Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
J Exp Clin Cancer Res ; 43(1): 136, 2024 May 06.
Article in English | MEDLINE | ID: mdl-38711082

ABSTRACT

BACKGROUND: Hepatocellular carcinoma (HCC) is a fatal malignancy with poor prognosis due to lack of effective clinical interference. DCAF1 plays a vital role in regulating cell growth and proliferation, and is involved in the progression of various malignancies. However, the function of DCAF1 in HCC development and the underlying mechanism are still unknown. This study aimed to explore the effect of DCAF1 in HCC and the corresponding molecular mechanism. METHODS: Quantitative real-time PCR, Western blot and immunostaining were used to determine DCAF1 expression in tumor tissues and cell lines. Subsequently, in vitro and in vivo experiments were conducted to explore the function of DCAF1 in tumor growth and metastasis in HCC. Coimmunoprecipitation, mass spectrometry and RNA sequencing were performed to identify the underlying molecular mechanisms. RESULTS: In this study, we found that DCAF1 was observably upregulated and associated with poor prognosis in HCC. Knockdown of DCAF1 inhibited tumor proliferation and metastasis and promoted tumor apoptosis, whereas overexpressing DCAF1 yielded opposite effects. Mechanistically, DCAF1 could activate the Akt signaling pathway by binding to PARD3 and enhancing its expression. We also found that the combined application of DCAF1 knockdown and Akt inhibitor could significantly suppress subcutaneous xenograft tumor growth. CONCLUSIONS: Our study illustrates that DCAF1 plays a crucial role in HCC development and the DCAF1/PARD3/Akt axis presents a potentially effective therapeutic strategy for HCC.


Subject(s)
Carcinoma, Hepatocellular , Disease Progression , Liver Neoplasms , Neoplasm Metastasis , Proto-Oncogene Proteins c-akt , Signal Transduction , Animals , Female , Humans , Male , Mice , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , Apoptosis , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/genetics , Cell Line, Tumor , Cell Proliferation , Gene Expression Regulation, Neoplastic , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Liver Neoplasms/genetics , Mice, Nude , Prognosis , Proto-Oncogene Proteins c-akt/metabolism , Xenograft Model Antitumor Assays
2.
Org Lett ; 25(51): 9181-9185, 2023 Dec 29.
Article in English | MEDLINE | ID: mdl-38117207

ABSTRACT

We report the development of a phosphonium salt as a remarkable activating agent that enables the direct conversion of C2-H bonds of a variety of heterocyclic N-oxides into C2-N, C2-O, or C2-S bonds with high efficiency. The phosphonium salt was prepared on a >150 g scale in a single step and is tolerant of multiple functionalities.

3.
J Hepatol ; 79(2): 403-416, 2023 08.
Article in English | MEDLINE | ID: mdl-37040844

ABSTRACT

BACKGROUND & AIMS: Non-alcoholic steatohepatitis (NASH) is a chronic inflammatory disease that can further progress to cirrhosis and hepatocellular carcinoma. However, the key molecular mechanisms behind this process have not been clarified. METHODS: We analyzed human NASH and normal liver tissue samples by RNA-sequencing and liquid chromatography-mass spectrometry, identifying hepatocyte cytosolic protein Myc-interacting zinc-finger protein 1 (Miz1) as a potential target in NASH progression. We established a Western diet+fructose-induced NASH model in hepatocyte-specific Miz1 knockout and adeno-associated virus type 8-overexpressing mice. Human NASH liver organoids were used to confirm the mechanism, and immunoprecipitation and mass spectrometry were used to detect proteins that could interact with Miz1. RESULTS: We demonstrate that Miz1 is reduced in hepatocytes in human NASH. Miz1 is shown to bind to peroxiredoxin 6 (PRDX6), retaining it in the cytosol, blocking its interaction with mitochondrial Parkin at Cys431, and inhibiting Parkin-mediated mitophagy. In NASH livers, loss of hepatocyte Miz1 results in PRDX6-mediated inhibition of mitophagy, increased dysfunctional mitochondria in hepatocytes, and production of proinflammatory cytokines, including TNFα, by hepatic macrophages. Crucially, the increased production of TNFα results in a further reduction in hepatocyte Miz1 by E3-ubiquitination. This produces a positive feedback loop of TNFα-mediated hepatocyte Miz1 degradation, resulting in PRDX6-mediated inhibition of hepatocyte mitophagy, with the accumulation of dysfunctional mitochondria in hepatocytes and increased macrophage TNFα production. CONCLUSIONS: Our study identified hepatocyte Miz1 as a suppressor of NASH progression via its role in mitophagy; we also identified a positive feedback loop by which TNFα production induces degradation of cytosolic Miz1, which inhibits mitophagy and thus leads to increased macrophage TNFα production. Interruption of this positive feedback loop could be a strategy to inhibit the progression of NASH. IMPACT AND IMPLICATIONS: Non-alcoholic steatohepatitis (NASH) is a chronic inflammatory disease that can further develop into cirrhosis and hepatocellular carcinoma. However, the key molecular mechanism of this process has not been fully clarified. Herein, we identified a positive feedback loop of macrophage TNFα-mediated hepatocyte Miz1 degradation, resulting in PRDX6-mediated inhibition of hepatocyte mitophagy, aggravation of mitochondrial damage and increased macrophage TNFα production. Our findings not only provide mechanistic insight into NASH progression but also provide potential therapeutic targets for patients with NASH. Our human NASH liver organoid culture is therefore a useful platform for exploring treatment strategies for NASH development.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Non-alcoholic Fatty Liver Disease , Humans , Animals , Mice , Non-alcoholic Fatty Liver Disease/metabolism , Carcinoma, Hepatocellular/pathology , Tumor Necrosis Factor-alpha/metabolism , Mitophagy , Feedback , Hepatocytes/metabolism , Liver/pathology , Liver Cirrhosis/pathology , Liver Neoplasms/pathology , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Mice, Inbred C57BL , Disease Models, Animal , Protein Inhibitors of Activated STAT/metabolism , Protein Inhibitors of Activated STAT/therapeutic use
4.
J Affect Disord ; 327: 155-158, 2023 04 14.
Article in English | MEDLINE | ID: mdl-36731542

ABSTRACT

BACKGROUND: Stress and marital dissatisfaction have been identified as risk factors for depression in separate lines of research. However, the interaction between stress and marital satisfaction in predicting depression over time has rarely been examined, despite the fact that marital satisfaction may weaken (i.e., buffer) the impact of stress on depression. This longitudinal study evaluated marital satisfaction as a moderator of the association between stress and depressive symptoms in a probability sample of American married adults. METHODS: Married respondents from Wave I and Wave II of the Americans' Changing Lives (ACL) study (N = 1392) completed measures of marital satisfaction, stressful life events, and depressive symptoms at baseline and three-year follow-up. RESULTS: Marital satisfaction and stressful life events were significantly associated with depressive symptoms in cross-sectional analyses and uniquely predicted depressive symptoms three years later, controlling for prior depressive symptoms. However, marital satisfaction did not moderate the association between stressful life events and depression. A sensitivity analysis of data from Wave IV and Wave V of the ACL yielded similar findings, supporting the replicability of the results. LIMITATIONS: Broader assessment on stressful life events and assessment of perceived stress would provide a stronger test of the association between stress and depression as well as the degree to which this association is moderated by marital satisfaction. CONCLUSIONS: Attending to both stress and marital satisfaction may provide a more comprehensive understanding of risk for depression than exclusive focusing on either stress or marital satisfaction, which may have beneficial implications for preventing and treating depression.


Subject(s)
Depression , Marriage , Adult , Humans , Depression/diagnosis , Longitudinal Studies , Cross-Sectional Studies , Personal Satisfaction
5.
Immunology ; 169(1): 69-82, 2023 05.
Article in English | MEDLINE | ID: mdl-36420610

ABSTRACT

Hepatocellular carcinoma (HCC) is one of the most fatal tumours worldwide and has a high recurrence rate. Nevertheless, the mechanism of HCC genesis remains partly unexplored, while the efficiency of HCC treatments remains limited. The present study analysed the expression of nuclear receptor subfamily 4 group A member 1 (NR4A1) in tumour-infiltrating natural killer (NK) cells derived from both human patients with HCC and tumour-bearing mouse models, as well as the features of NR4A1high and NR4A1low NK cells. In addition, knockout of NR4A1 by CRISPR/Cas9 and adoptive transfer experiments were applied to verify the function of NR4A1 in both tumour-infiltrating NK cells and anti-PD-1 therapy. The present study found that NR4A1 was significantly highly expressed in tumour-infiltrating NK cells, which mediated the dysfunction of tumour-infiltrating NK cells by regulating the IFN-γ/p-STAT1/IRF1 signalling pathway. Knockout of NR4A1 in NK cells not only restored the antitumour function of NK cells but also enhanced the efficacy of anti-PD-1 therapy. The present findings suggest a regulatory role of NR4A1 in the immune progress of NK cells against HCC, which may provide a new direction for immunotherapies of HCC.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Humans , Mice , Animals , Carcinoma, Hepatocellular/pathology , Liver Neoplasms/pathology , Mice, Knockout , Killer Cells, Natural , Immunotherapy , STAT1 Transcription Factor/genetics , STAT1 Transcription Factor/metabolism , Interferon Regulatory Factor-1/genetics , Interferon Regulatory Factor-1/metabolism , Nuclear Receptor Subfamily 4, Group A, Member 1/genetics , Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism
6.
Adv Mater ; 34(38): e2203019, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35918816

ABSTRACT

Chemoimmunotherapy has shown great potential to activate an immune response, but the immunosuppressive microenvironment associated with T cell exhaustion remains a challenge in cancer therapy. The proper immune-modulatory strategy to provoke a robust immune response is to simultaneously regulate T-cell exhaustion and infiltration. Here, a new kind of carrier-free nanoparticle is developed to simultaneously deliver chemotherapeutic drug (doxorubicin, DOX), cytolytic peptide (melittin, MPI), and anti-TOX small interfering RNA (thymocyte selection-associated high mobility group box protein, TOX) using a fluorinated prodrug strategy. In this way, the enhanced immunogenic cell death (ICD) induced by the combination of DOX and MPI can act as "offense" signaling to increase CD8+ T-cell infiltration, while the decreased TOX expression interfered with siTOX can serve as "defense" signaling to mitigate CD8+ T-cell exhaustion. As a result, the integration of DOX, MPI, and siTOX in such a bifunctional system produced a potent antitumor immune response in liver cancer and metastasis, making it a promising delivery platform and effective strategy for converting "cold" tumors into "hot" ones.


Subject(s)
Liver Neoplasms , Neoplasms , Prodrugs , Cell Line, Tumor , Doxorubicin/pharmacology , Humans , Immunotherapy , Melitten/pharmacology , Neoplasms/drug therapy , Neoplasms/metabolism , Prodrugs/pharmacology , RNA, Small Interfering/genetics , Tumor Microenvironment
8.
Spectrochim Acta A Mol Biomol Spectrosc ; 268: 120686, 2022 Mar 05.
Article in English | MEDLINE | ID: mdl-34890874

ABSTRACT

A series of WO3-x thin films with defects were obtained by thermal treatments from laser irradiation and annealing, respectively. The corresponding tunability of localized surface plasmon resonance properties and the enhancement of Raman scattering intensity were realized due to the defects in the WO3-x thin films after thermal treatments. With the changes of either laser power or annealing temperature, the crystalline quality of WO3-x thin film was declined with a red shift of the surface plasmon resonance wavelength from 464 nm to 482 nm. The as-treated WO3-x film shows good uniformity and reproducibility in Surface-enhanced Raman spectroscopy measurement, the detection limit for dye methylene blue can reach 10-8 mol/L, and enhancement factor is 1.38 × 106. Furthermore, the simulation result of finite-difference time-domain showed a substantial agreement with experimental results.

10.
Theranostics ; 10(12): 5290-5304, 2020.
Article in English | MEDLINE | ID: mdl-32373213

ABSTRACT

Rationale: Inflammation plays a crucial role in the progression of nonalcoholic steatohepatitis (NASH). Protein tyrosine phosphatase receptor type O truncated isoform (PTPROt) is an integral membrane protein that has been identified in osteoclasts, macrophages, and B lymphocytes. However, its relationship between inflammation and NASH is largely unknown. Herein, we aimed to study the function of PTPROt in NASH progression. Methods: We established a NASH mouse model in wild-type (WT), PTPRO knockout mice by western diet (WD) and methionine-choline-deficient diet (MCD). In addition, MCD-induced NASH model was established in BMT mice. Moreover, we determined the expression of PTPROt in liver macrophages in human subjects without steatosis, with simple steatosis, and with NASH to confirm the relationship between PTPROt and NASH. In vitro assays were also performed to study the molecular role of PTPROt in NASH progression. Results: Human samples and animal model results illustrated that PTPROt is increased in liver macrophages during NASH progression and is positively correlated with the degree of NASH. Our animal model also showed that PTPROt in liver macrophages can enhance the activation of the NF-κB signaling pathway, which induces the transcription of genes involved in the inflammatory response. Moreover, PTPROt promotes the transcription of pro-oxidant genes and inhibits antioxidant and protective genes via increased activation of the NF-κB signaling pathway, thereby causing an increased level of reactive oxygen species (ROS) and damaged mitochondria. This triggers the NLRP3-IL1ß axis and causes a heightened inflammatory response. Notably, PTPROt partially limits inflammation and ROS production by promoting mitophagy, which participates in a negative feedback loop in this model. Conclusions: Our data strongly indicate that PTPROt plays a dual role in inflammation via the NF-κB signaling pathway in liver macrophages during NASH. Further studies are required to explore therapeutic strategies and prevention of this common liver disease through PTPROt.


Subject(s)
Bone Marrow Transplantation/methods , NF-kappa B/metabolism , Non-alcoholic Fatty Liver Disease/metabolism , Animals , Cells, Cultured , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Fluorescent Antibody Technique , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Electron, Transmission , Signal Transduction/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...