Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Nat Commun ; 14(1): 4436, 2023 07 22.
Article in English | MEDLINE | ID: mdl-37481670

ABSTRACT

Inhibition of immunocyte infiltration and activation has been suggested to effectively ameliorate nonalcoholic steatohepatitis (NASH). Paired immunoglobulin-like receptor B (PirB) and its human ortholog receptor, leukocyte immunoglobulin-like receptor B (LILRB2), are immune-inhibitory receptors. However, their role in NASH pathogenesis is still unclear. Here, we demonstrate that PirB/LILRB2 regulates the migration of macrophages during NASH by binding with its ligand angiopoietin-like protein 8 (ANGPTL8). Hepatocyte-specific ANGPTL8 knockout reduces MDM infiltration and resolves lipid accumulation and fibrosis progression in the livers of NASH mice. In addition, PirB-/- bone marrow (BM) chimeras abrogate ANGPTL8-induced MDM migration to the liver. And yet, PirB ectodomain protein could ameliorate NASH by sequestering ANGPTL8. Furthermore, LILRB2-ANGPTL8 binding-promoted MDM migration and inflammatory activation are also observed in human peripheral blood monocytes. Taken together, our findings reveal the role of PirB/LILRB2 in NASH pathogenesis and identify PirB/LILRB2-ANGPTL8 signaling as a potential target for the management or treatment of NASH.


Subject(s)
Non-alcoholic Fatty Liver Disease , Animals , Humans , Mice , Angiopoietin-Like Protein 8 , Macrophages , Membrane Glycoproteins , Monocytes , Receptors, Immunologic/genetics
2.
J Cell Mol Med ; 25(19): 9447-9459, 2021 10.
Article in English | MEDLINE | ID: mdl-34498380

ABSTRACT

Oestradiol (E2) is a critical factor for multiple systems' development during the embryonic period. Here, we aimed to investigate the effects of oestradiol on intrahepatic bile duct development, which may allow a better understanding of congenital bile duct dysplasia. DLK+ hepatoblasts were extracted from the C57BL/6CrSlc foetal mice and randomly divided into control group, oestradiol groups (1, 10, 100 nM) and oestradiol (10 nM) + DAPT (inhibitor of Notch signalling; 40 µM) group for in vitro experiments. For in vivo analysis, pregnant mice were divided into control group, oestradiol (intraperitoneal injection of 0.6 mg/kg/day) ± DAPT (subcutaneous injection of 10 mg/kg/day) groups and tamoxifen (gavage administration of 0.4 mg/kg/day) group. The results showed that oestradiol promoted hepatoblast differentiation into cholangiocytes and intrahepatic bile duct development during the embryonic period. Tamoxifen, an antioestrogenic drug, inhibited the above processes. Moreover, oestradiol promoted the expression of Notch signalling pathway-associated proteins and genes both in vitro and in vivo. Notably, DAPT addition inhibited the oestradiol-mediated effects. In conclusion, oestradiol can promote hepatoblast differentiation into cholangiocytes and intrahepatic bile duct development of C57BL/6CrSlc mice during embryonic period via the Notch signalling pathway.


Subject(s)
Bile Ducts, Intrahepatic/embryology , Bile Ducts, Intrahepatic/metabolism , Estradiol/metabolism , Organogenesis , Receptors, Notch/metabolism , Signal Transduction , Animals , Biomarkers , Cell Differentiation , Cells, Cultured , Estradiol/pharmacology , Gene Expression , Hepatocytes/metabolism , Immunohistochemistry , Immunophenotyping , Mice , Mice, Inbred C57BL , Organogenesis/drug effects , Stem Cells/metabolism
3.
Medicine (Baltimore) ; 98(39): e17305, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31574858

ABSTRACT

Until now, the recognition of sodium taurocholate cotransporting polypeptide (NTCP) deficiency has been mainly based on sporadic case reports. It was previously believed to be mildly symptomatic and resulting in mild liver dysfunction. However, to our knowledge, there have been no reports about the histopathologic and ultrastructural pathologic characteristics of the disease. The aim of the study was to analyze the clinical, histopathologic and ultrastructural pathologic characteristics of NTCP deficiency in 13 pediatric patients.From August 2012 to October 2018, this retrospective study conducted in the Department of Pediatrics of Tongji Hospital, China analyzed the data of 13 NTCP deficient patients with an SLC10A1 gene mutation. Except for NTCP deficiency, no other liver diseases were present in the patients, which was determined by both a genetic testing panel for jaundice and by reviewing medical records. The laboratory results, imaging, histopathologic, and ultrastructural pathologic information were recorded for analysis.The serum level of total bile acid was high in all 13 patients. All patients had adequate growth and development. Eight of the patients (8/13) presented with visible jaundice and 12 (12/13) were found to have hyperbilirubinemia. A needle liver biopsy was performed in 11 cases, which revealed slightly chronic inflammation in all 11 patients. One of the patients (1/13) was found to be suffering from gallstones.The data showed that although NTCP deficiency was often asymptomatic, some of the patients showed obvious clinical expressions, such as jaundice. Among the 13 pediatric patients with NTCP deficiency, both the biochemical and histopathologic features were similar to those of mild hepatocellular jaundice. In addition, it was determined that the clinical features in the patient with gallstones may have been caused by NTCP deficiency.


Subject(s)
Bile Acids and Salts/blood , Jaundice , Liver Diseases , Liver , Organic Anion Transporters, Sodium-Dependent , Symporters , Child Development , Child, Preschool , China/epidemiology , Genetic Testing/methods , Humans , Image-Guided Biopsy/methods , Infant , Jaundice/diagnosis , Jaundice/etiology , Liver/metabolism , Liver/pathology , Liver Diseases/diagnosis , Liver Diseases/genetics , Liver Diseases/physiopathology , Liver Diseases/therapy , Liver Function Tests/methods , Membrane Glycoproteins/metabolism , Mutation , Organic Anion Transporters, Sodium-Dependent/deficiency , Organic Anion Transporters, Sodium-Dependent/genetics , Pediatrics/methods , Retrospective Studies , Symporters/deficiency , Symporters/genetics
4.
Medicine (Baltimore) ; 98(19): e15593, 2019 May.
Article in English | MEDLINE | ID: mdl-31083246

ABSTRACT

RATIONALE: Neonatal cholestasis is one of the most serious diseases in infancy. Progressive familial intrahepatic cholestasis (PFIC) is a disease that leads to intrahepatic cholestasis. It is one of the common causes of neonatal cholestasis in addition to biliary atresia (BA). The differential diagnosis of neonatal cholestasis is clinically challenging for pediatricians. PATIENT CONCERNS: A 4-month-old female presented with severe jaundice, pruritus, and pale stool for 20 days. Abnormally strong echoes near the portal area, an abnormally small gallbladder with an irregularly stiff wall, and splenomegaly were identified on abdominal ultrasound. Blood tests showed elevated alanine aminotransferase, total bilirubin, conjugated bilirubin, gamma-glutamyltranspeptidase, and total bile acid levels. DIAGNOSIS: Intraoperative cholangiography showed BA. ABCB4 gene mutation IVS13+6G>A/G was confirmed by genetic testing. The patient was diagnosed with BA combined with PFIC3. INTERVENTIONS: Kasai portoenterostomy and ursodeoxycholic acid were used for treatment. OUTCOMES: Her clinical symptoms and blood tests improved gradually. No recurrence was noted during 1 year of follow-up. LESSONS: Additional examinations, such as genetic testing, should be considered in patients with BA who had refractory jaundice after Kasai portoenterostomy in order to exclude intrahepatic cholestasis.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B/deficiency , Biliary Atresia/complications , Biliary Atresia/diagnosis , Cholestasis, Intrahepatic/complications , Cholestasis, Intrahepatic/diagnosis , ATP Binding Cassette Transporter, Subfamily B/genetics , Biliary Atresia/genetics , Biliary Atresia/therapy , Biomarkers/blood , Cholestasis, Intrahepatic/genetics , Cholestasis, Intrahepatic/therapy , Diagnosis, Differential , Female , Humans , Infant , Jaundice/diagnosis , Jaundice/etiology , Jaundice/genetics , Jaundice/therapy
5.
Mol Med Rep ; 14(1): 790-6, 2016 Jul.
Article in English | MEDLINE | ID: mdl-27220549

ABSTRACT

Neuregulin 1 (Nrg1) is involved in multiple biological processes in the nervous system. The present study investigated changes in Nrg1 signaling in the major brain regions of mice subjected to lipopolysaccharide (LPS)-induced neuroinflammation. At 24 h post­intraperitoneal injection of LPS, mouse brain tissues, including tissues from the cortex, striatum, hippocampus and hypothalamus, were collected. Reverse transcription­polymerase chain reaction was used to determine the expression of Nrg1 and its receptors, Neu and ErbB4, at the mRNA level. Western blotting was performed to determine the levels of these proteins and the protein levels of phosphorylated extracellular signal-regulated kinases (Erk)1/2 and Akt1. Immunohistochemical staining was utilized to detect the levels of pNeu and pErbB4 in these regions. LPS successfully induced sites of neuroinflammation in these regions, in which changes in Nrg1, Neu and ErbB4 at the mRNA and protein levels were identified compared with controls. LPS induced a reduction in pNeu and pErbB4 in the striatum and hypothalamus, although marginally increased pErbB4 levels were found in the hippocampus. LPS increased the overall phosphorylation of Src but this effect was reduced in the hypothalamus. Moreover, increased phosphorylation of Akt1 was found in the striatum and hippocampus. These data suggest diverse roles for Nrg1 signaling in these regions during the process of neuroinflammation.


Subject(s)
Brain/metabolism , Encephalitis/etiology , Encephalitis/metabolism , Lipopolysaccharides/adverse effects , Neuregulin-1/metabolism , Signal Transduction , Animals , Brain/pathology , Disease Models, Animal , Encephalitis/pathology , Female , Gene Expression , Immunohistochemistry , Mice , Neuregulin-1/genetics , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Receptor, ErbB-2/genetics , Receptor, ErbB-2/metabolism , Receptor, ErbB-4/genetics , Receptor, ErbB-4/metabolism
6.
Curr Cancer Drug Targets ; 14(8): 685-99, 2014.
Article in English | MEDLINE | ID: mdl-25348016

ABSTRACT

AML with Mt NPM1 has relatively good responses to induction therapy. However, a proportion of NPMc+ AML cells cannot be cleared by conventional treatments. Therefore, we determined the therapeutic efficacy of deguelin that has demonstrated extensive biological activity with low toxicity. We previously reported that deguelin selectively reduces Mt NPM1, as well as induces differentiation and potentiates apoptosis in NPMc+ AML cells. Nevertheless, little information is available regarding the mechanism of deguelin-induced differentiation. Here, we investigated the role of deguelin in the induction of NPMc+ AML cell differentiation. Deguelin at the nontoxic concentration of 2 µM strongly inhibited cell growth but reduced apoptosis in OCI-AML3 cells carrying Mt NPM1, whereas the antiproliferative effect was minimal in OCIM2 cells harboring Wt NPM1. Compared with OCIM2 cells that showed no response, deguelin-treated OCI-AML3 cells exhibited the morphological features of granulocytic/monocytic differentiation, increased expression of differentiation antigens, and a nitroblue tetrazolium reduction activity. Induction of differentiation was associated with downregulation of Mt NPM1 and SIRT1, but not Wt NPM1, which was accompanied by an increase in CEBPß and G-CSFR expression, and further confirmed by sh-Mt NPM1 and sh-SIRT1. sh-Mt NPM1 treatment reduced SIRT1 expression, but did not change HDAC1/3 levels, suggesting that the decline of SIRT1 was partially accountable for the deguelin-induced, Mt-NPM1-related differentiation. Moreover, Mt NPM1 overexpression blocked deguelin-induced cell differentiation. Lastly, we showed that deguelin reduced the expression of Mt NPM1 via the ubiquitin-proteasome pathway. Taken together, our results suggest that deguelin may be a therapeutic candidate for NPMc+ AML.


Subject(s)
Histone Deacetylase 1/genetics , Histone Deacetylases/genetics , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Nuclear Proteins/genetics , Rotenone/analogs & derivatives , Sirtuin 1/genetics , Apoptosis/drug effects , Apoptosis/genetics , CCAAT-Enhancer-Binding Protein-beta/genetics , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/genetics , Down-Regulation/drug effects , Granulocytes/drug effects , Humans , Leukemia, Myeloid, Acute/pathology , Monocytes/drug effects , Nucleophosmin , Proteasome Endopeptidase Complex/genetics , Receptors, Granulocyte Colony-Stimulating Factor/genetics , Rotenone/pharmacology , Ubiquitin/genetics
7.
Acta Pharmacol Sin ; 35(6): 806-13, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24902788

ABSTRACT

AIM: Skewed cytoplasmic accumulation of NPM mutant protein (NPM1c+) is close related to leukemia pathogenesis. The aim of this study was to investigate whether oridonin, a diterpenoid isolated from the Chinese traditional medicine Rabdosia rubescens, was able to interfere with NPM1c+ protein trafficking and induce apoptosis in NPM1c+ acute myeloid leukemia cells in vitro. METHODS: OCI-AML3 cell line harboring a NPM1 gene mutation was examined. Cell growth was detected by MTT assay. Cell apoptosis was evaluated using flow cytometry and Hoechst 33258 staining. The expression and subcellular localization of relevant proteins were detected by Western blot and immunofluorescent staining. The mRNA expression was detected by RT-PCR. RESULTS: Oridonin (2-12 µmol/L) dose-dependently inhibited the viability of OCI-AML3 cells (the IC50 value was 3.27±0.23 µmol/L at 24 h). Moreover, oridonin induced OCI-AML3 cell apoptosis accompanied by activation of caspase-3 and nuclear translocation of NPM1c+ protein. Oridonin did not change the expression of Crm1 (the export receptor for nuclear export signal-containing proteins), but induced nuclear translocation of Crm1. Oridonin markedly increased the expression of nucleoporin98 (Nup98), which had an important role in Crm1-mediated nuclear protein export, and induced nuclear accumulation of Nup98. Furthermore, oridonin markedly increased the expression of p14arf and p53. CONCLUSION: In NPM1c+ leukemia cells, oridonin induces NPM1c+ protein translocation into the nucleus possibly via nuclear accumulation of Crm1; the compound markedly increases p53 and p14arf expression, which may contribute to cell apoptosis.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Diterpenes, Kaurane/pharmacology , Leukemia, Myeloid, Acute/drug therapy , Nuclear Proteins/genetics , Protein Transport/drug effects , Cell Line, Tumor , Humans , Isodon/chemistry , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Mutation , Nuclear Proteins/metabolism , Nucleophosmin
8.
Acta Pharmacol Sin ; 35(2): 257-66, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24374812

ABSTRACT

AIM: Mangiferin is glucosylxanthone extracted from plants of the Anacardiaceae and Gentianaceae families. The aim of this study was to investigate the effects of mangiferin on Nrf2-antioxidant response element (ARE) signaling and the sensitivity to etoposide of human myeloid leukemia cells in vitro. METHODS: Human HL-60 myeloid leukemia cells and mononuclear human umbilical cord blood cells (MNCs) were examined. Nrf2 protein was detected using immunofluorescence staining and Western blotting. Binding of Nrf2 to ARE was examined with electrophoretic mobility shift assay. The level of NQO1 was assessed with real-time RT-PCR and Western blotting. DCFH-DA was used to evaluate intracellular ROS level. Cell proliferation and apoptosis were analyzed using MTT and flow cytometry, respectively. RESULTS: Mangiferin (50 µmol/L) significantly increased Nrf2 protein accumulation in HL-60 cells, particularly in the nucleus. Mangiferin also enhanced the binding of Nrf2 to an ARE, significantly up-regulated NQO1 expression and reduced intracellular ROS in HL60 cells. Mangiferin alone dose-dependently inhibited the proliferation of HL-60 cells. Mangiferin (50 mol/L) did not attenuate etoposide-induced cytotoxicity in HL-60 cells, and combined treatment of mangiferin with low concentration of etoposide (0.8 µg/mL) even increased the cell inhibition rate. Nor did mangiferin change the rate of etoposide-induced apoptosis in HL-60 cells. In MNCs, mangiferin significantly relieved oxidative stress, but attenuated etoposide-induced cytotoxicity. CONCLUSION: Mangiferin is a novel Nrf2 activator that reduces oxidative stress and protects normal cells without reducing the sensitivity to etoposide of HL-60 leukemia cells in vitro. Mangiferin may be a potential chemotherapy adjuvant.


Subject(s)
Antioxidant Response Elements/genetics , Etoposide/pharmacology , Leukemia, Myeloid/drug therapy , NF-E2-Related Factor 2/metabolism , Signal Transduction/drug effects , Xanthones/pharmacology , Cell Line, Tumor , HL-60 Cells , Humans , Leukemia, Myeloid/genetics , Leukemia, Myeloid/metabolism , NF-E2-Related Factor 2/genetics , Signal Transduction/genetics
9.
Acta Pharmacol Sin ; 33(12): 1542-8, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23064721

ABSTRACT

AIM: To investigate the effects of betulinic acid (BA) on apoptosis and autophagic flux in multiple myeloma cells and the relationship between the two processes. METHODS: The proliferation of human multiple myeloma KM3 cells was measured with MTT assay. FITC/PI double-labeled flow cytometry (FCM) and Hoechst 33258 staining were used to analyze the cell apoptosis. Caspase 3, PARP, Beclin1, LC3-II, and P62 were detected using Western blotting. RESULTS: Treatment of KM3 cells with BA (5-25 µg/mL) suppressed the cell proliferation in time- and dose-dependent manners. The IC(50) values at 12, 24, and 36 h were 22.29, 17.36, and 13.06 µg/mL, respectively. BA treatment dose-dependently induced apoptosis of KM3 cells, which was associated with the activation of caspase 3. However, Z-DEVD-FMK, a specific inhibitor of caspase 3, did not decrease, but rather sensitized the cells to BA-induced apoptosis, suggesting an alternative mechanism involved. On other hand, BA treatment dose-dependently increased the accumulation of LC3-II and P62 in KM3 cells, representing the inhibition of autophagic flux. Furthermore, BA treatment dose-dependently downregulated the expression of Beclin 1, an important inducer of autophagy, in KM3 cells. In the presence of BA, Z-DEVD-FMK induced autophagy and increased the amount of LC3-II in KM3 cells, which may occur via attenuating BA-induced decrease in the level of Beclin 1. Similarly, rapamycin, an autophagy inducer, increased the amount of LC3-II in KM3 cells. In the presence of BA, rapamycin caused further increase in the amount of LC3-II. Furthermore, rapamycin sensitized BA-treated KM3 cells to apoptosis. CONCLUSION: The results demonstrate that BA induces apoptosis and blocks autophagic flux in KM3 cells. Furthermore, in addition to activation of caspase 3, the inhibition of autophagic flux also contributes to the BA-mediated apoptosis of KM3 cells.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Autophagy/drug effects , Multiple Myeloma/pathology , Triterpenes/pharmacology , Antineoplastic Agents, Phytogenic/therapeutic use , Blotting, Western , Caspase 3/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Humans , Multiple Myeloma/drug therapy , Oligopeptides/pharmacology , Pentacyclic Triterpenes , Triterpenes/therapeutic use , Betulinic Acid
SELECTION OF CITATIONS
SEARCH DETAIL
...