Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
Add more filters

Publication year range
1.
J Virol ; 96(2): e0159721, 2022 01 26.
Article in English | MEDLINE | ID: mdl-34757838

ABSTRACT

Porcine reproductive and respiratory syndrome virus (PRRSV) is a major economically significant pathogen and has evolved several strategies to evade host antiviral response and provide favorable conditions for survival. In the present study, we demonstrated that a host microRNA, miR-376b-3p, was upregulated by PRRSV infection through the viral components, nsp4 and nsp11, and that miR-376b-3p can directly target tripartite motif-containing 22 (TRIM22) to impair its anti-PRRSV activity, thus facilitating the replication of PRRSV. Meanwhile, we found that TRIM22 induced degradation of the nucleocapsid protein (N) of PRRSV by interacting with N protein to inhibit PRRSV replication, and further study indicated that TRIM22 could enhance the activation of the lysosomal pathway by interacting with LC3 to induce lysosomal degradation of N protein. In conclusion, PRRSV increased miR-376b-3p expression and hijacked the host miR-376b-3p to promote PRRSV replication by impairing the antiviral effect of TRIM22. Therefore, our finding outlines a novel strategy of immune evasion exerted by PRRSV, which is helpful for better understanding the pathogenesis of PRRSV. IMPORTANCE Porcine reproductive and respiratory syndrome virus (PRRSV) causes enormous economic losses each year in the swine industry worldwide. MicroRNAs (miRNAs) play important roles during viral infections via modulating the expression of viral or host genes at the posttranscriptional level. TRIM22 has recently been identified as a key restriction factor that inhibited the replication of a number of human viruses, such as HIV, encephalomyocarditis virus (ECMV), hepatitis C virus (HCV), HBV, influenza A virus (IAV), and respiratory syncytial virus (RSV). In this study, we showed that host miR-376b-3p could be upregulated by PRRSV and functioned to impair the anti-PRRSV role of TRIM22 to facilitate PRRSV replication. Meanwhile, we found that TRIM22 inhibited the replication of PRRSV by interacting with viral N protein and accelerating its degradation through the lysosomal pathway. Collectively, the findings reveal a novel mechanism that PRRSV used to exploit the host miR-376b-3p to evade antiviral responses and provide new insight into the study of virus-host interactions.


Subject(s)
MicroRNAs/genetics , Porcine respiratory and reproductive syndrome virus/physiology , Tripartite Motif Proteins/genetics , Virus Replication , Animals , Cell Line , Gene Expression Regulation , Host-Pathogen Interactions , Humans , Lysosomes/metabolism , MicroRNAs/antagonists & inhibitors , Microtubule-Associated Proteins/metabolism , Nucleocapsid Proteins/metabolism , Porcine respiratory and reproductive syndrome virus/metabolism , Tripartite Motif Proteins/metabolism
2.
J Transl Med ; 21(1): 443, 2023 07 06.
Article in English | MEDLINE | ID: mdl-37415153

ABSTRACT

BACKGROUND: Aberrant ubiquitin-proteasome system (UPS) triggers various disorders of biological events and contributes to progression of tumorigenesis. The tripartite motif containing 22 (TRIM22) was demonstrated to participate in the progression of multiple malignancies. Nevertheless, the role of TRIM22 in melanoma is still indefinite. This project aims to investigate the biological function of TRIM22 in melanoma and provide novel therapeutical targets. METHODS: Bioinformatic algorithms were used to investigate prognostic significance of TRIM22. The in vitro or in vivo assays were used to explore the functions of TRIM22 in melanoma. The Co-Immunoprecipitation (Co-IP) and in vivo ubiquitination assays were used to assess regulations of TRIM22 on lysine acetyltransferase 2 A (KAT2A). The Chromatin immunoprecipitation (ChIP) assays and luciferase reporter assay were utilized to explore epigenetic regulations of KAT2A on Notch1. RESULTS: Here, we utilized the bioinformatic methods to confirm that TRIM22 is decreased in melanoma than normal tissues. Patients with low TRIM22 levels had shorter survival months than those with high TRIM22 levels. Targeting TRIM22 favors melanoma cell migration, proliferation, and tumor development in vitro and in vivo. Mechanistically, TRIM22 interacts with KAT2A and promotes its degradation in a ubiquitination-dependent manner. Melanoma cells with TRIM22 deficiency depended on KAT2A to enhance malignant progression, including proliferation, migration, and in vivo growth. KEGG analysis determined the positive correlation between KAT2A and Notch signaling. Chromatin Immunoprecipitation (ChIP) assays implicated that KAT2A directly binds to the promoter region of Notch1 and mediates the enrichment of H3K9ac modification. KAT2A activates Notch1 transcriptional levels and sustains the stemness feature of melanoma cells. Nocth1 inhibitor (IMR-1) effectively suppresses the growth of TRIM22low melanoma in vitro and in vivo but fails to inhibit TRIM22high melanoma. CONCLUSION: Together, our study illustrates the mechanism by which the TRIM22-KAT2A-Notch1 axis promotes melanoma progression, and demonstrates that KAT2A/Nocth1 confers an epigenetic vulnerability in TRIM22low melanoma.


Subject(s)
Melanoma , Humans , Cell Line, Tumor , Melanoma/genetics , Signal Transduction , Ubiquitination , Epigenesis, Genetic , Tripartite Motif Proteins/genetics , Tripartite Motif Proteins/metabolism , Repressor Proteins/genetics , Minor Histocompatibility Antigens/metabolism , Histone Acetyltransferases/genetics , Histone Acetyltransferases/metabolism
3.
J Biochem Mol Toxicol ; 37(7): e23367, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37009741

ABSTRACT

2,2-Bis(4-hydroxyphenyl)propane (bisphenol A; BPA) is an environmental endocrine-disrupting chemical. It mimics the effects of estrogen at multiple levels by activating estrogen receptors (ERs); however, BPA also affects the proliferation of human breast cancer cells independent of ERs. Although BPA inhibits progesterone (P4) signaling, the toxicological significance of its effects remain unknown. Tripartite motif-containing 22 (TRIM22) has been identified as a P4-responsive and apoptosis-related gene. Nevertheless, it has not yet been established whether exogenous chemicals change TRIM22 gene levels. Therefore, the present study investigated the effects of BPA on P4 signaling and TRIM22 and TP53 expression in human breast carcinoma MCF-7 cells. In MCF-7 cells incubated with various concentrations of P4, TRIM22 messenger RNA (mRNA) levels increased in a dose-dependent manner. P4 induced apoptosis and decreased viability in MCF-7 cells. The knockdown of TRIM22 abolished P4-induced decreases in cell viability and P4-induced apoptosis. P4 increased TP53 mRNA expression and p53 knockdown decrease the basal level of TRIM22 and P4 increased TRIM22 mRNA expression independent of p53 expression. BPA attenuated P4-induced increases in the ratio of cell apoptosis in a concentration-dependent manner, and the P4-induced decreases in cell viability was abolished in the presence of 100 nM and higher BPA concentrations. Furthermore, BPA inhibited P4-induced TRIM22 and TP53 expression. In conclusion, BPA inhibited P4-induced apoptosis in MCF-7 cells via the inhibition of P4 receptor transactivation. TRIM22 gene has potential as a biomarker for investigating the disruption of P4 signaling by chemicals.


Subject(s)
Breast Neoplasms , Progesterone , Humans , Female , Progesterone/pharmacology , MCF-7 Cells , Receptors, Progesterone/genetics , Receptors, Progesterone/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Transcriptional Activation , Breast Neoplasms/pathology , Benzhydryl Compounds/pharmacology , Apoptosis
4.
Virol J ; 19(1): 228, 2022 12 31.
Article in English | MEDLINE | ID: mdl-36587218

ABSTRACT

BACKGROUND: Dengue virus type 2 (DENV-2) was used to infect primary human umbilical vein endothelial cells (HUVECs) to examine autophagy induced by activation of the adenosine monophosphate-activated protein kinase (AMPK)/extracellular signal-regulated kinase (ERK)/mammalian target of rapamycin (mTOR) signaling pathway following tripartite motif-containing 22 (TRIM22)-mediated DENV-2 infection to further reveal the underlying pathogenic mechanism of DENV-2 infection. METHODS: Quantitative real-time polymerase chain reaction (qRT-PCR) was used to screen putative interference targets of TRIM22 and determine the knockdown efficiency. The effect of TRIM22 knockdown on HUVEC proliferation was determined using the CCK8 assay. Following TRIM22 knockdown, transmission electron microscopy (TEM) was used to determine the ultrastructure of HUVEC autophagosomes and expression of HUVEC autophagy and AMPK pathway-related genes were measured by qRT-PCR. Moreover, HUVEC autophagy and AMPK pathway-related protein expression levels were determined by western blot analysis. Cell cycle and apoptosis were assessed by flow cytometry (FCM) and the autophagosome structure of the HUVECs was observed by TEM. RESULTS: Western blot results indicated that TRIM22 protein expression levels increased significantly 36 h after DENV-2 infection, which was consistent with the proteomics prediction. The CCK8 assay revealed that HUVEC proliferation was reduced following TRIM22 knockdown (P < 0.001). The TEM results indicated that HUVEC autolysosomes increased and autophagy was inhibited after TRIM22 knockdown. The qRT-PCR results revealed that after TRIM22 knockdown, the expression levels of antithymocyte globulin 7 (ATG7), antithymocyte globulin 5 (ATG5), Beclin1, ERK, and mTOR genes decreased (P < 0.01); however, the expression of AMPK genes (P < 0.05) and P62 genes (P < 0.001) increased. FCM revealed that following TRIM22 knockdown, the percentage of HUVECs in the G2 phase increased (P < 0.001) along with cell apoptosis. The effect of TRIM22 overexpression on HUVEC autophagy induced by DENV-2 infection and AMPK pathways decreased after adding an autophagy inhibitor. CONCLUSIONS: In HUVECs, TRIM22 protein positively regulates autophagy and may affect autophagy through the AMPK/ERK/mTOR signaling pathway. Autophagy is induced by activation of the AMPK/ERK/mTOR signaling pathway following TRIM22-mediated DENV-2 infection of HUVECs.


Subject(s)
AMP-Activated Protein Kinases , Extracellular Signal-Regulated MAP Kinases , Humans , AMP-Activated Protein Kinases/genetics , AMP-Activated Protein Kinases/metabolism , Human Umbilical Vein Endothelial Cells , Extracellular Signal-Regulated MAP Kinases/metabolism , Sirolimus/pharmacology , Antilymphocyte Serum/pharmacology , Signal Transduction , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , TOR Serine-Threonine Kinases/pharmacology , Autophagy , Tripartite Motif Proteins/genetics , Tripartite Motif Proteins/pharmacology , Repressor Proteins/metabolism , Minor Histocompatibility Antigens/pharmacology
5.
Cutan Ocul Toxicol ; 41(4): 304-309, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36170453

ABSTRACT

OBJECTIVE: To reveal the function and underlying mechanism of Tri-domain protein 22 (TRIM22) in psoriasis. METHODS: M5 cytokines were applied in HaCat cells to mimic psoriasis in vitro. The TRIM22-silencing viruses were established to knockdown TRIM22 in HaCat cells. Western blot and/or real-time PCR were used to detect the expression of TRIM22, KRT1, KRT6, p-P65, P65, LC3, Beclin 1, P62, p-PI3K, PI3K, p-Akt, Akt, p-mTOR, and mTOR. ELISA kits were applied to assess levels of TNF-α, IL-1ß, IL-18, and HMGB1. RESULTS: TRIM22 expression levels were upregulated in M5-treated HaCat cells. M5 treatment enhanced cell proliferation and inflammation, and inhibited autophagy in HaCat cells which were effectively reversed by TRIM22 deficiency. Activation of PI3K/Akt/mTOR pathway is an essential promoter of cell proliferation and inflammation, and inhibitor of autophagy in psoriasis. TRIM22 deficiency blocked M5-induced activation of PI3K/Akt/mTOR pathway in HaCat cells. CONCLUSIONS: TRIM22 facilitates cell proliferation and inflammation, and suppresses autophagy in M5-treated HaCat cells through activating PI3K/Akt/mTOR pathway, and inhibition of TRIM22 can be a novel potential treatment for psoriasis.


Subject(s)
Phosphatidylinositol 3-Kinases , Psoriasis , Humans , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , TOR Serine-Threonine Kinases/pharmacology , Autophagy , Cell Proliferation , Psoriasis/drug therapy , Psoriasis/metabolism , Inflammation/metabolism , Apoptosis , Tripartite Motif Proteins/genetics , Repressor Proteins/metabolism , Repressor Proteins/pharmacology , Minor Histocompatibility Antigens/pharmacology
6.
J Med Virol ; 93(6): 3412-3419, 2021 06.
Article in English | MEDLINE | ID: mdl-32803897

ABSTRACT

Respiratory syncytial virus (RSV) infection is a major cause of lower respiratory tract disease. Although RSV causes major economic losses every year, effective treatments have not been found so far. Recent studies have shown that the tripartite motif-containing (TRIM) superfamily plays an essential role in the immune response. In this study, we found that TRIM22 had an inhibitory effect on RSV infection, and downregulation of TRIM22 moderately enhanced RSV replication. Our data further demonstrated that RSV infection induced TRIM22 expression through the activation of JAK-STAT1/2 signaling. RSV infection also induced TRIM22 expression. Taken together, these data points showed that the TRIM family member, TRIM22, had an essential role in resisting RSV infection, and this effect was closely related to the JAK-STAT1/2 pathway. Our results provide promising evidence for a novel target for the prevention and treatment of RSV.


Subject(s)
Janus Kinases/genetics , Minor Histocompatibility Antigens/genetics , Repressor Proteins/genetics , Respiratory Syncytial Virus, Human/physiology , STAT1 Transcription Factor/genetics , Signal Transduction , Tripartite Motif Proteins/genetics , Virus Replication/genetics , A549 Cells , Cell Line , Gene Knockdown Techniques , Humans , Janus Kinases/metabolism , Minor Histocompatibility Antigens/immunology , Repressor Proteins/immunology , Respiratory Syncytial Virus, Human/immunology , STAT1 Transcription Factor/metabolism , Tripartite Motif Proteins/immunology , Virus Replication/immunology
7.
Virus Genes ; 55(5): 660-672, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31375995

ABSTRACT

Porcine reproductive and respiratory syndrome virus (PRRSV) infection causes one of the most economically important swine diseases worldwide. Tripartite motif-containing 22 (TRIM22), a TRIM family protein, has been identified as a crucial restriction factor that inhibits a group of human viruses. Currently, the role of cellular TRIM22 in PRRSV infection remains unclear. In the present study, we analyzed the effect of TRIM22 on PRRSV replication in vitro and explored the underlying mechanism. Ectopic expression of TRIM22 impaired the viral replication, while TRIM22-RNAi favored the replication of PRRSV in MARC-145 cells. Additionally, we observed that TRIM22 deletion SPRY domain or Nuclear localization signal (NLS) losses the ability to inhibit PRRSV replication. Finally, Co-IP analysis identified that TRIM22 interacts with PRRSV nucleocapsid (N) protein through the SPRY domain, while the NLS2 motif of N protein is involved in interaction with TRIM22. Although the concentration of PRRSV N protein was not altered in the presence of TRIM22, the abundance of N proteins from simian hemorrhagic fever virus (SHFV), equine arteritis virus (EAV), and murine lactate dehydrogenase-elevating virus (LDV) diminished considerably with increasing TRIM22 expression. Together, our findings uncover a previously unrecognized role for TRIM22 and extend the antiviral effects of TRIM22 to arteriviruses.


Subject(s)
Host-Pathogen Interactions , Nuclear Localization Signals , Porcine respiratory and reproductive syndrome virus/growth & development , Porcine respiratory and reproductive syndrome virus/immunology , Tripartite Motif Proteins/genetics , Tripartite Motif Proteins/metabolism , Virus Replication , Animals , Cell Line , Chlorocebus aethiops , Gene Silencing , Nucleocapsid Proteins/metabolism , Protein Interaction Mapping , Sequence Deletion
8.
J Cell Biochem ; 119(11): 8971-8980, 2018 11.
Article in English | MEDLINE | ID: mdl-30011088

ABSTRACT

Autophagy is a crucial host-defense mechanism against Mycobacterium tuberculosis (Mtb) infection by spanning innate and adaptive immune functions. TRIM22 is a member of tripartite motif family protein which involved in innate immunity and autophagy process. However, its role in the modulation of bacterial infection has not been investigated. Here, we demonstrated that TRIM22 is upregulated in a dose-dependent and time-dependent manner during Mtb infection of THP-1 cells. Downregulation of TRIM22 significantly decreased light chain 3 (LC3)-II protein level and the formation of LC3 puncta, while it markedly increased SQSTM1, a marker of autophagic degradation, in Mtb-infected THP-1 cells. What is more, enhanced bacterial survival was observed in TRIM22 knockdown THP-1 cells, while rapamycin abrogated this effect. In the presence of vector containing TRIM22 in THP-1 cells prior to infection, the survival of Mtb was decreased, while BafA restored this effect. Further study demonstrated that TRIM22 expression was regulated by MicroRNA-20b, and that TRIM22 regulates Mtb-infected THP-1 autophagy via the nuclear factor-κB/beclin 1 pathway. Using a nuclear factor-κB inhibitor BAY 11-7082, we found that TRIM22-induced high expression of LC3-II and the formation of LC3 was substantially attenuated, while the TRIM22-induced low expression of SQSTM1 was markedly increased in BAY 11-7082-treatment cells. In addition, the bacterial survival reduced by TRIM22 was significantly reversed by BAY 11-7082. Overall, these results suggest that TRIM22-augmented autophagy prevents intracellular Mtb to evade autophagic clearance, thereby inhibiting the persistence of Mtb infections.


Subject(s)
Autophagy/physiology , Beclin-1/metabolism , Macrophages/physiology , Minor Histocompatibility Antigens/metabolism , Mycobacterium tuberculosis/pathogenicity , NF-kappa B/metabolism , Repressor Proteins/metabolism , Tripartite Motif Proteins/metabolism , Autophagy/genetics , Beclin-1/genetics , Blotting, Western , Humans , Immunity, Innate/genetics , Immunity, Innate/physiology , Macrophages/metabolism , Minor Histocompatibility Antigens/genetics , NF-kappa B/genetics , Real-Time Polymerase Chain Reaction , Repressor Proteins/genetics , Sequestosome-1 Protein/genetics , Sequestosome-1 Protein/metabolism , THP-1 Cells , Tripartite Motif Proteins/genetics
9.
Cell Biol Int ; 42(9): 1192-1199, 2018 Sep.
Article in English | MEDLINE | ID: mdl-29762880

ABSTRACT

Tripartite motif-containing 22 (TRIM22) is reported to participate in numerous cellular activities. Recent studies confirm that TRIM22 is a target gene for P53, and inhibits clonogenic growth of leukemic U-937 cells. The current study aims to discover the effect of TRIM22 in progression of human chronic myeloid leukemia (CML) and explore the related mechanism. TRIM22 was knocked down by siRNA transfection in CML cell K562. We observed that TRIM22 knockdown decreased proliferation and invasion in K562 cells. TRIM22 knockdown significantly induced cell cycle arrest by regulating the level of CDK4, Cyclin D1, P70S6K, and P53 in K562 cell. Moreover, loss of TRIM22 also promoted apoptosis through modulation of Bcl-2, Bax and active Caspase 3 in K562 cell. Furthermore, we demonstrated that TRIM22 knockdown inhibited the activation of PI3K/Akt/mTOR pathway by decreasing the level of the phosphorylated form p-Akt and p-mTOR in K562 cell. In conclusion, loss of TRIM22 suppresses the progression and invasion of CML through regulation of PI3K/Akt/mTOR pathway, suggesting that TRIM22 might be as a potential target for the treatment strategy of CML.


Subject(s)
Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Minor Histocompatibility Antigens/metabolism , Phosphoinositide-3 Kinase Inhibitors , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Repressor Proteins/deficiency , Repressor Proteins/metabolism , TOR Serine-Threonine Kinases/antagonists & inhibitors , Tripartite Motif Proteins/deficiency , Tripartite Motif Proteins/metabolism , Apoptosis/drug effects , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Gene Knockdown Techniques , Humans , K562 Cells , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Minor Histocompatibility Antigens/genetics , Neoplasm Invasiveness , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Repressor Proteins/genetics , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Tripartite Motif Proteins/genetics
10.
J Infect Dis ; 213(6): 1008-12, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26525407

ABSTRACT

Long-term treatment with interferon (IFN) alfa plus ribavirin decreases the proviral human immunodeficiency virus type 1 (HIV) DNA level. However, the short-term impact of IFN alfa on persistent HIV and its effects on immune activation after antiretroviral therapy remain unknown. Our study showed that the cell-associated HIV RNA level and CD4(+) T-cell activation decreased in the IFN group (n = 10). No changes were detected in levels of residual plasma viremia, replication-competent reservoirs, proviral DNA, or 2-long-terminal repeat circles, although APOBEC3G, TRIM5α, BST2, and TRIM22 were upregulated in the IFN group. These data suggest that short-term treatment with IFN alfa combined with RBV decreases HIV expression, in part through inhibition of HIV transcription by TRIM22 and decrease in T-cell activation.


Subject(s)
Antiviral Agents/therapeutic use , CD4-Positive T-Lymphocytes/drug effects , HIV Infections/drug therapy , HIV-1/physiology , Hepatitis C/complications , Interferon-alpha/therapeutic use , Polyethylene Glycols/therapeutic use , Adult , Anti-HIV Agents/administration & dosage , Anti-HIV Agents/therapeutic use , Antiviral Agents/administration & dosage , Biomarkers , CD4-Positive T-Lymphocytes/classification , CD4-Positive T-Lymphocytes/physiology , Coinfection , Female , Gene Expression Regulation, Viral/drug effects , HIV Infections/complications , Hepacivirus , Humans , Interferon-alpha/administration & dosage , Lymphocyte Activation/drug effects , Male , Middle Aged , Polyethylene Glycols/administration & dosage , Recombinant Proteins/administration & dosage , Recombinant Proteins/therapeutic use , Ribavirin/administration & dosage , Ribavirin/therapeutic use
11.
J Transl Med ; 14: 257, 2016 09 02.
Article in English | MEDLINE | ID: mdl-27590274

ABSTRACT

BACKGROUND AND AIMS: TRIM5 and TRIM22 are restriction factors involved in innate immune response and exhibit anti-viral activity. Single nucleotide polymorphisms (SNPs) at TRIM5 and TRIM22 genes have shown to influence several viral infections such as human immunodeficiency virus (HIV), hepatitis B, as well as measles and rubella vaccination. The aim of this study is to analyze whether TRIM5 and TRIM22 polymorphisms are associated with liver fibrosis inflammation-related biomarkers and response to pegylated-interferon-alpha plus ribavirin (pegIFNα/RBV) therapy in HIV/hepatitis C virus (HCV) coinfected patients. METHODS: A retrospective study was performed in 319 patients who started pegIFNα/RBV therapy. Liver fibrosis stage was characterized in 288 patients. TRIM5 rs3824949 and TRIM22 polymorphisms (rs1063303, rs7935564, and rs7113258) were genotyped using the GoldenGate assay. The primary outcomes were: a) significant liver fibrosis (≥F2) evaluated by liver biopsy or transient elastography (liver stiffness values ≥7.1 Kpa); b) sustained virological response (SVR) defined as no detectable HCV viral load (<10 IU/mL) at week 24 after the end of the treatment. The secondary outcome variable was plasma chemokine levels. RESULTS: Patients with TRIM5 rs3824949 GG genotype had higher SVR rate than patients with TRIM5 rs3824949 CC/CG genotypes (p = 0.013), and they had increased odds of achieving SVR (adjusted odds ratio (aOR = 2.58; p = 0.012). Patients with TRIM22 rs1063303 GG genotype had higher proportion of significant liver fibrosis than patients with rs1063303 CC/CG genotypes (p = 0.021), and they had increased odds of having significant hepatic fibrosis (aOR = 2.19; p = 0.034). Patients with TRIM22 rs7113258 AT/AA genotype had higher SVR rate than patients with rs7113258 TT genotypes (p = 0.013), and they had increased odds of achieving SVR (aOR = 1.88; p = 0.041). The TRIM22 haplotype conformed by rs1063303_C and rs7113258_A was more frequent in patients with SVR (p = 0.018) and was significantly associated with achieving SVR (aOR = 2.80; p = 0.013). The TRIM5 rs3824949 GG genotype was significantly associated with higher levels of GRO-α (adjusted arithmetic mean ratio ((aAMR) = 1.40; p = 0.011) and MCP-1 (aAMR = 1.61; p = 0.003). CONCLUSIONS: TRIM5 and TRIM22 SNPs are associated to increased odds of significant liver fibrosis and SVR after pegIFNα/RBV therapy in HIV/HCV coinfected patients. Besides, TRIM5 SNP was associated to higher baseline levels of circulating biomarkers GRO and MCP-1.


Subject(s)
Antiviral Agents/therapeutic use , Carrier Proteins/genetics , Coinfection/genetics , HIV Infections/genetics , Hepacivirus/physiology , Hepatitis C, Chronic/genetics , Minor Histocompatibility Antigens/genetics , Polymorphism, Single Nucleotide/genetics , Repressor Proteins/genetics , Tripartite Motif Proteins/genetics , Adult , Antiviral Restriction Factors , Chemokines/blood , Coinfection/drug therapy , Female , Gene Frequency/genetics , Genetic Association Studies , Genetic Predisposition to Disease , HIV Infections/blood , HIV Infections/complications , Haplotypes/genetics , Hepatitis C, Chronic/blood , Hepatitis C, Chronic/drug therapy , Humans , Male , Middle Aged , Ubiquitin-Protein Ligases
12.
J Transl Med ; 14: 94, 2016 Apr 18.
Article in English | MEDLINE | ID: mdl-27089879

ABSTRACT

BACKGROUND: We previously demonstrated that the HLA class II transactivator CIITA inhibits HIV-1 replication in T cells by competing with the viral transactivator Tat for the binding to Cyclin T1 subunit of the P-TEFb complex. Here, we analyzed the anti-viral function of CIITA in myeloid cells, another relevant HIV-1 target cell type. We sinvestigated clones of the U937 promonocytic cell line, either permissive (Plus) or non-permissive (Minus) to HIV-1 replication. This different phenotype has been associated with the expression of TRIM22 in U937 Minus but not in Plus cells. METHODS: U937 Plus cells stably expressing CIITA were generated and HLA-II positive clones were selected by cell sorting and cloning. HLA and CIITA proteins were analyzed by cytofluorometry and western blotting, respectively. HLA-II DR and CIITA mRNAs were quantified by qRT-PCR. Tat-dependent transactivation was assessed by performing the HIV-1 LTR luciferase gene reporter assay. Cells were infected with HIV-1 and viral replication was evaluated by measuring the RT activity in culture supernatants. RESULTS: CIITA was expressed only in HLA-II-positive U937 Minus cells, and this was strictly correlated with inhibition of Tat-dependent HIV-1 LTR transactivation in Minus but not in Plus cells. Overexpression of CIITA in Plus cells restored the suppression of Tat transactivation, confirming the inhibitory role of CIITA. Importantly, HIV-1 replication was significantly reduced in Plus-CIITA cells with respect to Plus parental cells. This effect was independent of TRIM22 as CIITA did not induce TRIM22 expression in Plus-CIITA cells. CONCLUSIONS: U937 Plus and Minus cells represent an interesting model to study the role of CIITA in HIV-1 restriction in the monocytic/macrophage cell lineage. The differential expression of CIITA in CIITA-negative Plus and CIITA-positive Minus cells correlated with their capacity to support or not HIV-1 replication, respectively. In Minus cells CIITA targeted the viral transactivator Tat to inhibit HIV-1 replication. The generation of Plus-CIITA cells was instrumental to demonstrate the specific contribution of CIITA in terms of inhibition of Tat activity and HIV-1 restriction, independently from other cellular factors, including TRIM22. Thus, CIITA acts as a general restriction factor against HIV-1 not only in T cells but also in myeloid cells.


Subject(s)
HIV-1/physiology , Myeloid Cells/metabolism , Myeloid Cells/virology , Nuclear Proteins/metabolism , Trans-Activators/metabolism , Virus Replication/physiology , tat Gene Products, Human Immunodeficiency Virus/metabolism , Clone Cells , HEK293 Cells , Humans , Phenotype , Terminal Repeat Sequences/genetics , Transcriptional Activation
13.
J Recept Signal Transduct Res ; 35(4): 289-94, 2015.
Article in English | MEDLINE | ID: mdl-25510414

ABSTRACT

Tripartite motif 22 (TRIM22) is involved in various cellular processes. It has been reported that TRIM22 can activate nuclear factor-κB (NF-κB) pathway, but the precise mechanism remains unclear. In this study, we explored the exact role of TRIM22 in activating the NF-κB pathway. Different to tumor necrosis factor-α (TNF-α) induction, we found that the overexpression of TRIM22 could induce the processing of p100 to p52 in HEK293T cells. Furthermore, based on the results of co-immunoprecipitation and co-localization experiments, we demonstrated that TRIM22 could interact with IκB kinase (IKK)α but not IKKß and could increase the level and phosphorylation of IKKα through its really interesting new gene (RING) and spla-ryanodine receptor (SPRY) domains. These results suggest that TRIM22 is able to activate the noncanonical but not the canonical NF-κB pathway by activating IKKα. This finding will aid our understanding of the biological function of TRIM22.


Subject(s)
I-kappa B Kinase/metabolism , NF-kappa B/metabolism , Repressor Proteins/metabolism , HEK293 Cells , Humans , I-kappa B Kinase/genetics , Minor Histocompatibility Antigens , Models, Biological , Mutant Proteins/chemistry , Mutant Proteins/genetics , Mutant Proteins/metabolism , NF-kappa B/genetics , Protein Binding , Protein Interaction Domains and Motifs , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Repressor Proteins/chemistry , Repressor Proteins/genetics , Signal Transduction , Tripartite Motif Proteins
14.
Hum Mutat ; 35(9): 1072-81, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24863734

ABSTRACT

Tripartite motif protein 22 (TRIM22) is a novel interferon-induced protein that potently inhibits the replication of evolutionarily diverse viruses, including HIV-1. Altered TRIM22 expression is also associated with diseases, such as multiple sclerosis, cancer, and autoimmunity. The factors that influence TRIM22 expression and antiviral activity are largely unknown. In this study, we adopted an evolution-guided functional approach to identify potential genetic determinants of TRIM22 function. Evolutionary analysis of TRIM22 from mammals spanning >100 million years demonstrated that TRIM22 evolution has been shaped by ancient and variable positive selection. We showed that positive selection is operating on multiple TRIM22 residues that cluster in putative functional regions and that some are predicted to be functionally damaging. Interestingly, the second most prevalent TRIM22 SNP in humans (rs1063303) is located at one of these positively selected sites. We showed that the frequency of rs1063303:G>C varies up to 10-fold between ethnicities and that in some ethnicities SNP rs1063303:G>C is being actively maintained in the population. The SNP rs1063303:G>C variant also had an inverse functional impact where it increased TRIM22 expression and decreased the antiviral activity of TRIM22. Taken together, our data characterize the extensive genetic variation in TRIM22 and identify rs1063303:G>C as a highly prevalent SNP that influences its function.


Subject(s)
Adaptation, Biological , Disease Resistance/genetics , Evolution, Molecular , Polymorphism, Single Nucleotide , Repressor Proteins/genetics , Repressor Proteins/metabolism , Virus Diseases/genetics , Animals , Cell Line , Humans , Mammals , Minor Histocompatibility Antigens , Models, Molecular , Phylogeny , Protein Conformation , Protein Interaction Domains and Motifs/genetics , Repressor Proteins/chemistry , Selection, Genetic , Tripartite Motif Proteins
15.
Eur J Immunol ; 43(8): 2174-84, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23670564

ABSTRACT

Tripartite motif (TRIM) 22 plays an important role in IFN-mediated antiviral activity. We previously demonstrated that IFN regulatory factor-1 (IRF-1) was crucial for constitutive and IFN-induced TRIM22 expression via binding to a special cis-element named 5' extended IFN-stimulating response element. Here, we further investigate the molecular mechanisms of TRIM22 with a focus on the co-activators of IRF-1. Using an in vitro DNA affinity binding assay and an in vivo chromatin immunoprecipitation assay, we found that IFN-γ stimulation significantly enhanced the binding of p300 and p300/CBP-associated factor, but not other co-activators such as general control nondepressible 5, steroid receptor co-activator-1, and activator of thyroid and retinoic, to the 5' extended IFN-stimulating response element containing TRIM22 promoter region together with IRF-1. Overexpression and knockdown analysis demonstrated that it was p300, but not p300/CBP-associated factor, that functioned as a transcriptional co-activator of IRF-1 in IFN-γ induction of TRIM22. We further show that p300 contributed to both IFN-γ- and IRF-1-mediated TRIM22 transcription independent of its histone acetyltransferase activity, however, it was required for the recruitment of RNA polymerase II to TRIM22 promoter region. These data indicate that p300 plays a critical role in IFN-γ-induced TRIM22 expression via recruiting RNA polymerase II to the TRIM22 promoter, and might serve as a bridge between IRF-1 and the basal transcriptional apparatus in TRIM22 induction.


Subject(s)
Histone Acetyltransferases/metabolism , Interferon Regulatory Factor-1/metabolism , Repressor Proteins/biosynthesis , p300-CBP Transcription Factors/metabolism , Cell Line, Tumor , Chromatin Immunoprecipitation , Hep G2 Cells , Humans , Interferon-gamma/metabolism , Minor Histocompatibility Antigens , Nuclear Receptor Coactivator 1/metabolism , Nuclear Receptor Coactivator 3/metabolism , Promoter Regions, Genetic , RNA Interference , RNA Polymerase II/metabolism , RNA, Small Interfering , Tripartite Motif Proteins , p300-CBP Transcription Factors/genetics
16.
Brain Behav ; 14(3): e3465, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38468469

ABSTRACT

BACKGROUND: SP gene family, consisting of SP100, SP110, SP140, and SP140L, has been implicated in the initiation and advancement of numerous malignancies. Nevertheless, their clinical significance in glioma remains incompletely understood. METHOD: Expression levels and prognostic significance of SP family members were evaluated in the TCGA and CGGA datasets. Multifactorial analysis was used to identify SP gene family members that can independently impact the prognosis of glioma patients. A SP140-based predictive risk model/nomogram was developed in TCGA dataset and validated in CGGA dataset. The model's performance was evaluated through receiver operating characteristic (ROC) curves, calibration plots, and decision curve analyses. Phenotypic associations of SP140 and TRIM22 were examined through CancerSEA and TIMER. The effect of SP140 inhibitor in glioma progress and TRIM22/PI3K/AKT signaling pathway was confirmed in U251/U87 glioma cells. RESULTS: The SP family members exhibited elevated expression in gliomas and were negatively correlated with prognosis. SP140 emerged as an independent prognostic factor, and a SP140-based nomogram/predictive risk model demonstrated high accuracy. SP140 inhibitor, GSK761, lead to the suppression of TRIM22 expression and the PI3K/AKT signaling pathway. GSK761 also restrain glioma proliferation, migration, and invasion. Furthermore, SP140 and TRIM22 coexpressed in glioma cells with high level of vascular proliferation, TRIM22 is closely associated with the immune cell infiltration. CONCLUSION: SP140-based nomogram proved to be a practical tool for predicting the survival of glioma patients. SP140 inhibitor could suppress glioma progress via TRIM22/PI3K/AKT signaling pathway.


Subject(s)
Glioma , Proto-Oncogene Proteins c-akt , Humans , Proto-Oncogene Proteins c-akt/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Cell Proliferation , Signal Transduction , Glioma/drug therapy , Glioma/genetics , Tripartite Motif Proteins/genetics , Tripartite Motif Proteins/pharmacology , Repressor Proteins/metabolism , Minor Histocompatibility Antigens/pharmacology , Transcription Factors , Antigens, Nuclear/metabolism , Antigens, Nuclear/pharmacology
17.
Redox Rep ; 29(1): 2404794, 2024 Dec.
Article in English | MEDLINE | ID: mdl-39314036

ABSTRACT

OBJECTIVES: Diabetic nephropathy (DN) is one of the most serious microvascular complications of diabetes and is the most common cause of end-stage renal disease. Tripartite motif-containing (TRIM) proteins are a large family of E3 ubiquitin ligases that contribute to protein quality control by regulating the ubiquitin - proteasome system. However, the detailed mechanisms through which various TRIM proteins regulate downstream events have not yet been fully elucidated. The current research aimed to determine the function and mechanism of TRIM22 in DN. METHODS: DN models were established by inducing HK-2 cells using high glucose (HG) and diabetic mice (db/db mice). Cell viability, apoptosis, mitochondrial reactive oxygen species, and mitochondrial membrane potential were detected by Cell Counting Kit-8 and flow cytometry, respectively. Pathological changes were evaluated using hematoxylin and eosin, periodic acid schiff and Masson staining. The binding between TRIM22 and optic atrophy 1 (OPA1) was analyzed using co-immunoprecipitation. The m6A level of TRIM22 5'UTR was detected using RNA immunoprecipitation. RESULTS: TRIM22 was highly expressed in patients with DN. TRIM22 silencing inhibited HG-induced apoptosis and mitochondrial dysfunction in HK-2 cells. Promoting mitochondrial fusion alleviated TRIM22 overexpression-induced cell apoptosis, mitochondrial dysfunction in HK-2 cells, and kidney damage in mice. Mechanistically, TRIM22 interacted with OPA1 and induced its ubiquitination. Wilms tumor 1-associating protein (WTAP) promoted m6A modification of TRIM22 through the m6A reader insulin-like growth factor 2 mRNA-binding protein 1 (IGF2BP1). DISCUSSION: TRIM22 silencing inhibited the progression of DN by interacting with OPA1 and inducing its ubiquitination. Furthermore, WTAP promoted m6A modification of TRIM22 via IGF2BP1.


Subject(s)
Diabetic Nephropathies , GTP Phosphohydrolases , Minor Histocompatibility Antigens , Mitochondria , Tripartite Motif Proteins , Ubiquitination , Diabetic Nephropathies/metabolism , Diabetic Nephropathies/pathology , Tripartite Motif Proteins/metabolism , Tripartite Motif Proteins/genetics , Animals , Humans , Mice , GTP Phosphohydrolases/metabolism , GTP Phosphohydrolases/genetics , Mitochondria/metabolism , Minor Histocompatibility Antigens/metabolism , Minor Histocompatibility Antigens/genetics , Male , Repressor Proteins/metabolism , Repressor Proteins/genetics , Apoptosis , Diabetes Mellitus, Experimental/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/genetics
18.
mBio ; 15(2): e0232023, 2024 Feb 14.
Article in English | MEDLINE | ID: mdl-38275298

ABSTRACT

Replication of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) genome is mediated by a complex of non-structural proteins (NSPs), of which NSP7 and NSP8 serve as subunits and play a key role in promoting the activity of RNA-dependent RNA polymerase (RdRp) of NSP12. However, the stability of subunits of the RdRp complex has rarely been reported. Here, we found that NSP8 was degraded by the proteasome in host cells, and identified tripartite motif containing 22 (TRIM22) as its E3 ligase. The interferon (IFN) signaling pathway was activated upon viral invasion into host cells, and TRIM22 expression increased. TRIM22 interacted with NSP8 and ubiquitinated it at Lys97 via K48-type ubiquitination. TRIM22 overexpression significantly reduced viral RNA and protein levels. Knockdown of TRIM22 enhanced viral replication. This study provides a new explanation for treating patients suffering from SARS-CoV-2 with IFNs and new possibilities for drug development targeting the interaction between NSP8 and TRIM22.IMPORTANCENon-structural proteins (NSPs) play a crucial role in the replication of severe acute respiratory syndrome coronavirus 2, facilitating virus amplification and propagation. In this study, we conducted a comprehensive investigation into the stability of all subunits comprising the RNA-dependent RNA polymerase complex. Notably, our results reveal for the first time that NSP8 is a relatively unstable protein, which is found to be readily recognized and degraded by the proteasome. This degradation process is mediated by the host E3 ligase tripartite motif containing 22 (TRIM22), which is also a member of the interferon stimulated gene (ISG) family. Our study elucidates a novel mechanism of antiviral effect of TRIM22, which utilizes its own E3 ubiquitin ligase activity to hinder viral replication by inducing ubiquitination and subsequent degradation of NSP8. These findings provide new ideas for the development of novel therapeutic strategies. In addition, the conserved property of NSP8 raises the possibility of developing broad antiviral drugs targeting the TRIM22-NSP8 interaction.


Subject(s)
COVID-19 , Ubiquitin-Protein Ligases , Humans , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , SARS-CoV-2/metabolism , Proteasome Endopeptidase Complex , RNA-Dependent RNA Polymerase/metabolism , Interferons , Virus Replication , Tripartite Motif Proteins/genetics , Repressor Proteins/genetics , Minor Histocompatibility Antigens
19.
J Cancer ; 15(8): 2137-2146, 2024.
Article in English | MEDLINE | ID: mdl-38495497

ABSTRACT

Background. Tripartite motif-containing 22 (TRIM22) is characterized by a canonical RING domain with ubiquitin E3 ligase activity and is closely associated with tumorigenesis. As a product of TRIM22 transcription, whether hsa_circ_TRIM22 has a function of regulating tumorigenesis is unclear. Thus, we aimed to explore the role and mechanism of hsa_circ_TRIM22 in human papillomavirus (HPV) 16 positive cervical cancer (CC). Methods. We collected HPV16-positive cervical tissues including chronic cervicitis, high-grade squamous intraepithelial lesions (HSIL), low-grade squamous intraepithelial lesions (LSIL), and CC, and along with CC cell lines to detect the hsa_circ_TRIM22 level using real-time fluorescence quantitative polymerase chain reaction (RT-qPCR). Hsa_circ_TRIM22 was silenced using specific short hairpin ribonucleic acid (shRNA) in CC cell lines and functional assays were performed thereafter. Mechanistically, the targeting and regulatory relationship between hsa_circ_TRIM22 and miR-154-5p were confirmed using the luciferase report assay and rescue experiments. Results. We found hsa_circ_TRIM22 expression level was significantly higher in CC cells and tissues. Further, hsa_circ_TRIM22 knockdown inhibited migration, proliferation, invasiveness, enhanced apoptosis, and slowed the cell cycle. Mechanistically, hsa_circ_TRIM22 could bind miR-154-5p and prevent miR-154-5p from reducing the levels of Cullin2 (CUL2). Notably, the application of miR-154-5p inhibitor significantly rescued hsa_circ_TRIM22-mediated tumorigenesis. Conclusions. Our observations suggest hsa_circ_TRIM22 is upregulated in HPV16-positive CC and promotes CC progression by regulating the miR-154-5p/CUL2 axis, thereby serving as a promising candidate for diagnosis and treatments of CC.

20.
Biochem Biophys Res Commun ; 441(3): 600-6, 2013 Nov 22.
Article in English | MEDLINE | ID: mdl-24183724

ABSTRACT

Tripartite-motif containing 22 (TRIM22) is a direct p53 target gene and inhibits the clonogenic growth of leukemic cells. Its expression in Wilms tumors is negatively associated with disease relapse. This study addresses if TRIM22 expression is de-regulated in breast carcinoma. Western blotting analysis of a panel of 10 breast cancer cell lines and 3 non-malignant mammary epithelial cell lines with a well-characterized TRIM22 monoclonal antibody showed that TRIM22 protein is greatly under-expressed in breast cancer cells as compared to non-malignant cell lines. Similarly, TRIM22 protein is significantly down-regulated in breast tumors as compared to matched normal breast tissues. Study of cell lines with methylation inhibitor and bisulfite sequencing indicates that TRIM22 promoter hypermethylation may not be the cause for TRIM22 under-expression in breast cancer. Instead, we found that TRIM22 protein level correlates strongly (R=0.79) with p53 protein level in normal breast tissue, but this correlation is markedly impaired (R=0.48) in breast cancer tissue, suggesting that there is some defects in p53 regulation of TRIM22 gene in breast cancer. This notion is supported by cell line studies, which showed that TRIM22 was no longer inducible by p53-activating genotoxic drugs in breast cancer cell lines and in a p53 null cell line H1299 transfected with wild type p53. In conclusion, this study shows that TRIM22 is greatly under-expressed in breast cancer. p53 dysfunction may be one of the mechanisms for TRIM22 down-regulation.


Subject(s)
Breast Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Repressor Proteins/genetics , Tumor Suppressor Protein p53/genetics , Antineoplastic Agents/pharmacology , Azacitidine/pharmacology , Camptothecin/pharmacology , DNA Damage , DNA Methylation/drug effects , Down-Regulation , Female , HeLa Cells , Humans , MCF-7 Cells , Minor Histocompatibility Antigens , Paclitaxel/pharmacology , Promoter Regions, Genetic , Topoisomerase I Inhibitors/pharmacology , Tripartite Motif Proteins , Tubulin Modulators/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL