Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 31
1.
Biol Direct ; 19(1): 15, 2024 02 23.
Article En | MEDLINE | ID: mdl-38388961

BACKGROUND: Lenvatinib is an oral small molecule inhibitor approved for treating patients with unresectable hepatocellular carcinoma (HCC) worldwide. Increasing cell sensitivity to lenvatinib would be an effective method of improving therapeutic efficacy. METHODS: High throughput methods was used to scan the differentially expressed genes (DEGs) related to lenvatinib sensitivity in HCC cells. Gain- and loss-function experiments were used to explore the functions of these DEGs in HCC and lenvatinib sensitivity. CO-IP assay and rescue experiments were utilized to investigate the mechanism. RESULTS: We identified that RAR responder protein 1 (RARRES1), a podocyte-specific growth arrest gene, was among significantly upregulated DEGs in HCC cells following lenvatinib treatment. Functional analysis showed that ectopic RARRES1 expression decreased HCC progression in vitro and in vivo, as well as improving tumor sensitivity to lenvatinib, while RARRES1 silencing increased HCC cell proliferation and migration. Mechanistically, co-immunoprecipitation assays demonstrated that RARRES1 interacted with serine protease inhibitor Kazal-type 2 (SPINK2) in HCC cells. Further, SPINK2 overexpression suppressed HCC cell proliferation and migration, as well as increasing sensitivity to lenvatinib whereas SPINK2 knockdown promoted cell progression and decreased lenvatinib sensitivity. The mRNA and protein levels of RARRES1 and SPINK2 were low in HCC tissue samples, relative to those in normal liver tissue. CONCLUSIONS: Our findings highlighted that RARRES1 can inhibit HCC progression and regulate HCC sensitivity to lenvatinib by interacting SPINK2, representing a new tumor suppressor RARRES1/SPINK2 axis in HCC that modulates sensitivity to lenvatinib.


Carcinoma, Hepatocellular , Liver Neoplasms , Quinolines , Humans , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/genetics , Liver Neoplasms/drug therapy , Liver Neoplasms/genetics , Serine Proteinase Inhibitors/therapeutic use , Phenylurea Compounds/pharmacology , Phenylurea Compounds/therapeutic use , Cell Line, Tumor , Membrane Proteins/metabolism
2.
Funct Integr Genomics ; 24(1): 8, 2024 Jan 10.
Article En | MEDLINE | ID: mdl-38200280

Members of the E26 transformation-specific (ETS) variant transcription factor family act as either tumor suppressors or oncogenic factors in numerous types of cancer. ETS variant transcription factor 7 (ETV7) participates in the development of malignant tumors, whereas its involvement in colorectal cancer (CRC) is less clear. In this study, The Cancer Genome Atlas (TCGA) and immunochemistry staining were applied to check the clinical relevance of ETV7 and interferon-induced protein with tetratricopeptide repeats 3 (IFIT3) in CRC patients. Overexpression and knockdown of ETV7 and IFIT3 were conducted by transfecting the cells with pCDNA3.1 plasmids and siRNAs, respectively. Western blotting was used to detect the protein expression of ETV7 in CRC cells. Cell Counting Kit-8, cell colony formation, and Transwell assays, as well as flow cytometry, were used to evaluate the proliferation, migration, cell cycle, and apoptosis of CRC cells. Furthermore, western blotting, RT-qPCR, and luciferase assay were used to explore the regulation of ETV7 on IFIT3. Rescue assay was used to investigate the significance of ETV7/IFIT3 axis on CRC progression. We found that ETV7 was upregulated in CRC tissues and cells. Overexpression of ETV7 stimulated the proliferation, migration, and cell cycle amplification, and reduced the apoptosis of CRC cells. Downregulation of ETV7 exerted the opposite effect on CRC cell progression. Moreover, we demonstrated that ETV7 stimulated the transcription activity, the mRNA and protein expression of IFIT3 in CRC cells. There was a positive correlation between ETV7 and IFIT3 in CRC patients. IFIT3 knockdown reversed the promotive effect exerted by overexpression of ETV7 on the amplification and migration of CRC cells. By contrast, overexpression of IFIT3 blocked the inhibitory effect of ETV7-targeting siRNA. In summary, ETV7 induces progression of CRC by activating the transcriptional expression of IFIT3. The EVT7/IFIT3 axis may be a novel target for CRC therapy.


Apoptosis , Colorectal Neoplasms , Humans , Up-Regulation , Down-Regulation , Apoptosis/genetics , Colorectal Neoplasms/genetics , Proto-Oncogene Proteins c-ets , Intracellular Signaling Peptides and Proteins
3.
Altern Ther Health Med ; 30(2): 171-177, 2024 Feb.
Article En | MEDLINE | ID: mdl-37856812

Tumor necrosis factor alpha-induced protein-3, also called A20, is a zinc-finger protein that participates in various inflammatory responses; however, the putative relationship between A20 and hepatic fibrosis remains unelucidated. Therefore, we investigated the role and mechanism of action of A20 in activating hepatic stellate cells (HSC) during the progression of hepatic fibrosis. Cell counting kit-8 (CCK8), colony growth, transwell assays, cell cycle analysis, and apoptosis assays were performed to explore the effect of A20 on cell function in vitro. An interspecies intravenous injection of the adeno-associated virus was used to assess the in vivo role of A20. The regulation of A20 on p65 was detected using mass spectrometry and immunoprecipitation. Our findings revealed that A20 was highly expressed in the liver tissues of patients with hepatic fibrosis and that the expression level of A20 in the liver tissue was closely correlated with the stage of liver fibrosis. In the LX-2 cell line, the downregulation of A20 upregulated the expression of fibrosis-related proteins and increased the expression of inflammatory factors, indicating the activation of HSC and vice versa. In addition, overexpression of A20 in mice reduced the degree of liver fibrosis in thioacetamide model mice. Finally, co-immunoprecipitation demonstrated that A20 could interact with p65. Hence, A20 inhibits HSC activation by binding to p65.


NF-kappa B , Tumor Necrosis Factor-alpha , Humans , Mice , Animals , NF-kappa B/metabolism , NF-kappa B/pharmacology , Tumor Necrosis Factor alpha-Induced Protein 3/metabolism , Tumor Necrosis Factor alpha-Induced Protein 3/pharmacology , Tumor Necrosis Factor-alpha/pharmacology , Hepatic Stellate Cells/metabolism , Hepatic Stellate Cells/pathology , Signal Transduction , Liver Cirrhosis/chemically induced , Liver Cirrhosis/pathology
4.
Acad Radiol ; 2023 Dec 06.
Article En | MEDLINE | ID: mdl-38061942

RATIONALE AND OBJECTIVES: An accurate prognostic model is essential for the development of treatment strategies for gallbladder cancer (GBC). This study proposes an integrated model using clinical features, radiomics, and deep learning based on contrast-enhanced computed tomography (CT) images for survival prediction in patients with GBC after surgical resection. METHODS: A total of 167 patients with GBC who underwent surgical resection at two medical institutions were retrospectively enrolled. After obtaining the pre-treatment CT images, the tumor lesions were manually segmented, and handcrafted radiomics features were extracted. A clinical prognostic signature and radiomics signature were built using machine learning algorithms based on the optimal clinical features or handcrafted radiomics features, respectively. Subsequently, a DenseNet121 model was employed for transfer learning on the radiomics image data and as the basis for the deep learning signature. Finally, we used logistic regression on the three signatures to obtain the unified multimodal model for comprehensive interpretation and analysis. RESULTS: The integrated model performed better than the other models, exhibiting the highest area under the curve (AUC) of 0.870 in the test set, and the highest concordance index (C-index) of 0.736 in predicting patient survival rates. A Kaplan-Meier analysis demonstrated that patients in high-risk group had a lower survival probability compared to those in low-risk group (log-rank p < 0.05). CONCLUSION: The nomogram is useful for predicting the survival of patients with GBC after surgical resection, helping in the identification of high-risk patients with poor prognosis and ultimately facilitating individualized management of patients with GBC.

5.
World J Gastrointest Surg ; 15(8): 1825-1830, 2023 Aug 27.
Article En | MEDLINE | ID: mdl-37701685

BACKGROUND: Embryonic hepatic artery anatomy simplifies its identification during liver transplantation. Injuries to the donor hepatic artery can cause complications in this process. The hepatic artery's complex anatomy in adults makes this step challenging; however, during embryonic development, the artery and its branches have a simpler relationship. By restoring the embryonic hepatic artery anatomy, surgeons can reduce the risk of damage and increase the procedure's success rate. This approach can lead to improved patient outcomes and lower complication rates. CASE SUMMARY: In this study, we report a case of donor liver preparation using a donor hepatic artery preparation based on human embryology. During the preparation of the hepatic artery, we restored the anatomy of the celiac trunk, superior mesenteric artery, and their branches to the state of the embryo at 5 wk. This allowed us to dissect the variant hepatic artery from the superior mesenteric artery and left gastric artery during the operation. After implanting the donor liver into the recipient, we observed normal blood flow in the donor hepatic artery, main hepatic artery, and variant hepatic artery, without any leakage. CONCLUSION: Donor hepatic artery preparation based on human embryology can help reduce the incidence of donor hepatic artery injuries during liver transplantation.

6.
Front Microbiol ; 14: 1174740, 2023.
Article En | MEDLINE | ID: mdl-37350783

Compared to traditional herbage, functional native herbage is playing more important role in ruminant agriculture through improving digestion, metabolism and health of livestock; however, their effects on rumen microbial communities and hindgut fermentation are still not well understood. The objective of present study was to evaluate the effects of dietary addition of Allium mongolicum on bacterial communities in rumen and feces of claves. Sixteen 7-month-old male calves were randomly divided into four groups (n = 4). All calves were fed a basal ration containing roughage (alfalfa and oats) and mixed concentrate in a ratio of 60:40 on dry matter basis. In each group, the basal ration was supplemented with Allium mongolicum 0 (SL0), 200 (SL200), 400 (SL400), and 800 (SL800) mg/kg BW. The experiment lasted for 58 days. Rumen fluid and feces in rectum were collected, Rumen fluid and hindgut fecal were collected for analyzing bacterial community. In the rumen, Compared with SL0, there was a greater relative abundance of phylum Proteobacteria (p < 0.05) and genera Rikenellaceae_RC9_gut_group (p < 0.01) in SL800 treatment. In hindgut, compared with SL0, supplementation of A. mongolicum (SL200, SL400, or SL800) decreased in the relative abundances of Ruminococcaceae_UCG-014 (p < 0.01), Ruminiclostridium_5 (p < 0.01), Eubacterium_coprostanoligenes_group (p < 0.05), and Alistipes (p < 0.05) in feces; Whereas, the relative abundances of Christensenellaceae_R-7_group (p < 0.05), and Prevotella_1 (p < 0.01) in SL800 were higher in feces, to maintain hindgut stability. This study provided evidence that A. mongolicum affects the gastrointestinal of calves, by influencing microbiota in their rumen and feces.

7.
Heliyon ; 9(2): e13691, 2023 Feb.
Article En | MEDLINE | ID: mdl-36852065

A new ent-abietane diterpenoid, named Euphejolkinolide A (1), was isolated from the whole plant of Euphorbia peplus L. Its structure, including absolute configurations, was determined by spectroscopic analyses and was corroborated by single-crystal X-ray diffraction analysis. This new compound was assessed for its activity to induce lysosome biogenesis through Lyso-Tracker Red staining, in which compound 1 could significantly induce lysosome biogenesis. In addition, quantitative real-time PCR (qRT-PCR) analysis demonstrated a direct correlation between the observed lysosome biogenesis and the transcriptional activation of the lysosomal genes after treatment with the compound 1. Moreover, compound 1 promoted autophagic flux by upregulating LC3-II and downregulating SQSTM1 in both human microglia cells and U251 cells, which is required for cellular homeostasis. Further results suggested 1 induced lysosome biogenesis and autophagy which was mediated by TFEB (transcription factor EB). The structure activity relationships (SAR) analysis suggested that the carbony1 at C-7 in 1 might be a key active group. Overall, the current data suggested that 1 could be a potential compound for lysosome disorder therapy by induction of autophagy.

8.
Bioorg Chem ; 132: 106375, 2023 03.
Article En | MEDLINE | ID: mdl-36682148

Ganoapplins A and B (1 and 2) with a 6/6/6/5/6-fused pentacyclic skeleton containing an aromatic E ring, were obtained from Ganoderma applanatum. Their structures were established through extensive spectroscopic analyses, quantum chemical calculations, including calculated chemical shifts with DP4 + analysis and electronic circular dichroism (ECD). A plausible biosynthetic pathway for 1 and 2 was proposed. Furthermore, their roles in activating autophagy were investigated and the cellular assays showed that 1 and 2 can inhibit tau pathology by inducing autophagy, suggesting their potential against Alzheimer's disease (AD).


Ganoderma , Molecular Structure , Ganoderma/chemistry , Circular Dichroism , Autophagy , Skeleton
9.
Int J Mol Sci ; 24(2)2023 Jan 12.
Article En | MEDLINE | ID: mdl-36675001

Eleven monoterpenoid indole alkaloids, including three new ones, tabercrassines A-C (1-3), were isolated from the seeds of Tabernaemontana crassa. Tabercrassine A (1) is an ibogan-ibogan-type bisindole alkaloid which is formed by the polymerization of two classic ibogan-type monomers through a C3 unit aliphatic chain. Their structures were established by extensive analysis of HRESIMS, NMR, and ECD spectra. Cellular assays showed that alkaloids 1-3 all reduce Aß42 production and inhibit phospho-tau (Thr217), a new biomarker of Alzheimer's disease [AD] associated with BACE1-, NCSTN-, GSK3ß-, and CDK5-mediated pathways, suggesting these alkaloids' potential against AD.


Antineoplastic Agents, Phytogenic , Secologanin Tryptamine Alkaloids , Tabernaemontana , Secologanin Tryptamine Alkaloids/pharmacology , Secologanin Tryptamine Alkaloids/chemistry , Indole Alkaloids/pharmacology , Tabernaemontana/chemistry , Amyloid Precursor Protein Secretases , Antineoplastic Agents, Phytogenic/pharmacology , Drug Screening Assays, Antitumor , Aspartic Acid Endopeptidases , Molecular Structure
10.
Phytochemistry ; 203: 113392, 2022 Nov.
Article En | MEDLINE | ID: mdl-36030903

Three undescribed monoterpenoid indole alkaloid dimers (kopoffines A-C, which are connected via a methylene unit) and with nine known alkaloids were isolated and identified from the fruits of Kopsia arborea Blume. Their structures, including their absolute configurations, were established by HRESIMS, NMR, single-crystal X-ray diffraction, and ECD analyses. Kopoffines A-C showed significant inhibition against cyclin-dependent kinase 5 (IC50: 0.34-2.18 µM). Western blotting analyses showed that kopoffines A-C significantly decreased the protein levels of CDK5 and phospho-CDK5 (Tyr15) (pCDK5) at concentrations of 2.5 and 10 µM. The levels of phospho-Tau (Thr217) (pTau217, a new biomarker of AD), and phospho-Tau (Ser396) (pTau396), which play major roles in the formation of neurofibrillary tangles , were decreased by the kopoffines A-C treatment. Molecular docking studies indicated that kopoffines A-C could form stable interactions with CDK5.


Apocynaceae , Secologanin Tryptamine Alkaloids , Apocynaceae/chemistry , Cyclin-Dependent Kinase 5 , Indole Alkaloids/chemistry , Molecular Docking Simulation , Molecular Structure , Monoterpenes , Phosphorylation , Secologanin Tryptamine Alkaloids/pharmacology
11.
Sci Total Environ ; 829: 154628, 2022 Jul 10.
Article En | MEDLINE | ID: mdl-35304148

Forage crops are widely cultivated as livestock feed to relieve grazing pressure in agro-pastoral regions with arid climates. However, gaseous losses of soil nitrogen (N) following N fertilizer application have been considerable in response to the pursuit of increased crop yield. A two-year experiment was carried out in a typical saline field under a temperate continental arid climate to investigate the effect of N application rate on N2O emissions from barley (Hordeum vulgare L.), corngrass (Zea mays × Zea Mexicana), rye (Secale cereale L.), and sorghum-sudangrass hybrid (Sorghum bicolor × Sorghum sudanense). The dynamics of N2O emissions, hay yield, and crude protein (CP) yield were measured under four N application rates (0, 150, 200, and 250 kg ha-1) in 2016 and 2017. An N2O emission peak was observed for all crop species five days after each N application. Cumulative N2O fluxes in the growing season ranged from 0.66 to 2.40 kg ha-1 and responded exponentially to N application rate. Emission factors of N2O showed a linear increase with N application rate for all crop species, but the linear slopes significantly differed between barley or rye and corngrass and sorghum-sudangrass hybrid. The hay and CP yields of all forage grasses significantly increased with the increase of N application rate from 0 to 200 kg ha-1. Barley and rye with lower hay and CP yields showed higher N2O emission intensities. The increased level of N2O emission intensity was higher from 200 to 250 kg ha-1 than from 150 to 200 kg ha-1. At N application rates of 200 and 250 kg ha-1, CP yield had a significantly negative correlation with cumulative N2O emission and explained 50.5% and 62.9% of the variation, respectively. In conclusion, ~200 kg ha-1 is the optimal N rate for forage crops to minimize N2O emission while maintaining yield in continental arid regions.


Fertilizers , Nitrous Oxide , Agriculture , Crops, Agricultural , Edible Grain/chemistry , Fertilizers/analysis , Nitrogen/analysis , Nitrous Oxide/analysis , Soil
12.
Biochem Biophys Res Commun ; 586: 177-184, 2022 01 01.
Article En | MEDLINE | ID: mdl-34856418

Gastric cancer (GC) is a common tumor with high metastatic rate worldwide. Promoting chemosensitivity is effective for improving therapeutic outcome and survival rate for GC patients. Tripartite motif-containing 21 (TRIM21), a member of TRIM-containing proteins, plays crucial roles in regulating numerous cellular events involved in tumor progression. However, it's regulatory effects on GC growth and drug sensitivity are still unclear. In the present study, we identified that TRIM21 expression was remarkably decreased in human GC tissues compared with the adjacent normal ones, and its down-regulation was closely linked to higher recurrence and lower overall survival rate among GC patients. We then found that apatinib (APA)-reduced GC cell proliferation was significantly abolished by TRIM21 knockdown; however, promoting TRIM21 expression further improved the sensitivity of GC cells to APA treatment, as proved by the remarkably decreased cell viability and colony formation. Furthermore, TRIM21 over-expression dramatically enhanced apoptosis, while its knockdown markedly diminished apoptotic cell death in APA-incubated GC cells. Moreover, stem cell properties of GC cells were also restrained by TRIM21. Our in vivo experiments showed that APA-repressed tumor growth was considerably abolished by TRIM21 knockdown, whereas being further elevated by TRIM21 over-expression. In addition, we showed that TRIM21 markedly decreased enhancer of zeste homolog 1 (EZH1) protein expression levels in GC cells, and importantly, a direct interaction between TRIM21 and EZH1 was verified. Of note, our in vitro studies revealed that EZH1 over-expression remarkably abolished the function of TRIM21 to restrain cell viability and induce apoptosis in APA-incubated GC cells, indicating that EZH1 suppression was necessary for TRIM21 to inhibit GC progression. Together, our findings demonstrated that TRIM21 may be a novel therapeutic target for GC treatment through reducing EZH1 to improve chemosensitivity.


Antineoplastic Agents, Phytogenic/pharmacology , Drug Resistance, Neoplasm/genetics , Polycomb Repressive Complex 2/genetics , Pyridines/pharmacology , Ribonucleoproteins/genetics , Stomach Neoplasms/genetics , Animals , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/genetics , Gene Expression Regulation, Neoplastic , Humans , Male , Mice , Mice, Nude , Polycomb Repressive Complex 2/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Ribonucleoproteins/antagonists & inhibitors , Ribonucleoproteins/metabolism , Signal Transduction , Spheroids, Cellular/drug effects , Spheroids, Cellular/metabolism , Spheroids, Cellular/pathology , Stomach Neoplasms/drug therapy , Stomach Neoplasms/mortality , Stomach Neoplasms/pathology , Survival Analysis , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
13.
Mol Med Rep ; 24(6)2021 Dec.
Article En | MEDLINE | ID: mdl-34676872

The tumor suppressor gene adenomatous polyposis coli (APC) is frequently inactivated or absent in colorectal carcinoma (CRC). Loss­of­function of APC promotes the expression of ß­catenin, which is critical for CRC development. Since ß­catenin acts as an important transcription factor, blockage of ß­catenin may have side effects, including impairment of tissue homeostasis and regeneration, thus limiting the application of ß­catenin inhibitors for the treatment of patients with CRC. Therefore, identifying a novel substrate of APC/ß­catenin may provide essential clues to develop effective drugs. Small interfering RNA technology and lentivirus­mediated overexpression were performed for knockdown and overexpression of pleckstrin 2 (PLEK2) in CRC cells. Cell Counting Kit­8 and colony formation assays, and cell cycle analysis and cell apoptosis detection were used to detect the capacity of cell proliferation, cell cycle distribution and apoptosis. The present study demonstrated that the APC/ß­catenin signaling cascade transcriptionally activated PLEK2 in CRC cells. PLEK2 expression was markedly increased in CRC tissues. There was an inverse correlation between APC and PLEK2 expression in patients with CRC. In vitro, overexpression of PLEK2 increased the proliferation of CRC cells. Opposite results were observed in the cells with knockdown of PLEK2. Furthermore, PLEK2 promoted cell cycle progression and suppressed apoptosis. In summary, upregulation of PLEK2 contributed to CRC proliferation and colony formation activated by the APC/ß­catenin signal pathway. Targeting PLEK2 may be important for the treatment of patients with CRC with activation of the APC/ß­catenin signaling pathway.


Colorectal Neoplasms/drug therapy , Drug Delivery Systems/methods , Membrane Proteins/drug effects , beta Catenin/metabolism , Adenomatous Polyposis Coli/genetics , Adenomatous Polyposis Coli Protein/genetics , Apoptosis , Cell Cycle , Cell Line, Tumor , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic , Genes, APC , HCT116 Cells , Humans , Membrane Proteins/genetics , RNA, Small Interfering , Up-Regulation , Wnt Signaling Pathway , beta Catenin/genetics
14.
Org Biomol Chem ; 19(44): 9637-9640, 2021 11 18.
Article En | MEDLINE | ID: mdl-34714900

Two new quassinoids (1 and 2) were isolated from the twigs of Harrisonia perforata (Blanco) Merr. Perforalactone E (2) possesses an uncommon hexacyclic 1α,12α:5α,13α-dicyclo-9ßH-picrasane skeleton. Its structure was determined based on spectroscopic data and X-ray crystallography. Compounds 1 and 2 could significantly induce lysosomal biogenesis through transcriptional activation of lysosomal genes.


Simaroubaceae
15.
Exp Ther Med ; 22(3): 919, 2021 Sep.
Article En | MEDLINE | ID: mdl-34335880

Vertical bone augmentation is an important challenge in dental implantology. Existing vertical bone augmentation techniques, along with bone grafting materials, have achieved certain clinical progress but continue to have numerous limitations. In order to evaluate the possibility of using biomaterials to develop bone substitutes, medical devices and/or new bone grafting techniques for vertical bone augmentation, it is essential to establish clinically relevant animal models to investigate their biocompatibility, mechanical properties, applicability and safety. The present review discusses recent animal experiments related to vertical bone augmentation. In addition, surgical protocols for establishing relevant preclinical models with various animal species were reviewed. The present study aims to provide guidance for selecting experimental animal models of vertical bone augmentation.

16.
Cell Biosci ; 11(1): 73, 2021 Apr 15.
Article En | MEDLINE | ID: mdl-33858520

OBJECTIVE: Dysregulation of KLF7 participates in the development of various cancers, but it is unclear whether there is a link between HCC and aberrant expression of KLF7. The aim of this study was to investigate the role of KLF7 in proliferation and migration of hepatocellular carcinoma (HCC) cells. METHODS: CCK8, colony growth, transwell, cell cycle analysis and apoptosis detection were performed to explore the effect of KLF7, VPS35 and Ccdc85c on cell function in vitro. Xenografted tumor growth was used to assess in vivo role of KLF7. Chip-qPCR and luciferase reporter assays were applied to check whether KLF7 regulated VPS35 at transcriptional manner. Co-IP assay was performed to detect the interaction between VPS35 and Ccdc85c. Immunohistochemical staining and qRT-PCR analysis were performed in human HCC sampels to study the clinical significance of KLF7, VPS35 and ß-catenin. RESULTS: Firstly, KLF7 was highly expressed in human HCC samples and correlated with patients' differentiation and metastasis status. KLF7 overexpression contributed to cell proliferation and invasion of HCC cells in vitro and in vivo. KLF7 transcriptional activation of VPS35 was necessary for HCC tumor growth and metastasis. Further, co-IP studies revealed that VPS35 could interact with Ccdc85c in HCC cells. Rescue assay confirmed that overexpression of VPS35 and knockdown of Ccdc85c abolished the VPS35-medicated promotion effect on cell proliferation and invasion. Finally, KLF7/VPS35 axis regulated Ccdc85c, which involved in activation of ß-catenin signaling pathway, confirmed using ß-catenin inhibitor, GK974. Functional studies suggested that downregulation of Ccdc85c partly reversed the capacity of cell proliferation and invasion in HCC cells, which was regulated by VPS35 upregulation. Lastly, there was a positive correlation among KLF7, VPS35 and active-ß-catenin in human HCC patients. CONCLUSION: We demonstrated that KLF7/VPS35 axis promoted HCC cell progression by activating Ccdc85c-medicated ß-catenin pathway. Targeting this signal axis might be a potential treatment strategy for HCC.

17.
Br J Cancer ; 125(1): 101-111, 2021 07.
Article En | MEDLINE | ID: mdl-33772151

BACKGROUND: CHK1 is considered an oncogene with overexpression in numerous cancers. However, CHK1 signalling regulation in hepatocellular carcinoma (HCC) remains unclear. METHODS: CHEK1 mRNA, protein, pri-miR-195 and miR-195 expression in HCC tissue was determined by qPCR, WB and IF staining assay. Survival analyses in HCC with high- and low-CHEK1 mRNA expression was performed using TCGA database. Relative luciferase activity was investigated in HCC cells transfected with p-CHEK1 3'UTR. Apoptosis was detected by TUNEL assay. NK and CD8+ T cells were analysed by flow cytometry. RESULTS: CHK1 is increased in human HCC tumours compared with non-cancerous liver. High CHK1 predicts worse prognosis. IFN-γ suppresses CHK1 via IRF-1 in HCC cells. The molecular mechanism of IRF-1 suppressing CHK1 is post-transcriptional by promoting miR-195 binding to CHEK1 mRNA 3'UTR, which exerts a translational blockade. Upregulated IRF-1 inhibits CHK1, which induces apoptosis of HCC cells. Likewise, CHK1 inhibition augments cellular apoptosis in HCC tumours. This effect may be a result of increased tumour NK cell infiltration. However, IRF-1 expression or CHK1 inhibition also upregulates PD-L1 expression via increased STAT3 phosphorylation. CONCLUSIONS: IRF-1 induces miR-195 to suppress CHK1 protein expression. Both increased IRF-1 and decreased CHK1 upregulate cellular apoptosis and PD-L1 expression in HCC.


B7-H1 Antigen/metabolism , Carcinoma, Hepatocellular/metabolism , Checkpoint Kinase 1/genetics , Interferon Regulatory Factor-1/metabolism , Liver Neoplasms/metabolism , MicroRNAs/genetics , Animals , Apoptosis , Carcinoma, Hepatocellular/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Checkpoint Kinase 1/metabolism , Female , Hep G2 Cells , Humans , Liver Neoplasms/genetics , Mice , Neoplasm Transplantation , Phosphorylation , Prognosis , Pyrazines/pharmacology , Pyrazoles/pharmacology , STAT3 Transcription Factor/metabolism , Up-Regulation/drug effects
18.
Front Mol Biosci ; 8: 635470, 2021.
Article En | MEDLINE | ID: mdl-33778005

Hydrogen sulfide (H2S) is a gasotransmitter along with nitric oxide and carbon oxide, which is involved in plant growth and development as well as biotic and abiotic stress resistance. In a previous study, we reported that mitogen-activated protein kinases, especially MPK4, are important downstream components of H2S involved in alleviating cold stress; however the underlying mechanism is unclear. In this study, we determined that the ability of H2S to alleviate cold stress is impaired in mpk4 mutants, but not in the upstream mek2 and crlk1 mutants. MPK4 was basically persulfidated, and NaHS (H2S donor) further increased the persulfidation level of MPK4. MEK2 was not persulfidated by H2S. NaHS treatments increased the MPK4 activity level nearly tenfold. The persulfidation signal of MPK4 did not disappear after eight cystein residues in MPK4 were site-mutated, respectively. Above all, our results suggested that H2S alleviates cold stress directly by persulfidating MPK4 and increasing the MPK4 kinase activity.

19.
Transl Oncol ; 14(6): 101059, 2021 Jun.
Article En | MEDLINE | ID: mdl-33735820

Hepatocellular carcinoma (HCC) is the third leading cause of cancer-related death worldwide. Lenvatinib oral chemotherapy is approved as a first-line treatment of patients with unresectable HCC. The efficacy and therapeutic duration of lenvatinib are limited by drug resistance, and the mechanism is unclear. IRF2 is a constitutive transcription factor associated with the development of various cancers by regulating cancer cell growth, apoptosis, and drug resistance. However, the potential role of IRF2 in lenvatinib resistance in HCC has not been explored. In this study, we found that IRF2 promoted proliferation, inhibited apoptosis, and increased lenvatinib resistance of HCC cells by regulating ß-catenin expression. Silencing IRF2 downregulated the expression of ß-catenin, while overexpressing IRF2 upregulated ß-catenin. Moreover, the expression of ß-catenin and IRF2 was positively correlated in HCC tissues. Inhibiting ß-catenin with XAV-939 effectively abrogated ß-catenin expression caused by lenvatinib treatment. These findings identify an important function of IRF2 in HCC and demonstrate a mechanism of lenvatinib resistance of HCC cells. Targeting IRF2 may be a potential strategy to improve the therapeutic effect of lenvatinib on HCC.

20.
Cancer Lett ; 506: 95-106, 2021 05 28.
Article En | MEDLINE | ID: mdl-33689775

Interferon regulatory factor 1 (IRF-1) is a tumor suppressor gene in cancer biology with anti-proliferative and pro-apoptotic effect on cancer cells, however mechanisms of IRF-1 regulating tumor microenvironment (TME) in hepatocellular carcinoma (HCC) remain only partially characterized. Here, we investigated that IRF-1 regulates C-X-C motif chemokine 10 (CXCL10) and chemokine receptor 3 (CXCR3) to activate anti-tumor immunity in HCC. We found that IRF-1 mRNA expression was positively correlated with CXCL10 and CXCR3 through qRT-PCR assay in HCC tumors and in analysis of the TCGA database. IRF-1 response elements were identified in the CXCL10 promoter region, and ChIP-qPCR confirmed IRF-1 binding to promote CXCL10 transcription. IRF-2 is a competitive antagonist for IRF-1 mediated transcriptional effects, and overexpression of IRF-2 decreased basal and IFN-γ induced CXCL10 expression. Although IRF-1 upregulated CXCR3 expression in HCC cells, it inhibited proliferation and exerted pro-apoptotic effects, which overcome proliferation partly mediated by activating the CXCL10/CXCR3 autocrine axis. In vitro and in vivo studies showed that IRF-1 increased CD8+ T cells, NK and NKT cells migration, and activated IFN-γ secretion in NK and NKT cells to induce tumor apoptosis through the CXCL10/CXCR3 paracrine axis. Conversely, this effect was markedly abrogated in HCC tumor bearing mice deficient in CXCR3. Therefore, the IRF-1/CXCL10/CXCR3 axis contributes to the anti-tumor microenvironment in HCC.


Carcinoma, Hepatocellular/immunology , Chemokine CXCL10/physiology , Interferon Regulatory Factor-1/physiology , Liver Neoplasms/immunology , Receptors, CXCR3/physiology , Animals , Apoptosis , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Chemokine CXCL10/genetics , Female , Humans , Liver Neoplasms/pathology , Mice , Mice, Inbred C57BL , Promoter Regions, Genetic , T-Lymphocytes/immunology
...