Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters











Publication year range
2.
Int J Mol Sci ; 25(2)2024 Jan 16.
Article in English | MEDLINE | ID: mdl-38256161

ABSTRACT

The thymus is one of the most crucial immunological organs, undergoing visible age-related shrinkage. Thymic epithelial cells (TECs) play a vital role in maintaining the normal function of the thymus, and their degeneration is the primary cause of age-induced thymic devolution. Thymosin ß4 (Tß4) serves as a significant important G-actin sequestering peptide. The objective of this study was to explore whether Tß4 influences thymocyte differentiation by regulating the cytoskeletal rearrangement and mitochondrial transfer of TECs. A combination of H&E staining, immunofluorescence, transmission electron microscopy, RT-qPCR, flow cytometry, cytoskeletal immunolabeling, and mitochondrial immunolabeling were employed to observe the effects of Tß4 on TECs' skeleton rearrangement, mitochondrial transfer, and thymocyte differentiation. The study revealed that the Tß4 primarily regulates the formation of microfilaments and the mitochondrial transfer of TECs, along with the formation and maturation of double-negative cells (CD4-CD8-) and CD4 single-positive cells (CD3+TCRß+CD4+CD8-) thymocytes. This study suggests that Tß4 plays a crucial role in thymocyte differentiation by influencing the cytoskeletal rearrangement and mitochondrial transfer of TECs. These effects may be associated with Tß4's impact on the aggregation of F-actin. This finding opens up new avenues for research in the field of immune aging.


Subject(s)
Thymocytes , Thymosin , Cytoskeleton , Epithelial Cells , Actins
3.
Virol J ; 21(1): 35, 2024 01 31.
Article in English | MEDLINE | ID: mdl-38297280

ABSTRACT

BACKGROUND: Progressive hepatitis B virus (HBV) infection can result in cirrhosis, hepatocellular cancer, and chronic hepatitis. While antiviral drugs that are now on the market are efficient in controlling HBV infection, finding a functional cure is still quite difficult. Identifying host factors involved in regulating the HBV life cycle will contribute to the development of new antiviral strategies. Zinc finger proteins have a significant function in HBV replication, according to earlier studies. Zinc finger protein 148 (ZNF148), a zinc finger transcription factor, regulates the expression of various genes by specifically binding to GC-rich sequences within promoter regions. The function of ZNF148 in HBV replication was investigated in this study. METHODS: HepG2-Na+/taurocholate cotransporting polypeptide (HepG2-NTCP) cells and Huh7 cells were used to evaluate the function of ZNF148 in vitro. Northern blotting and real-time PCR were used to quantify the amount of viral RNA. Southern blotting and real-time PCR were used to quantify the amount of viral DNA. Viral protein levels were elevated, according to the Western blot results. Dual-luciferase reporter assays were used to examine the transcriptional activity of viral promoters. ZNF148's impact on HBV in vivo was investigated using an established rcccDNA mouse model. RESULTS: ZNF148 overexpression significantly decreased the levels of HBV RNAs and HBV core DNA in HBV-infected HepG2-NTCP cells and Huh7 cells expressing prcccDNA. Silencing ZNF148 exhibited the opposite effects in both cell lines. Furthermore, ZNF148 inhibited the activity of HBV ENII/Cp and the transcriptional activity of cccDNA. Mechanistic studies revealed that ZNF148 attenuated retinoid X receptor alpha (RXRα) expression by binding to the RXRα promoter sequence. RXRα binding site mutation or RXRα overexpression abolished the suppressive effect of ZNF148 on HBV replication. The inhibitory effect of ZNF148 was also observed in the rcccDNA mouse model. CONCLUSIONS: ZNF148 inhibited HBV replication by downregulating RXRα transcription. Our findings reveal that ZNF148 may be a new target for anti-HBV strategies.


Subject(s)
Hepatitis B virus , Hepatitis B , Animals , Humans , Mice , DNA, Viral/genetics , Hep G2 Cells , Hepatitis B virus/physiology , Transcription Factors/genetics , Transcription Factors/metabolism , Virus Replication
4.
Inflamm Regen ; 44(1): 3, 2024 Jan 08.
Article in English | MEDLINE | ID: mdl-38191481

ABSTRACT

BACKGROUND: Thymosin beta family has a significant role in promoting hair regeneration, but which type of T cells play a key role in this process has not been deeply studied. This research aimed to find out the subtypes of T cell that play key role in hair regeneration mediated by thymosin beta 15 (Tß15). METHODS: Ready-to-use adenovirus expressing mouse Tmsb15b (thymosin beta 15 overexpression, Tß15 OX) and lentivirus-Tß15 short hairpin RNA (Tß15 sh) were used to evaluate the role of Tß15 in hair regeneration and development. The effect of Th22 cells on hair regeneration was further studied by optimized Th22-skewing condition medium and IL-22 binding protein (IL-22BP, an endogenous antagonist of IL-22, also known as IL-22RA2) in both ex vivo culture C57BL/6J mouse skin and BALB/c nude mice transplanted with thymus organoid model. RESULTS: The results show that Tß15, the homologous of Tß4, can promote hair regeneration by increasing the proliferation activity of hair follicle cells. In addition, high-level expression of Tß15 can not only increase the number of Th22 cells around hair follicles but also accelerate the transformation of hair follicles to maturity. Consistent with the expected results, when the IL-22BP inhibitor was used to interfere with Th22, the process of hair regeneration was blocked. CONCLUSIONS: In conclusion, Th22 is the key effector cell of Tß15 inducing hair regeneration. Both Tß15 and Th22 may be the potential drug targets for hair regeneration.

5.
Phytomedicine ; 123: 155216, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38061285

ABSTRACT

BACKGROUND: Thymus is the most crucial organ connecting immunity and aging. The progressive senescence of thymic epithelial cells (TECs) leads to the involution of thymus under aging, chronic stress and other factors. Ligustilide (LIG) is a major active component of the anti-aging Chinese herbal medicine Angelica sinensis (Oliv.) Diels, but its role in preventing TEC-based thymic aging remains elusive. PURPOSE: This study explored the protective role of Ligustilide in alleviating ADM (adriamycin) -induced thymic immune senescence and its underlying molecular mechanisms. METHOD: The protective effect of Ligustilide on ADM-induced thymic atrophy was examined by mouse and organotypic models, and conformed by SA-ß-gal staining in TECs. The abnormal spatial distribution of TECs in the senescent thymus was analyzed using H&E, immunofluorescence and flow cytometry. The possible mechanisms of Ligustilide in ADM-induced thymic aging were elucidated by qPCR, fluorescence labeling and Western blot. The mechanism of Ligustilide was subsequently validated through actin polymerization inhibitor, genetic engineering to regulate Thymosin ß15 (Tß15) and Tß4 expression, molecular docking and ß Thymosin-G-actin cross-linking assay. RESULTS: At a 5 mg/kg dose, Ligustilide markedly ameliorated ADM-induced weight loss and limb grip weakness in mice. It also reversed thymic damage and restored positive selection impaired by ADM. In vitro, ADM disrupted thymic structure, reduced TECs number and hindered double negative (DN) T cell differentiation. Ligustilide counteracted these effects, promoted TEC proliferation and reticular differentiation, leading to an increase in CD4+ single positive (CD4SP) T cell proportion. Mechanistically, ADM diminished the microfilament quantity in immortalized TECs (iTECs), and lowered the expression of cytoskeletal marker proteins. Molecular docking and cross-linking assay revealed that Ligustilide inhibited the protein binding between G-actin and Tß15 by inhibiting the formation of the Tß15-G-actin complex, thus enhancing the microfilament assembly capacity in TECs. CONCLUSION: This study, for the first time, reveals that Ligustilide can attenuate actin depolymerization, protects TECs from ADM-induced acute aging by inhibiting the binding of Tß15 to G-actin, thereby improving thymic immune function. Moreover, it underscores the interesting role of Ligustilide in maintaining cytoskeletal assembly and network structure of TECs, offering a novel perspective for deeper understanding of anti thymic aging.


Subject(s)
4-Butyrolactone/analogs & derivatives , Actins , Thymosin , Mice , Animals , Actins/metabolism , Thymosin/pharmacology , Thymosin/metabolism , Molecular Docking Simulation , Epithelial Cells
6.
Biosci Rep ; 43(11)2023 11 30.
Article in English | MEDLINE | ID: mdl-37883219

ABSTRACT

OBJECTIVES: Alanine aminotransferase (ALT) expression is highly elevated in the serum of patients with hepatocellular carcinoma. However, the role of ALT isoenzymes in the total ALT activity remains unclear. In the present study, we systematically investigated the role of ALT isoenzymes in alcoholic and non-alcoholic fatty liver cancer. MATERIALS AND METHODS: The expression of ALT1 and ALT2 at the mRNA and protein levels in 25 paired primary liver cancer tissues was detected by reverse transcription quantitative PCR (RT-qPCR), Western blotting, and immunohistochemistry. Serum ALT activity was determined using an automated biochemical analyzer. RESULTS: The mRNA and protein expression levels of ALT1 and ALT2 were lower in the tissues of alcoholic and non-alcoholic fatty liver cancers than in the paracancerous tissues. Notably, ALT2 was highly expressed in non-alcoholic fatty liver cancer tissues compared with alcoholic fatty liver cancer tissues. Total serum ALT activity was mainly contributed by ALT1 in alcoholic fatty liver cancer, whereas ALT1 contributed only marginally more to the total ALT activity than ALT2 in non-alcoholic fatty liver cancer. ALT2/ALT1 ratio can well discriminate normal control group, alcoholic liver cancer and non-alcoholic liver cancer. CONCLUSION: ALT1 contributed more to the total ALT activity than ALT2 in both alcoholic and non-alcoholic fatty liver cancer. Serum ALT2 to ALT activity was higher in non-alcoholic fatty liver cancer than that in alcoholic fatty liver cancer. ALT2/ALT1 ratio has some diagnostic significance for alcoholic and non-alcoholic liver cancer.


Subject(s)
Fatty Liver, Alcoholic , Liver Neoplasms , Non-alcoholic Fatty Liver Disease , Humans , Alanine Transaminase , Isoenzymes/metabolism , Fatty Liver, Alcoholic/diagnosis , Fatty Liver, Alcoholic/genetics , Fatty Liver, Alcoholic/metabolism , Non-alcoholic Fatty Liver Disease/diagnosis , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/metabolism , Liver Neoplasms/diagnosis , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , RNA, Messenger/metabolism , Liver/metabolism
7.
Molecules ; 28(8)2023 Apr 17.
Article in English | MEDLINE | ID: mdl-37110771

ABSTRACT

Thymosin α1 (Tα1) is an immunostimulatory peptide that is commonly used as an immune enhancer in viral infectious diseases such as hepatitis B, hepatitis C, and acquired immune deficiency syndrome (AIDS). Tα1 can influence the functions of immune cells, such as T cells, B cells, macrophages, and natural killer cells, by interacting with various Toll-like receptors (TLRs). Generally, Tα1 can bind to TLR3/4/9 and activate downstream IRF3 and NF-κB signal pathways, thus promoting the proliferation and activation of target immune cells. Moreover, TLR2 and TLR7 are also associated with Tα1. TLR2/NF-κB, TLR2/p38MAPK, or TLR7/MyD88 signaling pathways are activated by Tα1 to promote the production of various cytokines, thereby enhancing the innate and adaptive immune responses. At present, there are many reports on the clinical application and pharmacological research of Tα1, but there is no systematic review to analyze its exact clinical efficacy in these viral infectious diseases via its modulation of immune function. This review offers an overview and discussion of the characteristics of Tα1, its immunomodulatory properties, the molecular mechanisms underlying its therapeutic effects, and its clinical applications in antiviral therapy.


Subject(s)
Acquired Immunodeficiency Syndrome , Thymosin , Humans , Thymalfasin , NF-kappa B , Toll-Like Receptor 2 , Toll-Like Receptor 7 , Acquired Immunodeficiency Syndrome/drug therapy
8.
Curr Protein Pept Sci ; 24(1): 78-88, 2023.
Article in English | MEDLINE | ID: mdl-36464872

ABSTRACT

Thymosin ß4 (Tß4) is the ß-thymosin (Tßs) with the highest expression level in human cells; it makes up roughly 70-80% of all Tßs in the human body. Combining the mechanism and activity studies of Tß4 in recent years, we provide an overview of the subtle molecular mechanism, pharmacological action, and clinical applications of Tß4. As a G-actin isolator, Tß4 inhibits the polymerization of G-actin by binding to the matching site of G-actin in a 1:1 ratio through conformational and spatial effects. Tß4 can control the threshold concentration of G-actin in the cytoplasm, influence the balance of depolymerization and polymerization of F-actin (also called Tread Milling of F-actin), and subsequently affect cell's various physiological activities, especially motility, development and differentiation. Based on this, Tß4 is known to have a wide range of effects, including regulation of inflammation and tumor metastasis, promotion of angiogenesis, wound healing, regeneration of hair follicles, promotion of the development of the nervous system, and improving bone formation and tooth growth. Tß4 therefore has extensive medicinal applications in many fields, and serves to preserve the kidney, liver, heart, brain, intestine, and other organs, as well as hair loss, skin trauma, cornea repairing, and other conditions. In this review, we focus on the mechanism of action and clinical application of Tß4 for its main biological functions.


Subject(s)
Actins , Thymosin , Humans , Actins/genetics , Actins/metabolism , Actin Cytoskeleton/metabolism , Thymosin/pharmacology , Thymosin/chemistry , Thymosin/metabolism , Wound Healing
9.
Cells ; 11(22)2022 11 19.
Article in English | MEDLINE | ID: mdl-36429107

ABSTRACT

The thymus is the most sensitive organ under various pathophysiological conditions, such as aging, starvation, and infection. As a key stromal cell for T cell development, it is well-known that thymic epithelial cells (TECs) play an important role in the thymus response to the external environment. Thymosin beta 15 (Tß15) is a G-actin binding protein secreted by TECs, it plays an important role in maintaining the dynamic balance of actin, angiogenesis, axonal formation, and wound healing, but the relationship between Tß15 and TECs is not clear yet. Here, we show the impact of Tß15 on the TEC's spatial development, as well as the T-cell differentiation and thymic output. As a result, TEC is the main effector cell of Tß15 in the thymus. Tß15 OX inhibits the chemotaxis of TECs to the medulla and subsequently blocks the positive selection of thymocytes from CD3+TCRß+CD4+CD8+ double positive cells to CD3+TCRß+CD4+CD8- single-positive (CD4SP) cells. Tß15-knockdown accelerates the reticular differentiation of astral TECs and medullary TECs. Importantly, mice implanted with Tß15-knockdown iTECs show high thymic output but low peripheral T cell maturity and activity. In a word, our results explain the role of Tß15 on the differentiation and function of TECs and provide a new perspective for understanding the process of thymus development and degeneration.


Subject(s)
Cytoskeletal Proteins , Thymosin , Animals , Mice , Epithelial Cells , Thymus Gland , Thymocytes
10.
Front Microbiol ; 13: 836446, 2022.
Article in English | MEDLINE | ID: mdl-35663860

ABSTRACT

Chronic hepatitis B (CHB) virus infection is one of the leading causes of cirrhosis and liver cancer. Although the major drugs against CHB including nucleos(t)ide analogs and PEG-interferon can effectively control human hepatitis B virus (HBV) infection, complete cure of HBV infection is quite rare. Targeting host factors involved in the viral life cycle contributes to developing innovative therapeutic strategies to improve HBV clearance. In this study, we found that the mRNA and protein levels of SIRT2, a class III histone deacetylase, were significantly upregulated in CHB patients, and that SIRT2 protein level was positively correlated with HBV viral load, HBsAg/HBeAg levels, HBcrAg, and ALT/AST levels. Functional analysis confirmed that ectopic SIRT2 overexpression markedly increased total HBV RNAs, 3.5-kb RNA and HBV core DNA in HBV-infected HepG2-Na+/taurocholate cotransporting polypeptide cells and primary human hepatocytes. In contrast, SIRT2 silencing inhibited HBV transcription and replication. In addition, we found a positive correlation between SIRT2 expression and HBV RNAs synthesis as well as HBV covalently closed circular DNA transcriptional activity. A mechanistic study suggested that SIRT2 enhances the activities of HBV enhancer I/HBx promoter (EnI/Xp) and enhancer II/HBc promoter (EnII/Cp) by targeting the transcription factor p53. The levels of HBV EnI/Xp and EnII/Cp-bound p53 were modulated by SIRT2. Both the mutation of p53 binding sites in EnI/Xp and EnII/Cp as well as overexpression of p53 abolished the effect of SIRT2 on HBV transcription and replication. In conclusion, our study reveals that, in terms of host factors, a SIRT2-targeted program might be a more effective therapeutic strategy for HBV infection.

11.
Cancer Lett ; 481: 1-14, 2020 07 01.
Article in English | MEDLINE | ID: mdl-32268166

ABSTRACT

UBE2L3 is a ubiquitin-conjugating protein belonging to the E2 family that consists of 153 amino acid residues. In this study, we found that UBE2L3 was generally upregulated in clinical HCC samples compared to non-tumour samples and that there was a strong association between high UBE2L3 expression and tumour size, clinical grade and prognosis in HCC patients. UBE2L3 depletion inhibited the proliferation and induced the apoptosis of HCC cells. At the molecular level, we observed that UBE2L3 depletion enhanced the protein stability of GSK3ß, thus promoting the expression and activation of GSK3ß. Subsequently, activated GSK3ß phosphorylated p65 and promoted its nuclear translocation to increase the expression of target genes, including PUMA, Bax, Bim, Bad, and Bid. In vivo, knockout of UBE2L3 in HCC cells inhibited tumour growth in orthotopic liver injection nude mouse models. Moreover, inhibition of p65 or GSK3ß significantly restored the effects induced by UBE2L3 knockout in HCC. Together, this study reveals the stimulatory effect of UBE2L3 on HCC cell proliferation, suggesting that UBE2L3 may be an important pro-tumorigenic factor in liver carcinogenesis and a potential therapeutic target of HCC.


Subject(s)
Apoptosis/genetics , Carcinoma, Hepatocellular/genetics , Glycogen Synthase Kinase 3 beta/genetics , Liver Neoplasms/genetics , Signal Transduction/genetics , Transcription Factor RelA/genetics , Ubiquitin-Conjugating Enzymes/genetics , Animals , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Proliferation/genetics , Humans , Liver Neoplasms/pathology , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Phosphorylation/genetics , Up-Regulation/genetics
12.
Oncol Lett ; 17(6): 5030-5038, 2019 Jun.
Article in English | MEDLINE | ID: mdl-31186714

ABSTRACT

Hepatocellular carcinoma (HCC) is one of the leading causes of cancer-associated mortalities worldwide. The role of ornithine transcarbamylase (OTC) in HCC remains unclear. In the present study, the expression of OTC in HCC was analyzed based on datasets from the Gene Expression Omnibus database of the National Center for Biotechnology Information and further confirmed by immunohistochemistry, western blotting analysis and reverse transcription-quantitative polymerase chain reaction assays on clinical samples and cell lines. Furthermore, the associations between OTC expression and clinicopathological parameters as well as clinical outcome, including the overall and disease-free survival rates were analyzed. Finally, the effect of OTC on HCC cells was measured using proliferation, bromodeoxyuridine and colony-formation assays. Lower OTC expression was observed in HCC cells and tissues compared with primary human hepatocytes. Further investigation demonstrated that low expression of OTC in HCC was associated with larger tumor size and advanced grade. A Kaplan-Meier analysis revealed that patients with lower levels of OTC exhibited shorter overall and disease-free survival times. Notably, OTC silencing with RNA interference facilitated cell proliferation in HCC SK-Hep-1 and Huh-7 cells. However, overexpression of OTC led to inhibition of cell proliferation. In conclusion, the present study identified a novel role of OTC in HCC development, providing a potential novel therapeutic target for this disease.

13.
Cancer Lett ; 451: 156-167, 2019 06 01.
Article in English | MEDLINE | ID: mdl-30867140

ABSTRACT

NAD(P)H: quinone oxidoreductase 1 (NQO1) is an antioxidant enzyme which is associated with poor prognosis in human breast, colon, lung and liver cancers. However, the molecular mechanisms underlying the pro-tumorigenic function of NQO1 remains unclear. This study investigated the function of NQO1 in the context of hepatocellular carcinoma (HCC) development. We found that NQO1 was frequently up-regulated in human liver cancer, and its high expression level was correlated with the tumor stage and low survival rate of HCC patients. Loss-of-function of NQO1 inhibited growth in HCC cells with increased apoptosis in vitro, and suppressed orthotopic tumorigenicity in vivo. Mechanistically, high level of NQO1 in HCC cells enhanced protein stability of X-linked inhibitor of apoptosis protein (XIAP) by increasing its phosphorylation at Ser 87. Reintroduction of wile type XIAP and the phospho-mimic mutants XIAPS87D significantly reversed NQO1 knock-down/out induced growth inhibition and apoptosis. In mouse model with orthotopically implanted hepatocarcinoma, NQO1 suppression and NQO1 inhibitor suppressed tumor growth and induced apoptosis. NQO1 plays an important role in sustaining HCC cell proliferation and may thus act as a potential therapeutic target in HCC treatment.


Subject(s)
Apoptosis , Carcinoma, Hepatocellular/enzymology , Liver Neoplasms/enzymology , NAD(P)H Dehydrogenase (Quinone)/metabolism , X-Linked Inhibitor of Apoptosis Protein/metabolism , Animals , Carcinoma, Hepatocellular/pathology , Cell Line, Transformed , Humans , Liver Neoplasms/pathology , Male , Mice , Mice, Inbred BALB C , NAD(P)H Dehydrogenase (Quinone)/genetics , Phosphorylation
14.
Hepatology ; 68(4): 1260-1276, 2018 10.
Article in English | MEDLINE | ID: mdl-29624717

ABSTRACT

Hepatitis B virus (HBV) infection remains a major health problem worldwide. Maintenance of the covalently closed circular DNA (cccDNA), which serves as a template for HBV RNA transcription, is responsible for the failure of eradicating chronic HBV during current antiviral therapy. cccDNA is assembled with cellular histone proteins into chromatin, but little is known about the regulation of HBV chromatin by histone posttranslational modifications. In this study, we identified silent mating type information regulation 2 homolog 3 (SIRT3) as a host factor restricting HBV transcription and replication by screening seven members of the sirtuin family, which is the class III histone deacetylase. Ectopic SIRT3 expression significantly reduced total HBV RNAs, 3.5-kb RNA, as well as replicative intermediate DNA in HBV-infected HepG2-Na+ /taurocholate cotransporting polypeptide cells and primary human hepatocytes. In contrast, gene silencing of SIRT3 promoted HBV transcription and replication. A mechanistic study found that nuclear SIRT3 was recruited to the HBV cccDNA, where it deacetylated histone 3 lysine 9. Importantly, occupancy of SIRT3 on cccDNA could increase the recruitment of histone methyltransferase suppressor of variegation 3-9 homolog 1 to cccDNA and decrease recruitment of SET domain containing 1A, leading to a marked increase of trimethyl-histone H3 (Lys9) and a decrease of trimethyl-histone H3 (Lys4) on cccDNA. Moreover, SIRT3-mediated HBV cccDNA transcriptional repression involved decreased binding of host RNA polymerase II and transcription factor Yin Yang 1 to cccDNA. Finally, hepatitis B viral X protein could relieve SIRT3-mediated cccDNA transcriptional repression by inhibiting both SIRT3 expression and its recruitment to cccDNA. CONCLUSION: SIRT3 is a host factor epigenetically restricting HBV cccDNA transcription by acting cooperatively with histone methyltransferase; these data provide a rationale for the use of SIRT3 activators in the prevention or treatment of HBV infection. (Hepatology 2018).


Subject(s)
DNA, Viral/genetics , Epigenesis, Genetic/genetics , Hepatitis B/genetics , PR-SET Domains/genetics , Sirtuin 3/genetics , Virus Replication/genetics , DNA, Complementary/genetics , Hepatitis B/physiopathology , Hepatitis B virus/genetics , Histone Methyltransferases/metabolism , Humans , Real-Time Polymerase Chain Reaction/methods , Sensitivity and Specificity
15.
J Gen Virol ; 99(5): 645-654, 2018 05.
Article in English | MEDLINE | ID: mdl-29561254

ABSTRACT

Hepatitis B virus (HBV) infection is a major health problem worldwide. Interleukin-35 (IL-35) is a definite immunosuppressive cytokine belonging to the IL-12 family. Nevertheless, the role of IL-35 in HBV replication remains elusive. In this study, we found that the level of HBV DNA replicative intermediates detected by qPCR and Southern blotting analysis was significantly increased by rhIL-35 in a dose-dependent manner. Moreover, HBV 3.5 kb mRNA levels were up-regulated by rhIL-35. The HBV core protein level as well as the HBsAg and HBeAg secretion levels were also increased by rhIL-35. Moreover, a mechanistic study demonstrated that IL-35 promoted HBV replication by enhancing the HBV core promoter activity. Importantly, hepatocyte nuclear factor 4α (HNF4α) was probably the target of IL-35. Mutation of the HNF4α-binding site on HBV core promoter or silencing HNF4α abolished the enhancement of HBV replication induced by IL-35. Finally, rhIL-35 was able to increase HBV replication in HBV transgenic mice. Taken together, our findings demonstrated that IL-35 has a novel role in HBV replication.


Subject(s)
Hepatitis B virus/drug effects , Hepatitis B virus/genetics , Hepatocyte Nuclear Factor 4/genetics , Interleukins/pharmacology , Virus Replication/drug effects , Animals , Gene Expression Regulation , Gene Silencing , Hep G2 Cells , Hepatitis B/immunology , Hepatitis B Surface Antigens/genetics , Hepatitis B e Antigens/genetics , Hepatitis B virus/physiology , Hepatocyte Nuclear Factor 4/antagonists & inhibitors , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Promoter Regions, Genetic , RNA, Messenger , Recombinant Proteins/pharmacology , Signal Transduction , Up-Regulation
16.
Biochim Biophys Acta Gen Subj ; 1862(3): 440-450, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29107812

ABSTRACT

BACKGROUND: The functions of autism-associated Neuroligins (Nlgs) are modulated by their post-translational modifications, such as proteolytic cleavage. A previous study has shown that there are different endogenous forms of DNlg3 in Drosophila, indicating it may undergo proteolytic processing. However, the molecular mechanism underlying DNlg3 proteolytic processing is unknown. Here, we report a novel proteolytic mechanism that is essential for DNlg3 maturation and function in the nervous system. METHODS: Molecular cloning, cell culture, immunohistochemistry, western blotting and genetic studies were employed to map the DNlg3 cleavage region, identify the protease and characterize the cleavage manner. Behavior analysis, immunohistochemistry and genetic manipulations were employed to study the functions of different DNlg3 forms in the nervous system and neuromuscular junction (NMJs). RESULTS: Tumor necrosis factor α-converting enzyme (TACE) cleaved DNlg3 exclusively at its extracellular acetylcholinesterase-like domain to generate the N-terminal fragment and the short membrane-anchored fragment (sDNlg3). DNlg3 was constitutively processed in an activity-independent manner. Interestingly, DNlg3 was cleaved intracellularly in the Golgi apparatus before it arrived at the cell surface, a unique cleavage mechanism that is distinct from 'conventional' ectodomain shedding of membrane proteins, including rodent Nlg1. Genetic studies showed that sDNlg3 was essential for maintaining proper locomotor activity in Drosophila. CONCLUSIONS: Our results revealed a unique cleavage mechanism of DNlg3 and a neuron-specific role for DNlg3 maturation which is important in locomotor activity. GENERAL SIGNIFICANCE: Our study provides a new insight into a cleavage mechanism of Nlgs maturation in the nervous system.


Subject(s)
ADAM17 Protein/metabolism , Cell Adhesion Molecules, Neuronal/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Membrane Proteins/metabolism , Nerve Tissue Proteins/metabolism , Protein Processing, Post-Translational , ADAM17 Protein/genetics , Animals , Animals, Genetically Modified , Cell Adhesion Molecules, Neuronal/genetics , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Drosophila melanogaster/physiology , Gene Knockout Techniques , Golgi Apparatus/metabolism , Locomotion/physiology , Membrane Proteins/genetics , Nerve Tissue Proteins/genetics , Neurons/enzymology , Organ Specificity , Peptide Fragments/metabolism , Protein Domains
17.
J Mol Cell Biol ; 9(3): 231-242, 2017 06 01.
Article in English | MEDLINE | ID: mdl-28498949

ABSTRACT

Neuroligins (Nlgs) are transmembrane cell adhesion molecules playing essential roles in synapse development and function. Genetic mutations in neuroligin genes have been linked with some neurodevelopmental disorders such as autism. These mutated Nlgs are mostly retained in the endoplasmic reticulum (ER). However, the mechanisms underlying normal Nlg maturation and trafficking have remained largely unknown. Here, we found that Drosophila neuroligin 2 (DNlg2) undergoes proteolytic cleavage in the ER in a variety of Drosophila tissues throughout developmental stages. A region encompassing Y642-T698 is required for this process. The immature non-cleavable DNlg2 is retained in the ER and non-functional. The C-terminal fragment of DNlg2 instead of the full-length or non-cleavable DNlg2 is able to rescue neuromuscular junction defects and GluRIIB reduction induced by dnlg2 deletion. Intriguingly, the autism-associated R598C mutation in DNlg2 leads to similar marked defects in DNlg2 proteolytic process and ER export, revealing a potential role of the improper Nlg cleavage in autism pathogenesis. Collectively, our findings uncover a specific mechanism that controls DNlg2 maturation and trafficking via proteolytic cleavage in the ER, suggesting that the perturbed proteolytic cleavage of Nlgs likely contributes to autism disorder.


Subject(s)
Cell Adhesion Molecules, Neuronal/metabolism , Drosophila Proteins/metabolism , Endoplasmic Reticulum/metabolism , Nerve Tissue Proteins/metabolism , Animals , Animals, Genetically Modified , Autistic Disorder/genetics , Cell Adhesion Molecules, Neuronal/genetics , Drosophila/metabolism , Drosophila Proteins/genetics , Humans , Mutation , Nerve Tissue Proteins/genetics , Neuromuscular Junction/metabolism , Protein Transport
18.
Biochem Biophys Res Commun ; 485(4): 713-719, 2017 04 15.
Article in English | MEDLINE | ID: mdl-28238784

ABSTRACT

SIRT6 is a class III histone deacetylase that has been implicated in HCC development. We previously reported that SIRT6 potentiated apoptosis evasion in hepatocellular carcinoma by inhibiting both Bax expression and mitochondrial translocalization. However, the mechanism underlying SIRT6-mediated inhibition of Bax mitochondrial localization remains elusive. In this study, we found that although SIRT6 had no effect on the expression level of Ku70, SIRT6 could interact with Ku70 and deacetylate it. The increased acetylation of Ku70 in SIRT6-depleted cells disrupt its interaction with Bax, which finally resulted in Bax mitochondrial translocalization. Furthermore, lysine K542 on Ku70 was the target for deacetylation by SIRT6. Ku70K542Q mutation abolished suppression of association between Ku70 and Bax and caused redistribution of Bax to the cytosol in SIRT6-depleted cells. Finally, Ku70K542Q mutation could reversed the inhibition of growth and apoptosis promotion mediated by SIRT6 silencing. Together, our findings revealed SIRT6 could block the mitochondrial translocation of Bax and decrease the apoptotic ratio of HCC cells by deacetylation of Ku70. SIRT6 may serve as a promising target for developing targeted therapies for HCC in the future.


Subject(s)
Apoptosis , Ku Autoantigen/metabolism , Sirtuins/metabolism , bcl-2-Associated X Protein/metabolism , Acetylation , Blotting, Western , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Gene Expression , Humans , Ku Autoantigen/genetics , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Lysine/genetics , Lysine/metabolism , Microscopy, Fluorescence , Mitochondria/metabolism , Mutation , Protein Binding , Protein Transport/genetics , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Sirtuins/genetics , bcl-2-Associated X Protein/genetics
19.
Oncotarget ; 7(31): 50117-50130, 2016 Aug 02.
Article in English | MEDLINE | ID: mdl-27367026

ABSTRACT

SIRT3, a class III histone deacetylase, has been implicated in various cancers as a novel therapeutic target. In hepatocellular carcinoma (HCC), we previously reported that SIRT3 induced cell apoptosis by regulating GSK-3ß/Bax signaling pathway. Downregulation of SIRT3 in HCC cells facilitates tumor cell survival. In this study, we found that chemotherapeutic agents (doxorubicin, cisplatin and epirubicin) and sorafenib treatment downregulated SIRT3 mRNA and protein levels in three HCC cell lines. MTS assay found that SIRT3 overexpression sensitized liver cancer cells to chemotherapeutic agents and sorafenib in SMMC-7721, Huh-7 and PLC/PRF/5 cell lines. Moreover, SIRT3 overexpression promoted chemotherapeutic agents-induced or sorafenib-induced apoptosis as evidenced by flow cytometry, enhanced PARP cleavage and enhanced Caspase-9 cleavage in three HCC cells. In contrast, SIRT3 silencing increased drug resistance of HCC cells to chemotherapeutic agents. Mechanistic study found that SIRT3 downregulated the mRNA and protein levels of glutathione S-transferase pi 1 (GSTP1), which is a member of phase II detoxification enzymes families involved in metabolizing for chemotherapeutic agents. Moreover, SIRT3 decreased the amount of GSTP1 that was associated with JNK, which finally contributed the activation of JNK activity and activation of downstream target c-Jun and Bim. Importantly, GSTP1 overexpression or JNK inhibitor abolished SIRT3-induced apoptosis in HCC cells exposed to chemotherapeutic agents. Finally, there was a negative correlation between SIRT3 expression and GSTP1 expression in human HCC tissues. Together, our findings revealed SIRT3 could enhance the drug sensitivity of HCC cells to an array of chemotherapeutic agents. SIRT3 may serve as a potential target for improving the chemosensitivity of HCC patients.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Glutathione S-Transferase pi/metabolism , Liver Neoplasms/metabolism , MAP Kinase Kinase 4/metabolism , Sirtuin 3/metabolism , Antineoplastic Agents/pharmacology , Apoptosis , Carcinoma, Hepatocellular/drug therapy , Cell Line, Tumor , Cell Separation , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Glutathione , Glycogen Synthase Kinase 3/metabolism , Humans , Liver Neoplasms/drug therapy , MAP Kinase Signaling System/drug effects , Niacinamide/analogs & derivatives , Niacinamide/pharmacology , Phenylurea Compounds/pharmacology , RNA, Messenger/metabolism , Signal Transduction , Sorafenib
20.
PLoS One ; 11(3): e0150961, 2016.
Article in English | MEDLINE | ID: mdl-26950437

ABSTRACT

BACKGROUND/AIM: The hepatitis B virus (HBV) infection is accompanied by the induction of oxidative stress, especially mediated by HBV X protein (HBx). Oxidative stress has been implicated in a series of pathological states, such as DNA damage, cell survival and apoptosis. However, the host factor by which cells protect themselves under this oxidative stress is poorly understood. METHODOLOGY/PRINCIPAL FINDINGS: In this study, we first confirmed that HBV infection significantly induced oxidative stress. Moreover, viral protein HBx plays a major role in the oxidative stress induced by HBV. Importantly, we found that mitochondrial protein SIRT3 overexpression could decrease reactive oxygen species (ROS) induced by HBx while SIRT3 knockdown increased HBx-induced ROS. Importantly, SIRT3 overexpression abolished oxidative damage of HBx-expressing cells as evidenced by γH2AX and AP sites measurements. In contrast, SIRT3 knockdown promoted HBx-induced oxidative damage. In addition, we also observed that oxidant H2O2 markedly promoted HBV replication while the antioxidant N-acetyl-L-cysteine (NAC) inhibited HBV replication. Significantly, SIRT3 overexpression inhibited HBV replication by reducing cellular ROS level. CONCLUSIONS/SIGNIFICANCE: Collectively, these data suggest HBx expression induces oxidative stress, which promotes cellular oxidative damage and viral replication during HBV pathogenesis. Mitochondrial protein SIRT3 protected HBx expressing-cells from oxidative damage and inhibited HBV replication possibly by decreased cellular ROS level. These studies shed new light on the physiological significance of SIRT3 on HBx-induced oxidative stress, which can contribute to the liver pathogenesis.


Subject(s)
Oxidative Stress , Sirtuin 3/metabolism , Trans-Activators/genetics , Cell Line , Gene Expression , Hepatitis B virus/metabolism , Hepatitis B virus/physiology , Humans , Liver/cytology , Liver/metabolism , Liver/virology , Reactive Oxygen Species/metabolism , Viral Regulatory and Accessory Proteins , Virus Replication
SELECTION OF CITATIONS
SEARCH DETAIL