Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 10 de 10
1.
Biochem Genet ; 2024 Apr 11.
Article En | MEDLINE | ID: mdl-38602597

To investigate the expression level of miR-25-3p in patients with type 2 diabetes mellitus (T2DM) and diabetic nephropathy (DN), and its effect on proliferation, apoptosis and inflammatory response of mesangial cells cultured with high glucose. Blood samples of all clinical subjects were collected for RT-qPCR analysis to detect serum miR-25-3p levels. Human mesangial cells (HMCs) cultured with high glucose were used to construct DN model in vitro. MTT assay, flow cytometry and ELISA were used to evaluate the effects of miR-25-3p on the proliferation, apoptosis, and inflammatory response of DN cell models. Serum miR-25-3p was decreased in both T2DM group and DN group, but more in DN group. Serum miR-25-3p was positively correlated with eGFR and negatively correlated with UAER. The expression of miR-25-3p was reduced in HMCs induced by high glucose. Transfection of miR-25-3p mimic could significantly up-regulate the miR-25-3p level in HMCs. Besides, high glucose culture resulted in abnormal proliferation of HMCs, reduced apoptotic cells, and increased inflammation. The addition of miR-25-3p mimic significantly inhibited cell proliferation and promoted cell apoptosis and reduced the production of inflammatory factors. The abnormal reduction of serum miR-25-3p in DN indicates that it may be a potential biomarker for clinical diagnosis of DN. In in vitro experiments, miR-25-3p was involved in the progression of DN by regulating cell proliferation, apoptosis, and inflammatory response.

2.
Cell Rep ; 42(11): 113362, 2023 11 28.
Article En | MEDLINE | ID: mdl-37938970

Upregulation of FGL1 helps tumors escape from immune surveillance, and therapeutic antibodies targeting FGL1 have potential as another immune checkpoint inhibitor. However, the underlying mechanism of high FGL1 protein level in cancers is not well defined. Here, we report that FBXO38 interacts with and ubiquitylates FGL1 to negatively regulate its stability and to mediate cancer immune response. Depletion of FBXO38 markedly augments FGL1 abundance, not only suppressing CD8+ T cell infiltration and enhancing immune evasion of tumor but also increasing inflammation in mice. Importantly, we observe a negative correlation of FBXO38 with FGL1 and IL-6 in non-small cell lung cancer specimens. FGL1 and IL-6 levels positively correlate with TNM (tumor, lymph node, metastasis) stages, while FBXO38 and the infiltrating CD8+ T cells negatively correlate with TNM stages. Our study identifies a mechanism regulating FGL1 stability and a target to enhance the immunotherapy and suggests that the combination of anti-FGL1 and anti-IL-6 is a potential therapeutic strategy for cancer immunotherapy.


Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Animals , Mice , CD8-Positive T-Lymphocytes , Inflammation , Interleukin-6 , Ubiquitination
4.
Cell Discov ; 9(1): 84, 2023 Aug 08.
Article En | MEDLINE | ID: mdl-37550284

Tumor development, involving both cell growth (mass accumulation) and cell proliferation, is a complex process governed by the interplay of multiple signaling pathways. TET2 mainly functions as a DNA dioxygenase, which modulates gene expression and biological functions via oxidation of 5mC in DNA, yet whether it plays a role in regulating cell growth remains unknown. Here we show that TET2 suppresses mTORC1 signaling, a major growth controller, to inhibit cell growth and promote autophagy. Mechanistically, TET2 functions as a 5mC "eraser" by mRNA oxidation, abolishes YBX1-HuR binding and promotes decay of urea cycle enzyme mRNAs, thus negatively regulating urea cycle and arginine production, which suppresses mTORC1 signaling. Therefore, TET2-deficient tumor cells are more sensitive to mTORC1 inhibition. Our results uncover a novel function for TET2 in suppressing mTORC1 signaling and inhibiting cell growth, linking TET2-mediated mRNA oxidation to cell metabolism and cell growth control. These findings demonstrate the potential of mTORC1 inhibition as a possible treatment for TET2-deficient tumors.

5.
J Transl Med ; 21(1): 421, 2023 06 29.
Article En | MEDLINE | ID: mdl-37386465

BACKGROUND: We aimed to evaluate whether extracellular vesicles (EV)-derived microRNAs (miRNAs) can be used as biomarkers for advanced adenoma (AA) and colorectal cancer (CRC). METHODS: We detected the changes in the plasma EV-delivered miRNA profiles in healthy donor (HD), AA patient, and I-II stage CRC patient groups using miRNA deep sequencing assay. We performed the TaqMan miRNA assay using 173 plasma samples (two independent cohorts) from HDs, AA patients, and CRC patients to identify the candidate miRNA(s). The accuracy of candidate miRNA(s) in diagnosing AA and CRC was determined using the area under the receiver-operating characteristic curve (AUC) values. Logistic regression analysis was performed to evaluate the association of candidate miRNA(s) as an independent factor for the diagnosis of AA and CRC. The role of candidate miRNA(s) in the malignant progression of CRC was explored using functional assays. RESULTS: We screened and identified four prospective EV-delivered miRNAs, including miR-185-5p, which were significantly upregulated or downregulated in AA vs. HD and CRC vs. AA groups. In two independent cohorts, miR-185-5p was the best potential biomarker with the AUCs of 0.737 (Cohort I) and 0.720 (Cohort II) for AA vs. HD diagnosis, 0.887 (Cohort I) and 0.803 (Cohort II) for CRC vs. HD diagnosis, and 0.700 (Cohort I) and 0.631 (Cohort II) for CRC vs. AA diagnosis. Finally, we demonstrated that the upregulated expression of miR-185-5p promoted the malignant progression of CRC. CONCLUSION: EV-delivered miR-185-5p in the plasma of patients is a promising diagnostic biomarker for colorectal AA and CRC. Trial registration The study protocol was approved by the Ethics Committee of Changzheng Hospital, Naval Medical University, China (Ethics No. 2022SL005, Registration No. of China Clinical Trial Registration Center: ChiCTR220061592).


Adenoma , Colorectal Neoplasms , Extracellular Vesicles , MicroRNAs , Humans , Prospective Studies , MicroRNAs/genetics , Adenoma/diagnosis , Adenoma/genetics , Colorectal Neoplasms/diagnosis , Colorectal Neoplasms/genetics
6.
Heliyon ; 9(6): e16417, 2023 Jun.
Article En | MEDLINE | ID: mdl-37251444

With the progress of society, the health problems of pets have attracted more and more attention. Recent studies have shown that intestinal microflora and related fecal metabolites play a crucial role in the healthy growth of cats. However, the potential role and related metabolic characteristics of gut microbiota in different age groups of pet cats need to be further clarified. 16S rRNA gene sequencing was used to analyze the intestinal microbial composition of young and old cats. LC-MS metabonomic analysis is used to characterize the changes in the metabolic spectrum in feces. The potential relationship between intestinal microorganisms and metabolites, as well as the differences in different age groups, were studied. The species composition of intestinal microflora in the young group and old group is significantly different, T-test algorithm shows 36 different ASVs and 8 different genuses, while the Wilcoxon algorithm shows 81 different ASVs and 17 different genuses. The metabolomics analysis identified 537 kinds of fecal metabolites, which are rich in differences between young and old cats, and may be potential biomarkers indicating the health of cats. 16S rRNA analysis showed significant differences in fructose and mannose metabolism, while metabonomics KEGG analysis showed significant difference in choline metabolism in cancer. Our study compared the differences between the intestinal microbiome and fecal metabolites in young and old cats. This difference provides a new direction for further exploring the relationship between the composition and metabolism of intestinal microbiota in cats of different age groups. It also provides a reference for cat health research.

7.
Cancer Res ; 82(11): 2097-2109, 2022 06 06.
Article En | MEDLINE | ID: mdl-35176127

Inactivating mutations of von Hippel-Lindau (VHL) are highly prevalent in clear cell renal cell carcinoma (ccRCC). Improved understanding of the vulnerabilities of VHL-deficient ccRCC could lead to improved treatment strategies. The activity of DNA dioxygenase ten-eleven translocation (TET)2 is significantly reduced in multiple cancers by different mechanisms, but its role in ccRCC progression remains unclear. Here, we report that increased expression of TET2, but not TET1 and TET3, is negatively associated with tumor metastasis and advanced tumor stage and is positively associated with good prognosis uniquely in ccRCC among all 33 types of cancer in The Cancer Genome Atlas datasets. TET2 restrained glycolysis and pentose phosphate pathway metabolism in a VHL deficiency-dependent manner, thereby suppressing ccRCC progression. Notably, TET2 and VHL mutations tended to cooccur in ccRCC, providing genetic evidence that they cooperate to inhibit the progression of ccRCC. Mechanistically, TET2 was recruited by transcription factor HNF4α to activate FBP1 expression, which antagonized the function of hypoxia-inducible factor-1/2α (HIF1/2α) in metabolic reprogramming to impede ccRCC growth. Stimulating the TET2-FBP1 axis with vitamin C repressed the growth of VHL-deficient ccRCC with wild-type TET2 and increased the sensitivity to glycolysis inhibitors. Moreover, combined expression levels of the HNF4α-TET2-FBP1 axis served as a biomarker of prognosis in patients with ccRCC. This study reveals a unique function of TET2 in the suppression of tumor metabolism and HIF signaling, and it also provides therapeutic targets, potential drugs, and prognostic markers for the management of ccRCC. SIGNIFICANCE: The identification of TET2-mediated inhibition of HIF signaling and tumor metabolic reprogramming provides insights for new therapeutic strategies for VHL-deficient ccRCC.


Carcinoma, Renal Cell , Dioxygenases , Kidney Neoplasms , Basic Helix-Loop-Helix Transcription Factors/metabolism , Carcinoma, Renal Cell/pathology , Cell Line, Tumor , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Dioxygenases/genetics , Gene Expression Regulation, Neoplastic , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Kidney Neoplasms/pathology , Von Hippel-Lindau Tumor Suppressor Protein/genetics , Von Hippel-Lindau Tumor Suppressor Protein/metabolism
8.
Proc Natl Acad Sci U S A ; 119(8)2022 02 22.
Article En | MEDLINE | ID: mdl-35181605

Breast cancer is the most frequent malignancy in women worldwide, and triple-negative breast cancer (TNBC) patients have the worst prognosis and highest risk of recurrence. The therapeutic strategies for TNBC are limited. It is urgent to develop new methods to enhance the efficacy of TNBC treatment. Previous studies demonstrated that D-mannose, a hexose, can enhance chemotherapy in cancer and suppress the immunopathology of autoimmune diseases. Here, we show that D-mannose can significantly facilitate TNBC treatment via degradation of PD-L1. Specifically, D-mannose can activate AMP-activated protein kinase (AMPK) to phosphorylate PD-L1 at S195, which leads to abnormal glycosylation and proteasomal degradation of PD-L1. D-mannose-mediated PD-L1 degradation promotes T cell activation and T cell killing of tumor cells. The combination of D-mannose and PD-1 blockade therapy dramatically inhibits TNBC growth and extends the lifespan of tumor-bearing mice. Moreover, D-mannose-induced PD-L1 degradation also results in messenger RNA destabilization of DNA damage repair-related genes, thereby sensitizing breast cancer cells to ionizing radiation (IR) treatment and facilitating radiotherapy of TNBC in mice. Of note, the effective level of D-mannose can be easily achieved by oral administration in mice. Our study unveils a mechanism by which D-mannose targets PD-L1 for degradation and provides methods to facilitate immunotherapy and radiotherapy in TNBC. This function of D-mannose may be useful for clinical treatment of TNBC.


B7-H1 Antigen/metabolism , Mannose/pharmacology , Triple Negative Breast Neoplasms/drug therapy , AMP-Activated Protein Kinases/drug effects , AMP-Activated Protein Kinases/metabolism , Animals , B7-H1 Antigen/drug effects , Cell Line, Tumor , Female , Humans , Immunologic Factors/metabolism , Immunotherapy/methods , Lymphocytes, Tumor-Infiltrating/metabolism , Mannose/metabolism , Mice , Mice, Inbred BALB C , Phosphorylation , Proteolysis/drug effects , Radiotherapy/methods , T-Lymphocytes/metabolism , Triple Negative Breast Neoplasms/metabolism
9.
Nat Commun ; 10(1): 2925, 2019 07 02.
Article En | MEDLINE | ID: mdl-31266960

Bacteriophage Q protein engages σ-dependent paused RNA polymerase (RNAP) by binding to a DNA site embedded in late gene promoter and renders RNAP resistant to termination signals. Here, we report a single-particle cryo-electron microscopy (cryo-EM) structure of an intact Q-engaged arrested complex. The structure reveals key interactions responsible for σ-dependent pause, Q engagement, and Q-mediated transcription antitermination. The structure shows that two Q protomers (QI and QII) bind to a direct-repeat DNA site and contact distinct elements of the RNA exit channel. Notably, QI forms a narrow ring inside the RNA exit channel and renders RNAP resistant to termination signals by prohibiting RNA hairpin formation in the RNA exit channel. Because the RNA exit channel is conserved among all multisubunit RNAPs, it is likely to serve as an important contact site for regulators that modify the elongation properties of RNAP in other organisms, as well.


Bacteriophages/enzymology , Codon, Terminator/genetics , DNA-Directed RNA Polymerases/chemistry , DNA-Directed RNA Polymerases/metabolism , Transcription, Genetic , Viral Proteins/chemistry , Viral Proteins/metabolism , Bacteriophages/chemistry , Bacteriophages/genetics , Codon, Terminator/metabolism , Cryoelectron Microscopy , DNA-Directed RNA Polymerases/genetics , Escherichia coli/genetics , Escherichia coli/metabolism , Escherichia coli/virology , Promoter Regions, Genetic , Viral Proteins/genetics
10.
Brain Behav Immun ; 70: 293-304, 2018 05.
Article En | MEDLINE | ID: mdl-29548997

Neuropsychiatric (NP) involvement in systemic lupus erythematosus (SLE) severely impacts patients' quality of life and leads to a poor prognosis. The current therapeutic protocol, corticosteroid administration, can also induce neuropsychiatric disorders. FTY720 is an immunomodulator that selectively confines lymphocytes in lymph nodes and reduces autoreactive T cell recruitment to the central nervous system (CNS). This study aimed to identify a novel therapeutic strategy for NPSLE. B6.MRL-lpr mice were treated with oral administration of FTY720 (2 mg/kg) three times per week for 12 weeks, to evaluate its efficacy in a model of NPSLE. FTY720 significantly attenuated the impulsive and depression-like behavior of B6.MRL-lpr mice. Neuronal damage was reduced in the cortex, hippocampus, and amygdala of the FTY720-treated B6.MRL-lpr mice, as well as in TNF-α-treated HT22 cells. Additionally, FTY720 downregulated levels of inflammatory cytokines, and reduced the infiltration of T cells and neutrophils in the brain parenchyma. FTY720 also acted directly on cerebral endothelial cells and reduced the permeability of the blood-brain barrier (BBB) in B6.MRL-lpr mice, as evidenced by reduced central IgG and albumin levels. Finally, FTY720 significantly inhibited activation of PI3K/Akt/GSK3ß/p65 signaling, which further reduced the expression levels of adhesion molecules in bEND.3 cells treated with B6.MRL-lpr mouse serum. Collectively, our data indicate that oral administration of FTY720 at an early stage has beneficial effects in NPSLE-model B6.MRL-lpr mice, suggesting that it may represent an effective new therapeutic strategy for NPSLE.


Fingolimod Hydrochloride/pharmacology , Lupus Erythematosus, Systemic/immunology , Animals , Brain , Cytokines , Depression , Disease Models, Animal , Female , Fingolimod Hydrochloride/therapeutic use , Immunologic Factors/therapeutic use , Lupus Erythematosus, Systemic/therapy , Lymphocytes/drug effects , Mice , Mice, Inbred C57BL , Mice, Inbred MRL lpr , Quality of Life , Signal Transduction , T-Lymphocytes
...