Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 107
Filter
1.
Curr Med Sci ; 43(5): 1023-1032, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37615927

ABSTRACT

OBJECTIVE: Cisplatin is the first-line treatment for breast cancer, but it faces challenges of drug resistance. This study investigated new molecular mechanisms underlying cisplatin resistance in breast cancer. METHODS: We analyzed sequencing data from the TCGA database to identify potential associations between transmembrane emp24 protein transport domain containing 2 (TMED2) and breast cancer. Western blotting, real-time PCR, CCK-8, and TUNEL assays were used to measure the effects and molecular mechanism of TMED2 on cisplatin resistance in MCF-7 and MDA-MB-231 cell lines. RESULTS: TMED2 was overexpressed in breast cancer and associated with poor prognosis. TMED2 increased cisplatin resistance in breast cancer cells in vitro via promoting ubiquitination of Kelch-like ECH-associated protein 1 (KEAP1), relieving inhibition of KEAP1 on nuclear factor erythroid 2-related factor 2 (Nrf2), and increasing expression of downstream drug resistance related genes, such as heme oxygenase 1 (HO-1) and NAD (P) H quinone oxidoreductase 1 (NQO1). CONCLUSION: We identified a new molecular mechanism by which TMED2 affects cisplatin resistance in breast cancer. Our results provide theoretical guidance for future clinical applications.


Subject(s)
Breast Neoplasms , Cisplatin , Drug Resistance, Neoplasm , Female , Humans , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Cisplatin/pharmacology , Cisplatin/therapeutic use , Kelch-Like ECH-Associated Protein 1/genetics , Membrane Proteins/metabolism , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Vesicular Transport Proteins/metabolism , Drug Resistance, Neoplasm/genetics
2.
Acta Pharmacol Sin ; 44(11): 2282-2295, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37280363

ABSTRACT

Abnormalities of FGFR1 have been reported in multiple malignancies, suggesting FGFR1 as a potential target for precision treatment, but drug resistance remains a formidable obstacle. In this study, we explored whether FGFR1 acted a therapeutic target in human T-cell acute lymphoblastic leukemia (T-ALL) and the molecular mechanisms underlying T-ALL cell resistance to FGFR1 inhibitors. We showed that FGFR1 was significantly upregulated in human T-ALL and inversely correlated with the prognosis of patients. Knockdown of FGFR1 suppressed T-ALL growth and progression both in vitro and in vivo. However, the T-ALL cells were resistant to FGFR1 inhibitors AZD4547 and PD-166866 even though FGFR1 signaling was specifically inhibited in the early stage. Mechanistically, we found that FGFR1 inhibitors markedly increased the expression of ATF4, which was a major initiator for T-ALL resistance to FGFR1 inhibitors. We further revealed that FGFR1 inhibitors induced expression of ATF4 through enhancing chromatin accessibility combined with translational activation via the GCN2-eIF2α pathway. Subsequently, ATF4 remodeled the amino acid metabolism by stimulating the expression of multiple metabolic genes ASNS, ASS1, PHGDH and SLC1A5, maintaining the activation of mTORC1, which contributed to the drug resistance in T-ALL cells. Targeting FGFR1 and mTOR exhibited synergistically anti-leukemic efficacy. These results reveal that FGFR1 is a potential therapeutic target in human T-ALL, and ATF4-mediated amino acid metabolic reprogramming contributes to the FGFR1 inhibitor resistance. Synergistically inhibiting FGFR1 and mTOR can overcome this obstacle in T-ALL therapy.


Subject(s)
Amino Acids , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma , Humans , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , TOR Serine-Threonine Kinases/metabolism , Signal Transduction , T-Lymphocytes/metabolism , Cell Line, Tumor , Minor Histocompatibility Antigens , Amino Acid Transport System ASC/metabolism , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Activating Transcription Factor 4/metabolism
3.
Oncol Rep ; 50(2)2023 Aug.
Article in English | MEDLINE | ID: mdl-37387422

ABSTRACT

Glucosamine­phosphate N­acetyltransferase 1 (GNPNAT1) is a member of the acetyltransferase superfamily, related to general control non­depressible 5 (GCN5). It has been documented that GNPNAT1 expression is increased in lung cancer, whereas its involvement in breast cancer (BC) remains to be further investigated. The present study aimed to evaluate the expression levels of GNPNAT1 in BC and its effect on BC stem cells (BCSCs). The Cancer Genome Atlas (TCGA) database was used for the analysis of the expression of GNPNAT1 and its clinical significance. Cox regression and logistic regression analyses were used to evaluate prognosis­related factors. The GNPNAT1­binding protein network was constructed using the Search Tool for the Retrieval of Interacting Genes/Proteins (STRING) application. The biological signaling pathways implicated in GNPNAT1 were investigated through function enrichment analysis including Gene Ontology, Kyoto Encyclopedia of Genes and Genomes, and gene set enrichment analysis. The single­sample GSEA method was used to investigate the connection between the level of immune infiltration and GNPNAT1 expression in BC. GNPNAT1 expression was upregulated in patients with BC and was significantly associated with a poor prognosis. GNPNAT1 and its co­expressed genes were mostly enriched in nuclear transport, Golgi vesicle transport, ubiquitin­like protein transferase activity and ribonucleoprotein complex binding, as determined using functional enrichment analysis. GNPNAT1 expression was positively associated with Th2 cells and T­helper cells, and negatively associated with plasmacytoid dendritic cells, CD8+ T­cells and cytotoxic cells. Additionally, the GNPNAT1 expression levels were considerably increased in BCSCs. GNPNAT1 knockdown markedly decreased the stemness ability of SKBR3 and Hs578T cells, including the production of CSC markers and mammosphere or clone formation, while GNPNAT1 overexpression increased the stemness level. Hence, the findings of the present study demonstrate that GNPNAT1 may be exploited as a novel prognostic biomarker and therapeutic target for BC.


Subject(s)
Breast Neoplasms , Humans , Female , Breast Neoplasms/genetics , CD8-Positive T-Lymphocytes , Prognosis , Acetyltransferases , Biomarkers , Glucosamine 6-Phosphate N-Acetyltransferase
4.
Probiotics Antimicrob Proteins ; 15(3): 443-450, 2023 06.
Article in English | MEDLINE | ID: mdl-34599740

ABSTRACT

Microbiota of lower female reproductive tract is special in its microorganism composition with Lactobacillus as the predominant bacteria. A few of Lactobacillus species have been identified to benefit the inhibition of inflammatory and malignant diseases. Lacticaseibacillus casei LH23 is a strain isolated from traditional fermented food and had been demonstrate to ameliorate DSS-induced colitis in mice. In the present study, effects of Lacticaseibacillus casei LH23 on cervical cancer cells were investigated. Supernatants of lysates and heat-inactivated Lacticaseibacillus casei LH23 were found to inhibit the expression of human papillomavirus genes E6/E7 which is the main causative factor of cervical cancer. With MTT, EdU staining, and TUNEL staining assays, Lacticaseibacillus casei LH23 was shown to suppress the proliferation and induced the apoptosis of cervical cancer cells. Additionally, with wound-healing and Western-blot assays, Lacticaseibacillus casei LH23 was shown to slowdown the migration of cervical cancer cells and altered the expression of metastasis-related genes. These results demonstrated the anti-cervical cancer potential of Lacticaseibacillus casei LH23.


Subject(s)
Lacticaseibacillus casei , Papillomavirus Infections , Uterine Cervical Neoplasms , Female , Humans , Animals , Mice , Lacticaseibacillus , Papillomavirus E7 Proteins/genetics , Uterine Cervical Neoplasms/drug therapy , Gene Expression
5.
Macromol Biosci ; 23(3): e2200173, 2023 03.
Article in English | MEDLINE | ID: mdl-36448643

ABSTRACT

It is always big challenges for hyaluronic acid (HA) in transmembrane absorbing and efficient delivering to the skin. Pep-1, as one of the cell-penetrating peptides, has been documented to permeate various substances across cellular membranes without covalent binding. Here, a novel hyaluronic acid binding peptide (named HaBP) is designed, and then combined with Pep-1 to enhance the cell-penetrating efficiency of HA. The results of ELISA and immunofluorescence assay show that HaBP could bind with HA very well, and a combination of Pep-1 and HaBP could efficiently improve the transmembrane ability of HA. Furthermore, HA gradually enters the dermis from the surface of the skin in mice when it is administrated with both HaBP and Pep-1, while there are no obvious allergies or other adverse reactions during this process. This study finds a new method to promote the efficient transmembrane and transdermal absorption of HA, and throws some light on further research on the development of hyaluronic acid and its related cosmetics or drugs.


Subject(s)
Hyaluronic Acid , Skin Absorption , Mice , Animals , Hyaluronic Acid/chemistry , Peptides/chemistry , Skin/metabolism , Cell Membrane/metabolism
6.
Int J Mol Sci ; 23(19)2022 Oct 09.
Article in English | MEDLINE | ID: mdl-36233293

ABSTRACT

Thyroid cancer remains the most common endocrine malignancy worldwide, and its incidence has steadily increased over the past four years. Papillary Thyroid Cancer (PTC) is the most common differentiated thyroid cancer, accounting for 80-85% of all thyroid cancers. Mitochondrial proteins (MRPs) are an important part of the structural and functional integrity of the mitochondrial ribosomal complex. It has been reported that MRPL9 is highly expressed in liver cancer and promotes cell proliferation and migration, but it has not been reported in PTC. In the present study we found that MRPL9 was highly expressed in PTC tissues and cell lines, and lentivirus-mediated overexpression of MRPL9 promoted the proliferation and migration ability of PTC cells, whereas knockdown of MRPL9 had the opposite effect. The interaction between MRPL9 and GGCT (γ-glutamylcyclotransferase) was found by immunofluorescence and co-immunoprecipitation experiments (Co-IP). In addition, GGCT is highly expressed in PTC tissues and cell lines, and knockdown of GGCT/MRPL9 in vivo inhibited the growth of subcutaneous xenografts in nude mice and inhibited the formation of lung metastases. Mechanistically, we found that knockdown of GGCT/MRPL9 inhibited the MAPK/ERK signaling pathway. In conclusion, our study found that the interaction of GGCT and MRPL9 modulates the MAPK/ERK pathway, affecting the proliferation and migration of PTC cells. Therefore, GGCT/MRPL9 may serve as a potential biomarker for PTC monitoring and PTC treatment.


Subject(s)
MAP Kinase Signaling System , Thyroid Neoplasms , gamma-Glutamylcyclotransferase , Animals , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic , Humans , Mice , Mice, Nude , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Thyroid Cancer, Papillary/pathology , Thyroid Neoplasms/pathology , gamma-Glutamylcyclotransferase/genetics
7.
J Transl Med ; 20(1): 426, 2022 09 23.
Article in English | MEDLINE | ID: mdl-36138468

ABSTRACT

BACKGROUND: Long non-coding RNAs (lncRNAs) are closely related to the occurrence and development of cancer. Abnormally expressed lncRNA can be used as a diagnostic marker for cancer. In this study, we aim to investigate the clinical significance of MIR99AHG expression in lung adenocarcinoma (LUAD), and its biological roles in LUAD progression. METHODS: The relative expression of MIR99AHG in LUAD tissues and cell lines was analyzed using public databases and RT-qPCR. The biological functions of MIR99AHG were investigated using a loss-of-function approach. The effect of MIR99AHG on lung fibrosis was assessed by scratch assay, invasion assay and lung fibrosis rat model. FISH, luciferase reporter assay and immunofluorescence were performed to elucidate the underlying molecular mechanisms. RESULTS: LncRNA MIR99AHG expression level was downregulated in LUAD tissues and cell lines. Low MIR99AHG levels were associated with poorer patient overall survival. Functional analysis showed that MIR99AHG is associated with the LUAD malignant phenotype in vitro and in vivo. Further mechanistic studies showed that, MIR99AHG functions as a competitive endogenous RNA (ceRNA) to antagonize miR-136-5p-mediated ubiquitin specific protease 4 (USP4) degradation, thereby unregulated the expression of angiotensin-converting enzyme 2 (ACE2), a downstream target gene of USP4, which in turn affected alveolar type II epithelial cell fibrosis and epithelial-mesenchymal transition (EMT). In summary, the MIR99AHG/miR-136-5p/USP4/ACE2 signalling axis regulates lung fibrosis and EMT, thus inhibiting LUAD progression. CONCLUSION: This study showed that downregulated MIR99AHG leads to the development of pulmonary fibrosis. Therefore, overexpression of MIR99AHG may provide a new approach to preventing LUAD progression.


Subject(s)
Adenocarcinoma , Lung Neoplasms , MicroRNAs , Pulmonary Fibrosis , RNA, Long Noncoding , Adenocarcinoma/genetics , Angiotensin-Converting Enzyme 2 , Animals , Apoptosis/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation, Neoplastic , Lung Neoplasms/pathology , MicroRNAs/genetics , MicroRNAs/metabolism , Pulmonary Fibrosis/genetics , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Rats , Ubiquitin-Specific Proteases/genetics , Ubiquitin-Specific Proteases/metabolism
8.
Oncol Rep ; 48(4)2022 10.
Article in English | MEDLINE | ID: mdl-36004470

ABSTRACT

Cell division cycle­associated 5 (CDCA5) plays a critical role in the progression of various human cancers by regulating cell cycle­related proteins; however, the function of CDCA5 in breast cancer (BC) is poorly understood. The aim of the present study was to investigate the expression level of CDCA5 in BC and its effect on BC progression. CDCA5 was found to be highly expressed in patients with BC, as well as in BC cell lines. It was also found that a high CDCA5 expression in BC was significantly associated with a shorter survival rate. In addition, the expression level of CDCA5 was significantly increased in stem cells derived from suspension­cultured BC cells, as compared to adherent­cultured cells. CDCA5 knockdown in MCF7 and SKBR3 cells significantly reduced cell proliferation, migration and clone formation. At the same time, the stemness capacity of BC cells, determined by analyzing cancer stem cell marker expression and mammosphere formation, was also markedly diminished following the knockdown of CDCA5. In addition, in vivo experiments demonstrated that CDCA5 knockdown in MCF7 cells markedly reduced tumor growth. On the whole, the present study demonstrates that CDCA5 may be used as a prognostic biomarker and therapeutic target for BC.


Subject(s)
Breast Neoplasms , Adaptor Proteins, Signal Transducing/genetics , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Proliferation/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Prognosis
9.
Int J Mol Sci ; 23(12)2022 Jun 09.
Article in English | MEDLINE | ID: mdl-35742899

ABSTRACT

Gastric cancer (GC) is the fifth most common cancer and the third deadliest cancer in the world, and the occurrence and development of GC are influenced by epigenetics. Methyltransferase-like 3 (METTL3) is a prominent RNA n6-adenosine methyltransferase (m6A) that plays an important role in tumor growth by controlling the work of RNA. This study aimed to reveal the biological function and molecular mechanism of METTL3 in GC. The expression level of METTL3 in GC tissues and cells was detected by qPCR, Western blot and immunohistochemistry, and the expression level and prognosis of METTL3 were predicted in public databases. CCK-8, colony formation, transwell and wound healing assays were used to study the effect of METTL3 on GC cell proliferation and migration. In addition, the enrichment effect of METTL3 on DEK mRNA was detected by the RIP experiment, the m6A modification effect of METTL3 on DEK was verified by the MeRIP experiment and the mRNA half-life of DEK when METTL3 was overexpressed was detected. The dot blot assay detects m6A modification at the mRNA level. The effect of METTL3 on cell migration ability in vivo was examined by tail vein injection of luciferase-labeled cells. The experimental results showed that METTL3 was highly expressed in GC tissues and cells, and the high expression of METTL3 was associated with a poor prognosis. In addition, the m6A modification level of mRNA was higher in GC tissues and GC cell lines. Overexpression of METTL3 in MGC80-3 cells and AGS promoted cell proliferation and migration, while the knockdown of METTL3 inhibited cell proliferation and migration. The results of in vitro rescue experiments showed that the knockdown of DEK reversed the promoting effects of METTL3 on cell proliferation and migration. In vivo experiments showed that the knockdown of DEK reversed the increase in lung metastases caused by the overexpression of METTL3 in mice. Mechanistically, the results of the RIP experiment showed that METTL3 could enrich DEK mRNA, and the results of the MePIP and RNA half-life experiments indicated that METTL3 binds to the 3'UTR of DEK, participates in the m6A modification of DEK and promotes the stability of DEK mRNA. Ultimately, we concluded that METTL3 promotes GC cell proliferation and migration by stabilizing DEK mRNA expression. Therefore, METTL3 is a potential biomarker for GC prognosis and a therapeutic target.


Subject(s)
Stomach Neoplasms , Adenosine/analogs & derivatives , Adenosine/metabolism , Animals , Cell Transformation, Neoplastic , Methyltransferases/genetics , Methyltransferases/metabolism , Mice , RNA, Messenger/genetics , RNA, Messenger/metabolism , Stomach Neoplasms/pathology
10.
Exp Ther Med ; 24(1): 487, 2022 Jul.
Article in English | MEDLINE | ID: mdl-35761803

ABSTRACT

Peripheral blood monocytes acquire the phenotype of myeloid-derived suppressor cells (MDSCs) by induction of cytokine or co-culture with cancer cells and are widely used to model MDSCs for in vitro studies. However, the simplest method of plastic adhesive sorting is poorly described as the purity of monocyte resulting from this method is the lowest compared with flow cytometry cell-sorting and magnetic beads sorting. Therefore, the present study aimed at investigating the effect of the plastic adhesive monocyte isolation techniques on the resulting MDSCs phenotype. Monocytes were allowed to adhere for 1 h and cultured with IL6 and granulocyte-macrophage colony-stimulating factors (GM-CSF) for 7 days. Plastic adhesion sorting resulted in early low monocyte yield and purity, but high purity of MDSCs was obtained by refreshing the induction medium. The resulting MDSCs were the major subpopulation of CD33+CD11b+CD14+CD15-human leukocyte antigen (HLA)-/low cells and provided the potent capacity to suppress T cell proliferation and cytokine IFN-γ production. Moreover, the induced MDSCs were inhibited by STAT3 inhibitor WP1066, resulting in downregulation of phosphorylated-STAT3 and PD-L1 expression and upregulation of apoptosis respectively. In conclusion, the present study described the generation of monocytic MDSCs from adherence monocytes and the inhibition of STAT3 inhibitor WP1066 on the induced MDSCs. The present study contributed to the development of a new clinical drug, WP1066 targeting MDSC.

12.
Int J Oncol ; 60(6)2022 06.
Article in English | MEDLINE | ID: mdl-35475491

ABSTRACT

Liver cancer is a malignant cancer phenotype for which there currently remains a lack of reliable biomarkers and therapeutic targets for disease management. Tryptophan 2,3­dioxygenase (TDO2), a heme­containing polyoxygenase enzyme, is primarily expressed in cells of the liver and nervous systems. In the present study, through the combination of cancer bioinformatics and analysis of clinical patient samples, it was shown that TDO2 expression in liver cancer tissue samples was significantly higher than that in normal tissues, and liver cancer patients with high TDO2 expression had a poor prognosis. Mechanistic studies on liver cancer cells showed that TDO2 promoted cancer cell migration and invasion via signal transduction through the Wnt5a pathway. Such regulation impacted the expression of cancer­associated biomarkers, such as matrix metalloprotease 7 (MMP7) and the cell adhesion receptor CD44. Treatment with a calcium channel blocker (azelnidipine) reduced TDO2 levels and inhibited liver cancer cell migration and invasion. A mouse xenograft cancer model showed that TDO2 promoted tumorigenesis. Furthermore, azelnidipine treatment to downregulate TDO2 also decreased liver cancer development in this mouse cancer model. TDO2 is thus not only a useful liver cancer biomarker but a potential drug target for management of liver cancer.


Subject(s)
Liver Neoplasms , Tryptophan Oxygenase , Animals , Biomarkers, Tumor , Cell Line , Cell Movement , Humans , Liver Neoplasms/drug therapy , Liver Neoplasms/genetics , Mice , Tryptophan/metabolism , Tryptophan Oxygenase/genetics , Tryptophan Oxygenase/metabolism , Wnt-5a Protein/genetics
13.
Oncol Res ; 28(9): 885-897, 2022 Jan 31.
Article in English | MEDLINE | ID: mdl-34321149

ABSTRACT

Abnormal cell proliferation caused by abnormal transcription regulation mechanism seems to be one of the reasons for the progression of breast cancer and also the pathological basis. MicroRNA-142-5p (miR-142-5p) is a low-expressed miRNA in breast cancer. The role of MKL-1s regulation of DNMT1 in breast cancer cell proliferation and migration is still unclear. MKL-1 (myocardin related transcription factor A) can bind to the conserved cis-regulatory element CC (A/T) 6GG (called CarG box) in the promoter to regulate the transcription of miR-142-5p. The expressions of miR-142-5p and MKL-1 are positively correlated. In addition, it has been proved that DNMT1 is the target of miR-142-5p, which inhibits the expression of DNMT1 by targeting the 3-UTR of DNMT1, thereby forming a feedback loop and inhibiting the migration and proliferation of breast cancer. Our data provide important and novel insights into the MKL-1/miR-142-5p/DNMT1/maspin signaling pathway and may become a new idea for breast cancer diagnosis, treatment, and prognosis.


Subject(s)
Breast Neoplasms , MicroRNAs , 3' Untranslated Regions , Breast Neoplasms/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation , DNA (Cytosine-5-)-Methyltransferase 1 , Female , Gene Expression Regulation, Neoplastic , Humans , MicroRNAs/genetics
14.
Life Sci ; 289: 120242, 2022 Jan 15.
Article in English | MEDLINE | ID: mdl-34922939

ABSTRACT

Bulky DNA damage inducing chemotherapeutic cancer drugs such as cisplatin (CIS) and doxorubicin (DOX) are commonly used in the treatment of a variety of cancers. However, they often cause multi-organ toxicity, and the mechanisms underlying are not clear. Using cellular model, the present study showed that persistent endogenous reactive oxygen species (ROS) were stimulated after a single dose short treatment with CIS and DOX. ROS level correlated with the formation of DNA double-strand breaks (DSBs). Knockdown BRCA1, a key player involved in homologous recombination (HR), enhanced ROS accumulation. Whereas knockdown DNA-PKcs and overexpress BRCA1 to inhibit nonhomologous end-joining (NHEJ) repair pathway and restore HR can partially suppress ROS levels. These data indicated that ROS production is associated with DSB formation and repair which is likely a downstream event of DNA repair. Further studies showed that knockdown DNA repair regulators PP2A but not ATM, could partially reduce ROS too. The induction of ROS affected the level of proinflammatory cytokines interleukin-1ß (IL-1ß), interleukin-6 (IL-6) and tumor necrosis factor-alpha (TNF-α). Collectively, the present study reveals that DNA repair associated metabolism change and oxidative stress may be a direct cause of the severe side effects associated with genotoxic chemotherapy cancer drugs.


Subject(s)
Antineoplastic Agents , DNA Breaks, Double-Stranded , DNA End-Joining Repair/drug effects , DNA, Neoplasm , Neoplasm Proteins , Neoplasms , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacology , Cell Line, Tumor , DNA, Neoplasm/genetics , DNA, Neoplasm/metabolism , Humans , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology
15.
Aging (Albany NY) ; 13(21): 24402-24416, 2021 11 14.
Article in English | MEDLINE | ID: mdl-34775375

ABSTRACT

Tissue inhibitor matrix metalloproteinase 1 (TIMP1) has been reported to act as a tumor oncogene in colon cancer. However, little is known about the biological role of TIMP1 in gastric cancer. In this study, we found that the expression of TIMP1 in GC tissues was upregulated compared with the normal gastric tissues. TIMP1 was confirmed as a direct target of miR-6745 and silencing TIMP1 mimicked the effects of miR-6745 in GC cells. Further mechanism studies have shown that miR-6745 inhibits the Wnt/ß-catenin pathway by targeting TIMP1, thereby inhibiting cell proliferation, migration and invasion. In addition, through the analysis of GC tissues, a negative correlation between miR-6745 and TIMP1 was found in 42 GC tissues. Our findings indicate that the miR-6745-TIMP1 axis regulates Wnt/ßcatenin signaling and participates in GC tumorigenesis and provide a potential therapeutic target for preventing GC progression.


Subject(s)
MicroRNAs/genetics , Stomach Neoplasms , Tissue Inhibitor of Metalloproteinase-1/genetics , Animals , Cell Line, Tumor , Cell Proliferation/genetics , Humans , Male , Mice , Mice, Nude , Neoplasm Metastasis/genetics , Stomach/metabolism , Stomach/pathology , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , Stomach Neoplasms/mortality , Stomach Neoplasms/pathology , Wnt Signaling Pathway
16.
Virus Res ; 306: 198602, 2021 12.
Article in English | MEDLINE | ID: mdl-34662680

ABSTRACT

High risk human papillomavirus (HPV) is the main causative factor of cervical cancer. In addition, estrogen and its receptors are also involved in the development of carcinogenesis. The canonical estrogen receptor ERα is frequently deficient while its variant ERα-36 is highly expressed in cervical cancer cells. The biological significance for this receptor transition from ERα to ERα-36 remains unclear. In the present study, the results of RT-PCR and Western blot demonstrated that ERα and ERα-36 function antagonistically on the expression of the viral oncogenes HPV E6 and E7. At mRNA and protein levels, ERα inhibited HPV E6/E7 expression whereas ERα-36 stimulated HPV E6/E7 expression. Overexpression of ERα-36 promoted cell proliferation while reintroduction of ERα into cervical cancer cells did not significantly affect cell proliferation which is in line with the different effects of . ERα-36 and ERα on the expression of cell cycle regulator, namely p53, p21 and cyclin D1. Furthermore, ERα suppressed whereas ERα-36 promoted the migration and invasion of cervical cancer cells, which should be related to the oppositive roles of ERα and ERα-36 in the Wnt/ß-catenin/MRTF-A signaling pathway which is activated by HPV E7. Results of this study suggest that ERα functions as a tumor suppressor whereas ERα-36 is an oncoprotein in cervical cancer cells. ERα deficiency together with ERα-36 overexpression might enhance the expression of HPV E6/E7 genes and facilitate the development of cervical cancer. Targeting ERα-36 with selective antagonists should be a promising strategy for cervical cancer therapy.


Subject(s)
Oncogene Proteins, Viral , Papillomavirus Infections , Uterine Cervical Neoplasms , Estrogen Receptor alpha/genetics , Estrogens/pharmacology , Female , Humans , Oncogene Proteins, Viral/genetics , Oncogene Proteins, Viral/metabolism , Oncogenes , Papillomavirus E7 Proteins/genetics , Papillomavirus Infections/complications , Papillomavirus Infections/genetics , Phenotype , Repressor Proteins/genetics , Repressor Proteins/metabolism
17.
Int J Biol Sci ; 17(9): 2278-2293, 2021.
Article in English | MEDLINE | ID: mdl-34239355

ABSTRACT

Effectively targeting cancer stem cells to treat cancer has great therapeutic prospects. However, the effect of microRNA miR-17/MKL-1 on gastric cancer stem cells has not been studied yet. This study preliminarily explored the mechanism of miR-17/MKL-1 in gastric cancer stem cells. Many previous reports have indicated that microRNA and EMT regulated cancer stem cell characteristics, and miR-17 and MKL-1 were involved as a critical gene in migration and invasion in the EMT pathway. Through RT-PCR, Western Blot, flow cytometry, immunofluorescence, sphere formation xenograft tumor assays and drug resistance, the role of miR-17-5p and MKL-1 on promoting stem cell-like properties of gastric cancer were verified in vivo and vitro. Next, MKL-1 targets CD44, EpCAM, and miR -17-5p promoter verified by luciferase assay and ChIP. Besides, the TCGA database analysis found that both miR-17-5p and MKL-1 increased in gastric cancer, and the prognostic survival of the MKL-1 high expression group was reduced. It is found that MKL-1 promotes expression by targeting miR-17, CD44 and EpCAM promoters. Besides, the TCGA database analysis found that both miR-17-5p and MKL-1 increased in gastric cancer, and the prognostic survival of the MKL-1 high expression group was reduced. These findings reveal new regulatory signaling pathways for gastric cancer stem cells, thus it give new insights on potential early diagnosis and/or molecular therapy for gastric cancer.


Subject(s)
MicroRNAs/metabolism , Neoplastic Stem Cells/pathology , Stomach Neoplasms/pathology , Trans-Activators/metabolism , Animals , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Humans , Hyaluronan Receptors/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Nude , MicroRNAs/genetics , Prognosis , Promoter Regions, Genetic , Signal Transduction , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , Trans-Activators/genetics , Xenograft Model Antitumor Assays
18.
Chem Biodivers ; 18(10): e2100244, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34310845

ABSTRACT

Hypericum attenuatum Choisy is a traditional Chinese herbal plant with multiple therapeutic effects. In this study, bioactivity-guided fractionation of Hypericum attenuatum Choisy extracts afforded three major flavonoids (including astragalin, guaijaverin and quercetin), which possessed α-Glucosidase inhibitory activity with IC50 values of 33.90±0.68 µM, 17.23±0.75 µM and 31.90±0.34 µM, respectively. Circular dichroism analysis revealed that all the three compounds could interact with α-glucosidase by inducing conformational changes of the enzyme. Molecular docking results indicated that they could bind to the active site in α-glucosidase, and the binding force was driven mainly by hydrogen bond. Additionally, isobolographic analysis of the interactions between two compounds showed that all the combinations presented a synergistic α-glucosidase inhibitory effect at lower concentrations, and the combination between quercetin and guaijaverin or astragalin exhibited the best synergistic effect. This research might provide a theoretical basis for the application of Hypericum attenuatum Choisy in treating hyperglycemia.


Subject(s)
Flavonoids/pharmacology , Glycoside Hydrolase Inhibitors/pharmacology , Hypericum/chemistry , Plant Extracts/pharmacology , alpha-Glucosidases/metabolism , Dose-Response Relationship, Drug , Flavonoids/chemistry , Flavonoids/isolation & purification , Glycoside Hydrolase Inhibitors/chemistry , Glycoside Hydrolase Inhibitors/isolation & purification , Humans , Molecular Structure , Plant Extracts/chemistry , Plant Extracts/isolation & purification , Thermodynamics
19.
Bioengineered ; 12(1): 2095-2105, 2021 12.
Article in English | MEDLINE | ID: mdl-34047682

ABSTRACT

As gene delivery tools, lentiviral vectors (LV) have broad applications in chimeric antigen receptor therapy (CAR-T). Large-scale production of functional LV is limited by the adherent, serum-dependent nature of HEK293T cells used in the manufacturing. HEK293T adherent cells were adapted to suspension cells in a serum-free medium to establish large-scale processes for functional LV production in a stirred bioreactor without micro-carriers. The results showed that 293 T suspension was successfully cultivated in F media (293 CD05 medium and SMM293-TII with 1:1 volume ratio), and the cells retained the capacity for LV production. After cultivation in a 5.5 L bioreactor for 4 days, the cells produced 1.5 ± 0.3 × 107 TU/mL raw LV, and the lentiviral transduction efficiency was 48.6 ± 2.8% in T Cells. The yield of LV equaled to the previous shake flask. The critical process steps were completed to enable a large-scale LV production process. Besides, a cryopreservation solution was developed to reduce protein involvement, avoid cell grafting and reduce process cost. The process is cost-effective and easy to scale up production, which is expected to be highly competitive.


Subject(s)
Bioreactors/virology , Genetic Vectors , Immunotherapy, Adoptive , Lentivirus , Virus Cultivation/methods , Genetic Vectors/genetics , Genetic Vectors/metabolism , HEK293 Cells , Humans , Lentivirus/genetics , Lentivirus/metabolism , T-Lymphocytes
20.
Biosci Biotechnol Biochem ; 85(6): 1395-1404, 2021 May 25.
Article in English | MEDLINE | ID: mdl-33784390

ABSTRACT

Liraglutide is an analog of human glucagon-like peptide-1 which play essential roles in regulation of glycolipid metabolism. To investigate role of lactic acid bacteria (LAB) in lipid-lowering effect of liraglutide, 40 mice were divided into normal food diet (NFD), high-fat food (HFD), 10.0 mg/kg/d simvastatin-treated HFD (SIM + HFD), 200 and 400 µg/kg/d liraglutide-treated HFD (LL + HFD and HL + HFD) groups for 5 weeks. We found that liraglutide could upregulate cholesterol 7α-hydroxylase (CYP7A1) and LDL-receptor (LDLR), whereas downregulate 3-hydroxy-3-methylglutaryl coenzyme A reductase (HMGCR). Besides, liraglutide enhance abundance of lactobacillaceae in gut of hyperlipidemic mice and increase bile tolerance ability of LAB by upregulating bile salt hydrolases, and the lysate of liraglutide-sensitive LAB could also directly downregulate HMGCR, the key enzyme in cholesterol synthesis, and inhibit hepatocyte steatosis. These findings might provide new theoretical guidance for clinical application of liraglutide and research and development of antiobesity, hypolipidemic, and cholesterol-lowering drugs or functional foods.


Subject(s)
Bile/metabolism , Hypolipidemic Agents/pharmacology , Lactobacillus/drug effects , Lactobacillus/metabolism , Liraglutide/pharmacology , Animals , Cholesterol/metabolism , Cholesterol 7-alpha-Hydroxylase/metabolism , Diet, High-Fat/adverse effects , Dose-Response Relationship, Drug , Male , Mice
SELECTION OF CITATIONS
SEARCH DETAIL
...