Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 16 de 16
3.
J Biol Chem ; 299(7): 104909, 2023 07.
Article En | MEDLINE | ID: mdl-37307917

Sustainable TGF-ß1 signaling drives organ fibrogenesis. However, the cellular adaptation to maintain TGF-ß1 signaling remains unclear. In this study, we revealed that dietary folate restriction promoted the resolution of liver fibrosis in mice with nonalcoholic steatohepatitis. In activated hepatic stellate cells, folate shifted toward mitochondrial metabolism to sustain TGF-ß1 signaling. Mechanistically, nontargeted metabolomics screening identified that α-linolenic acid (ALA) is exhausted by mitochondrial folate metabolism in activated hepatic stellate cells. Knocking down serine hydroxymethyltransferase 2 increases the bioconversion of ALA to docosahexaenoic acid, which inhibits TGF-ß1 signaling. Finally, blocking mitochondrial folate metabolism promoted liver fibrosis resolution in nonalcoholic steatohepatitis mice. In conclusion, mitochondrial folate metabolism/ALA exhaustion/TGF-ßR1 reproduction is a feedforward signaling to sustain profibrotic TGF-ß1 signaling, and targeting mitochondrial folate metabolism is a promising strategy to enforce liver fibrosis resolution.


Folic Acid , Liver Cirrhosis , Mitochondria , alpha-Linolenic Acid , Animals , Mice , alpha-Linolenic Acid/deficiency , alpha-Linolenic Acid/metabolism , Hepatic Stellate Cells/metabolism , Liver/cytology , Liver/metabolism , Liver/pathology , Liver Cirrhosis/complications , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Non-alcoholic Fatty Liver Disease/complications , Non-alcoholic Fatty Liver Disease/metabolism , Transforming Growth Factor beta1/metabolism , Folic Acid/metabolism , Mitochondria/metabolism , Folic Acid Deficiency/complications , Folic Acid Deficiency/metabolism , Signal Transduction , Feedback, Physiological
4.
Rev Esp Enferm Dig ; 115(7): 407-408, 2023 07.
Article En | MEDLINE | ID: mdl-37314145

Laparoscopic cholecystectomy (LC) is currently the standard procedure for the treatment of benign gallbladder diseases. Although the ligature clip may fall off and shift after surgery, relevant reports are rare. We describe the formation of common bile duct stone in an elderly female in which a metal clip displaced into the common bile duct 6 years after LC.


Cholecystectomy, Laparoscopic , Gallstones , Humans , Female , Aged , Cholecystectomy, Laparoscopic/methods , Gallstones/diagnostic imaging , Gallstones/surgery , Common Bile Duct/surgery , Surgical Instruments
11.
Toxicol Appl Pharmacol ; 420: 115530, 2021 06 01.
Article En | MEDLINE | ID: mdl-33845055

Chronic Kidney Disease (CKD) is a serious threat to human health. In addition, kidney fibrosis is a key pathogenic intermediate for the progression of CDK. Moreover, excessive activation of fibroblasts is key to the development of kidney fibrosis and this process is difficult to control. Notably, fraxinellone is a natural compound isolated from Dictamnus dasycarpus and has a variety of pharmacological activities, including hepatoprotective, anti-inflammatory and anti-cancer effects. However, the effect of fraxinellone on kidney fibrosis is largely unknown. The present study showed that fraxinellone could alleviate folic acid-induced kidney fibrosis in mice in a dose dependent manner. Additionally, the results revealed that fraxinellone could effectively down-regulate the expression of CUGBP1, which was highly up-regulated in human and murine fibrotic renal tissues. Furthermore, expression of CUGBP1 was selectively induced by the Transforming Growth Factor-beta (TGF-ß) through p38 and JNK signaling in kidney fibroblasts. On the other hand, downregulating the expression of CUGBP1 significantly inhibited the activation of kidney fibroblasts. In conclusion, these findings demonstrated that fraxinellone might be a new drug candidate and CUGBP1 could be a promising target for the treatment of kidney fibrosis.


Benzofurans/pharmacology , CELF1 Protein/metabolism , Fibroblasts/drug effects , Kidney Diseases/prevention & control , Kidney/drug effects , Animals , CELF1 Protein/genetics , Cell Line , Disease Models, Animal , Fibroblasts/metabolism , Fibroblasts/pathology , Fibrosis , Folic Acid , JNK Mitogen-Activated Protein Kinases/metabolism , Kidney/metabolism , Kidney/pathology , Kidney Diseases/chemically induced , Kidney Diseases/metabolism , Kidney Diseases/pathology , Male , Mice, Inbred C57BL , Rats , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
12.
Int Immunopharmacol ; 95: 107567, 2021 Jun.
Article En | MEDLINE | ID: mdl-33756225

Coronavirus disease 2019 (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has become a global pandemic. However, the mechanism of tissue tropism of SARS-CoV-2 remains unclear. Here, recombinant receptor-binding subdomain 1 of spike protein of SARS-CoV-2 (RBD-SD1) was used as a probe to investigate the potential tropism of SARS-CoV-2 in thirty-three types of normal human tissues. RBD-SD1 probe was observed to interact with cells in reported SARS-CoV-2 infected organs. Interestingly, the RBD-SD1 probe strongly interacted with bone marrow cells in an angiotensin-converting enzyme 2 (ACE2)-independent manner. In addition, SARS-CoV-2 induced the ACE2 mRNA expression in human primary bone marrow cells, suggesting human bone marrow cells may be sensitive to SARS-CoV-2 infection. Therefore, human bone marrow cells could be strongly infected by SARS-CoV-2, which may play an important role in the pathogenesis of COVID-19. These findings provide a deeper understanding of SARS-CoV-2 infection routes, thus contributing to the treatment of COVID-19.


SARS-CoV-2/chemistry , SARS-CoV-2/metabolism , Spike Glycoprotein, Coronavirus/chemistry , Spike Glycoprotein, Coronavirus/metabolism , Angiotensin-Converting Enzyme 2/biosynthesis , Angiotensin-Converting Enzyme 2/genetics , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Humans , Lung/cytology , Lung/metabolism , Primary Cell Culture , Protein Binding , Protein Domains , Up-Regulation
13.
Redox Biol ; 36: 101634, 2020 09.
Article En | MEDLINE | ID: mdl-32863213

Nonalcoholic steatohepatitis (NASH), the progressive form of nonalcoholic fatty liver disease (NAFLD), is becoming a common chronic liver disease with the characteristics of steatosis, inflammation and fibrosis. Macrophage plays an important role in the development of NASH. In this study, Annexin A5 (Anx A5) is identified with the special effect on hepatic macrophage phenotype shift from M1 to M2. And it is further demonstrated that Anx A5 significantly switches metabolic reprogramming from glycolysis to oxidative phosphorylation in activated macrophages. Mechanistically, the main target of Anx A5 in energy metabolism is confirmed to be pyruvate kinase M2 (PKM2). And we following reveal that Anx A5 directly interacts with PKM2 at ASP101, LEU104 and ARG106, inhibits phosphorylation of Y105, and promotes PKM2 tetramer formation. In addition, based on the results of PKM2 inhibitor (compound 3k) and the phosphorylated mutation (PKM2 (Y105E)), it is proved that Anx A5 exhibits the function in macrophage polarization dependently on PKM2 activity. In vivo studies also show that Anx A5 improves steatosis, inflammation and fibrosis in NASH mice due to specially regulating hepatic macrophages via interaction with PKM2. Therefore, we have revealed a novel function of Anx A5 in hepatic macrophage polarization and HFD-induced NASH, providing important insights into the metabolic reprogramming, which is important for NASH therapy.


Non-alcoholic Fatty Liver Disease , Animals , Annexin A5 , Liver/metabolism , Macrophage Activation , Macrophages/metabolism , Mice , Non-alcoholic Fatty Liver Disease/drug therapy , Non-alcoholic Fatty Liver Disease/genetics , Pyruvate Kinase/genetics , Pyruvate Kinase/metabolism
14.
Int J Nanomedicine ; 14: 8121-8132, 2019.
Article En | MEDLINE | ID: mdl-31632022

INTRODUCTION: Exosomes are important mediators of intercellular communication. Previously, we characterized circulating exosomal miR-425-3p as a non-invasive prognostic marker for predicting clinical response to platinum-based chemotherapy in patients with non-small cell lung cancer (NSCLC). METHODS: Circulating exosomal miR-425-3p was validated by qRT-PCR in paired serum samples from NSCLC patients during the course of platinum-based chemotherapy. Cell coculture was performed to examine the effects of exosomal miR-425-3p on the sensitivity of recipient A549 cells to cisplatin. Using bioinformatics, ChIP and luciferase reporter assays, the transcription factor essential for miR-425-3p expression was identified. Autophagic activity in the recipient cells was determined by Western blot and fluorescence microscopy. RESULTS: Higher levels of exosomal miR-425-3p were found in serum samples from the patients in tolerance versus those at baseline. An upward trend in the expression of circulating exosomal miR-425-3p was revealed during chemotherapy. Furthermore, the expression of exosomal miR-425-3p could be induced by cisplatin in NSCLC cells. Exosomes isolated from either cisplatin-treated or cisplatin-resistant NSCLC cells conferred chemoresistance to sensitive A549 cells in a miR-425-3p-dependent manner. Cisplatin-induced c-Myc was found to directly bind the miR-425-3p promoter and transactivated its expression. Exosomal miR-425-3p facilitated autophagic activation in the recipient cells by targeting AKT1, eventually leading to chemoresistance. DISCUSSION: Our results suggest that apart from a prognostic marker of treatment response, exosomal miR-425-3p might be a potential dynamic biomarker to tailor cisplatin resistance in NSCLC patients during the treatment and represent a promising therapeutic target for therapy-resistant NSCLC.


Autophagy/drug effects , Carcinoma, Non-Small-Cell Lung/pathology , Cisplatin/pharmacology , Drug Resistance, Neoplasm/drug effects , Exosomes/metabolism , Lung Neoplasms/pathology , MicroRNAs/metabolism , A549 Cells , Apoptosis/drug effects , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Cisplatin/therapeutic use , Exosomes/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , MicroRNAs/genetics , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Up-Regulation/drug effects , Up-Regulation/genetics , beta Catenin/metabolism
15.
EBioMedicine ; 48: 169-177, 2019 Oct.
Article En | MEDLINE | ID: mdl-31521609

BACKGROUND: Tumor mutations and tumor microenvironment are associated with resistance to cancer immunotherapies. However, peripheral T cell in effective anti-programmed death 1 (PD-1) antibody treatment is poorly understood. METHODS: Mass spectrometry and conventional flow cytometry were used to investigate peripheral blood cells isolated from patients. Furthermore, melanoma mouse model was performed to assess the role of CXCR3 signaling in anti-PD-1 antibody treatment. FINDINGS: We revealed a marked increase in the percentage of CXCR3+ T cells in the blood of cancer patients after the first pembrolizumab infusion. This percentage decreased after the second infusion in responsive patients, whereas a sustained high percentage of CXCR3+ T cells was observed in patients with progressive disease. A low percentage of CXCR3+ T cells presented in patients with stable disease or a partial response was confirmed by conventional flow cytometry. Intriguingly, blockade of CXCR3 signaling exacerbated tumor growth in mice. Intratumoral injection with recombinant CXCL9/10 plus intraperitoneal injection of anti-PD1 antibody inhibited the tumor growth in mice. INTERPRETATION: The dynamic changes in CXCR3+ T cells in blood may be a prognostic factor in anti-PD-1 immunotherapy, and promotion of CXCR3-mediated signaling may be beneficial to the anti-PD-1 therapy. FUND: This work was supported by the National Natural Science Foundation of China (Nos. 81722047, 81871944, 81670553, 81874317, 81572389, 81730100) and Jiangsu province key medical talents (Nos. ZDRCA2016026), The "Deng Feng" Distinguished Scholars Program, National Science & Technology Major Project "Key New Drug Creation and Manufacturing Program", China (Number: 2018ZX09201002), and the Fundamental Research Funds for the Central Universities (020814380117).


Antineoplastic Agents, Immunological/pharmacology , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Receptors, CXCR3/metabolism , Signal Transduction/drug effects , Animals , Antineoplastic Agents, Immunological/therapeutic use , Cell Line, Tumor , Disease Models, Animal , Humans , Melanoma/diagnosis , Melanoma/etiology , Melanoma/metabolism , Melanoma, Experimental , Mice , Receptors, CXCR3/genetics , T-Lymphocyte Subsets/drug effects , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Tumor Microenvironment/drug effects , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology
16.
Biochem Pharmacol ; 123: 63-72, 2017 Jan 01.
Article En | MEDLINE | ID: mdl-27751819

Interferon gamma (IFN-γ) signaling in T cells plays an important role in developing T helper 1 (Th1)-mediated inflammation. Selective regulation of IFN-γ signaling is an attractive strategy for treating Th1-mediated immune diseases. In this study, we aimed to explore possible means of targeting IFN-γ signaling by using small molecule compound. A synthetic small molecule FC9 was identified as it selectively inhibited IFN-γ signaling in T cells without suppressing interleukin 4 (IL-4) signaling. Furthermore, FC9 inhibited IFN-γ-induced Janus kinase 2 (JAK2) activation via competing with IFN-γ for binding to IFN-γ receptor 1 (IFN-γ R1). Interestingly, we found that FC9 bound to IFN-γ R1 and selectively suppressed Th1 but not Th2 immune response in T cells, resulting in an improvement in 2,4,6-trinitrobenzene sulfonic acid (TNBS)-induced colitis in mice. In conclusion, FC9-induced competitive blockade of IFN-γ R1 for selective inhibition of IFN-γ signaling, demonstrated a novel mean of targeting IFN-γ signaling. These findings could lead to increased options for the treatment of Crohn's disease and other Th1-mediated inflammatory diseases.


Colitis/prevention & control , Ergot Alkaloids/pharmacology , Indole Alkaloids/pharmacology , Interferon-gamma/metabolism , Receptors, Interferon/metabolism , Th1 Cells/immunology , Animals , Cell Differentiation , Colitis/immunology , Mice , Mice, Inbred C57BL , STAT1 Transcription Factor/metabolism , Signal Transduction , Th1 Cells/cytology , Interferon gamma Receptor
...