed that FDI-6, a specific inhibitor of FOXM1, suppressed the proliferation of HCC cells resistant to sorafenib, as well as upregulation of FOXM1 or KIF23 abolished this effect. In addition, we found that FDI-6 combined with sorafenib significantly improved the therapeutic effect of sorafenib. Collectively, the present results revealed that FOXM augments sorafenib resistance and enhances HCC progression by upregulating KIF23 expression via an epigenetic mechanism, and targeting FOXM1 can be an effective treatment for HCC.


Assuntos
Carcinoma Hepatocelular , Neoplasias Hepáticas , Humanos , Carcinoma Hepatocelular/tratamento farmacológico , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Sorafenibe/farmacologia , Sorafenibe/uso terapêutico , Neoplasias Hepáticas/tratamento farmacológico , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Regulação para Cima , Ativação Transcricional , Resistencia a Medicamentos Antineoplásicos/genética , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica , Proliferação de Células , Proteína Forkhead Box M1/genética , Proteína Forkhead Box M1/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo
4.
J Invest Surg ; 36(1): 2164813, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-36603844

RESUMO

BACKGROUND: Hepatic ischemia-reperfusion injury (IRI) is a common innate immune-mediated sterile inflammatory response in liver transplantation and liver tumor resection. Neutrophil extracellular traps (NETs) can aggravate liver injury and activates innate immune response in the process of liver IRI. However, Curcumin (Cur) can reverse this damage and reduce NETs formation. Nevertheless, the specific regulatory mechanism is still unclear in liver IRI. This study aimed to explore the potential mechanisms that how does Cur alleviate hepatic IRI by inhibits NETs production and develop novel treatment regimens. METHODS: We established a hepatic IRI model by subjecting C57BL/6J mice to 60 min of ischemia, followed by reperfusion for 2 h, 6 h, 12 h, and 24 h respectively. Subsequently, we were separated into 5 groups, namely the I/R group, Cur group, DNase-1 group, Cur + DNase1 group and sham operation group. Serum alanine aminotransferase (ALT) and aspartate transaminase (AST), Hematoxylin-eosin staining, immunofluorescence, and TUNEL analysis were applied to assess liver injury degree and NETs levels. Western blot assay was used to detect the protein levels of apoptosis-related proteins and MEK pathway proteins. RESULTS: Cur could alleviate hepatic IRI by inhibiting the generation of NETs via suppressing the MEK/ERK pathway. In addition, this study also revealed that DNase-1 is vital for alleviating hepatic IRI by reducing the generation of NETs. CONCLUSIONS: Cur combined with DNase-1 was more effective than the two drugs administered alone in alleviating hepatic IRI by inhibiting the generation of NETs. These results also suggested that curcumin combined with DNase-1 was a potential therapeutic strategy to mitigate hepatic IRI.


Assuntos
Curcumina , Armadilhas Extracelulares , Traumatismo por Reperfusão , Camundongos , Animais , Curcumina/farmacologia , Curcumina/uso terapêutico , Curcumina/metabolismo , Armadilhas Extracelulares/metabolismo , Camundongos Endogâmicos C57BL , Fígado/patologia , Traumatismo por Reperfusão/tratamento farmacológico , Inflamação , Desoxirribonucleases/metabolismo , Desoxirribonucleases/farmacologia , Desoxirribonucleases/uso terapêutico , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Quinases de Proteína Quinase Ativadas por Mitógeno/farmacologia , Quinases de Proteína Quinase Ativadas por Mitógeno/uso terapêutico
5.
Cancer Gene Ther ; 29(11): 1616-1627, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-35504951

RESUMO

The Apoptosis Stimulating Protein of p53 2 (ASPP2) is a heterozygous insufficient tumor suppressor; however, its molecular mechanism(s) in tumor suppression is not completely understood. ASPP2 plays an essential role in cell growth, as shown by liver hepatocellular carcinoma (LIHC) RNA-seq assay using the Cancer Genome Atlas (TCGA) and High-Throughput-PCR assay using ASPP2 knockdown cells. These observations were further confirmed by in vivo and in vitro experiments. Mechanistically, N-terminus ASPP2 interacted with Keratin 18 (k18) in vivo and in vitro. Interestingly, the RDIVpSGP motif of ASPP2 associates with 14-3-3 and promotes ASPP2/k18/14-3-3 ternary-complex formation which promotes MEK/ERK signal activation by impairing 14-3-3 and BRAF association. Additionally, ASPP2-rAd injection promotes paclitaxel-suppressed tumor growth by suppressing cell proliferation in the BALB/c nude mice model. ASPP2 and k18 were preferentially downregulated in Hepatocellular Carcinoma (HCC), which predicted poor prognosis in HCC patients. Overall, these findings suggested that ASPP2 promoted BRAF/MEK/ERK signal activation by promoting the formation of an ASPP2/k18/14-3-3 ternary complex via the RDIVpSGP motif at the N terminus. Moreover, this study provides novel insights into the molecular mechanism of tumor suppression in HCC patients.


Assuntos
Carcinoma Hepatocelular , Neoplasias Hepáticas , Camundongos , Animais , Carcinoma Hepatocelular/patologia , Proteína Supressora de Tumor p53/metabolismo , Proteínas Proto-Oncogênicas B-raf , Neoplasias Hepáticas/metabolismo , Queratina-18/metabolismo , Camundongos Nus , Proteínas 14-3-3/genética , Proteínas 14-3-3/metabolismo , Proteínas Reguladoras de Apoptose/metabolismo , Proliferação de Células , Apoptose , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Linhagem Celular Tumoral
6.
J Healthc Eng ; 2022: 4470510, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35399855

RESUMO

Rare Earth up-conversion nanoparticles NaYF4:20%Yb,2%Er@PEI (UCNPs) were generated via a one-step hydrothermal technique at relatively reduced temperatures. Photosensitizer Ce6 and anti-EpCAM, a highly expressed monoclonal antibody in cancer stem cells of hepatocellular carcinoma, were linked to UCNP surfaces via the formation of amide linkage between carboxyl from Ce6 or anti-EpCAM and abundant amino from PEI, leading to the formation of Ps-Ce6 and anti-EpCAM-UCNPs-Ce6 nanoparticles. The synthesized nanoparticles characterized by XRD, TEM, and IR, and their zeta potential, ROS generation ability, Ce6 loading rate, and up-conversion fluorescence properties were investigated. It has been revealed that all the products were uniformly dispersed nanoparticles (25-32 nm), which crystallized primarily as hexagonal structures, and their up-conversion fluorescence spectra were similar to that of NaYF4:20%Yb,2%Er. The Ce6 loading rate in the anti-EpCAM-UCNPs-Ce6 nanoparticles was about 2.9%, thereby resulting in good ROS generation ability. For anti-EpCAM-UCNPs-Ce6, the biosafety, targeting effect, and PDT effect exposed under near-infrared (NIR) laser (980 nm) were evaluated using human liver cancer cells (BEL-7404). The results showed that it has good biocompatibility and biosafety as well as high targeting and PDT treatment efficiencies, which renders it a potential experimental material for the near-infrared PDT study.


Assuntos
Carcinoma Hepatocelular , Neoplasias Hepáticas , Nanopartículas , Fotoquimioterapia , Carcinoma Hepatocelular/tratamento farmacológico , Linhagem Celular Tumoral , Humanos , Neoplasias Hepáticas/tratamento farmacológico , Nanopartículas/química , Nanopartículas/uso terapêutico , Fotoquimioterapia/métodos , Fármacos Fotossensibilizantes/química , Fármacos Fotossensibilizantes/uso terapêutico , Espécies Reativas de Oxigênio
7.
Cell Death Dis ; 13(3): 213, 2022 03 07.
Artigo em Inglês | MEDLINE | ID: mdl-35256589

RESUMO

Tumor heterogeneity has been associated with immunotherapy and targeted drug resistance in hepatocellular carcinoma (HCC). However, communications between tumor and cytotoxic cells are poorly understood to date. In the present study, thirty-one clusters of cells were discovered in the tumor tissues and adjacent tissues through single-cell sequencing. Moreover, the quantity and function exhaustion of cytotoxic cells was observed to be induced in tumors by the TCR and apoptosis signal pathways. Furthermore, granzyme failure of cytotoxic cells was observed in HCC patients. Importantly, the GZMA secreted by cytotoxic cells was demonstrated to interact with the F2R expressed by the tumor cells both in vivo and in vitro. This interaction induced tumor suppression and T cell-mediated killing of tumor cells via the activation of the JAK2/STAT1 signaling pathway. Mechanistically, the activation of JAK2/STAT1 signaling promoted apoptosis under the mediating effect of the LDPRSFLL motif at the N-terminus of F2R, which interacted with GZMA. In addition, GZMA and F2R were positively correlated with PD-1 and PD-L1 in tumor tissues, while the expressions of F2R and GZMA promoted PD-1 mAb-induced tumor suppression in both mouse model and HCC patients. Finally, in HCC patients, a low expression of GZMA and F2R in the tumor tissues was correlated with aggressive clinicopathological characteristics and poor prognosis. Collectively, GZMA-F2R communication inefficient induces deficient PD-1 mAb therapy and provide a completely novel immunotherapy strategy for tumor suppression in HCC patients.


Assuntos
Carcinoma Hepatocelular , Neoplasias Hepáticas , Animais , Antígeno B7-H1/metabolismo , Carcinoma Hepatocelular/metabolismo , Linhagem Celular Tumoral , Granzimas/metabolismo , Humanos , Imunoterapia , Janus Quinase 2/metabolismo , Neoplasias Hepáticas/metabolismo , Camundongos , Receptor de Morte Celular Programada 1/metabolismo , Fator de Transcrição STAT1/metabolismo , Linfócitos T/metabolismo
8.
Biochem Biophys Res Commun ; 585: 185-190, 2021 12 31.
Artigo em Inglês | MEDLINE | ID: mdl-34808502

RESUMO

Hepatocellular carcinoma (HCC) is a prevalent solid cancer worldwide and sorafenib is a common treatment. Nevertheless, sorafenib resistance is a severe clinical problem. In the present study, we identified that epigenetic regulator, KAT6A, was overexpressed in clinical HCC tissues and sorafenib-resistant HCC samples. The depletion of KAT6A repressed the cell viability and Edu-positive cell numbers of HCC cells. The IC50 value of sorafenib was increased in sorafenib-resistant HCC cells. In addition, the expression of KAT6A was induced in sorafenib-resistant HCC cells. The depletion of KAT6A suppressed the IC50 of sorafenib. Mechanically, YAP was decreased by the depletion of KAT6A. KAT6A was able to enrich in the promoter of YAP. The silencing of KAT6A reduced the enrichment of histone H3 lysine 23 acetylation (H3K23ac) and RNA polymerase II (RNA pol II) on the promoter of YAP in sorafenib-resistant HCC cells. KAT6A inhibitor WM-1119 repressed the cell proliferation of sorafenib-resistant HCC cells, while overexpression of KAT6A or YAP could reverse the effect in the cells. Meanwhile, the treatment of sorafenib inhibited the viability of sorafenib-resistant HCC cells, while the co-treatment of WM-1119 could improve the effect of sorafenib. Collectively, KAT6A was associated with sorafenib resistance and contributes to progression of HCC by targeting YAP. Targeting KAT6A may be served as a promising therapeutic approach for HCC.


Assuntos
Carcinoma Hepatocelular/genética , Proteínas de Ciclo Celular/genética , Resistencia a Medicamentos Antineoplásicos/genética , Regulação Neoplásica da Expressão Gênica , Histona Acetiltransferases/genética , Neoplasias Hepáticas/genética , Sorafenibe/farmacologia , Fatores de Transcrição/genética , Antineoplásicos/farmacologia , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patologia , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular/genética , Progressão da Doença , Epigênese Genética , Células Hep G2 , Histona Acetiltransferases/metabolismo , Humanos , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Transcrição/metabolismo
9.
J Neuroimmunol ; 350: 577438, 2020 Nov 07.
Artigo em Inglês | MEDLINE | ID: mdl-33197842

RESUMO

Neurological complications are very common after liver transplantation. This study focuses on clinical risk factors and susceptibility gene polymorphisms of neurological complications after liver transplantation. A better predictive model is obtained. This study proves that MTRR is an independent susceptibility gene for neurological complications. Compared with the independent risk factor of abdominal infection, MTRR has a more advantageous value in predicting neurological complications after liver transplantation.

10.
Zhonghua Nan Ke Xue ; 23(3): 276-279, 2017 Mar.
Artigo em Chinês | MEDLINE | ID: mdl-29706052

RESUMO

The genitofemoral nerve (GFN) has its unique anatomic characteristics of location, run and function in the male urinary system and its relationship with the ureter, deferens and inguinal region is apt to be ignored in clinical anatomic application. Clinical studies show that GFN is closely correlated with postoperative ureteral complications and pain in the inguinal region after spermatic cord or hernia repair. GFN transplantation can be used in the management of erectile dysfunction caused by cavernous nerve injury. Therefore, GFN played an important role in the clinical application of uroandrology. This review summarizes the advances in the studies of GFN in relation to different diseases in uroandrology.


Assuntos
Disfunção Erétil/cirurgia , Hérnia Inguinal/cirurgia , Plexo Lombossacral/lesões , Complicações Pós-Operatórias/cirurgia , Sistema Urogenital/inervação , Disfunção Erétil/etiologia , Humanos , Canal Inguinal/inervação , Masculino , Dor Pós-Operatória/etiologia , Nervos Periféricos/transplante , Complicações Pós-Operatórias/etiologia , Ureter/inervação , Ureter/cirurgia , Ducto Deferente/inervação
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...