Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
Hepatol Commun ; 5(2): 334-348, 2021 02.
Article in English | MEDLINE | ID: mdl-33553979

ABSTRACT

CKLF-like MARVEL transmembrane domain containing 6 (CMTM6) was identified as a regulator of programmed death ligand 1 (PD-L1), which induces antitumor immunity in several cancers. This study aimed to clarify the relationship between CMTM6 and PD-L1 expression and clinical outcomes in patients with hepatocellular carcinoma (HCC). In total, 259 patients with HCC who had undergone hepatic resection were enrolled. Immunohistochemical staining for CMTM6 and PD-L1 was performed. The relationships between CMTM6 expression and the clinicopathological characteristics and outcomes were analyzed. Additionally, the stabilization of PD-L1 expression and regulation of malignant activities by CMTM6 were examined in vitro. Our patients were divided into high (n = 65, 25.1%) and low (n = 194, 74.9%) CMTM6 expression groups. High CMTM6 expression was significantly associated with malignant aggregates, including poor differentiation (P < 0.0001), microscopic intrahepatic metastasis (P = 0.0369), and multiple intrahepatic recurrences (P = 0.0211). CMTM6 expression was significantly correlated with PD-L1 expression in HCC tissues (P < 0.0001). The patients were classified into three groups: high CMTM6/PD-L1 positive (n = 21), high CMTM6/ PD-L1 negative (n = 44), and low CMTM6 (n = 194) expression pattern groups. Overall survival was significantly different among the three groups (P < 0.0001). Additionally, immunohistochemical double staining revealed that CMTM6 and PD-L1 were co-expressed on HCC cells. In vitro, PD-L1 expression was enhanced at late time points in the presence of CMTM6 expression. CMTM6 also regulated epithelial-to-mesenchymal transition and stemness phenotypes in HCC cells. Conclusion: Our large cohort study found that CMTM6 co-expressed with PD-L1 was strongly associated with the clinical outcome in patients with HCC. The evaluation of CMTM6 combined with PD-L1 in HCC might be useful for patient selection in immune checkpoint therapy.


Subject(s)
B7-H1 Antigen/biosynthesis , Carcinoma, Hepatocellular/metabolism , Liver Neoplasms/metabolism , MARVEL Domain-Containing Proteins/biosynthesis , Myelin Proteins/biosynthesis , Adult , Aged , Aged, 80 and over , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cohort Studies , Epithelial-Mesenchymal Transition , Female , Humans , Liver Neoplasms/pathology , Male , Middle Aged , Prognosis
3.
Cancer Immunol Immunother ; 70(2): 417-429, 2021 Feb.
Article in English | MEDLINE | ID: mdl-32770259

ABSTRACT

The "macrotrabecular-massive" (MTM) pattern of hepatocellular carcinoma (HCC) has been suggested to represent a distinct HCC subtype and is associated with specific molecular features. Since the immune microenvironment is heterogenous in HCC, it is important to evaluate the immune microenvironment of this novel variant. CMTM6, a key regulator of PD-L1, is an important immunocheckpoint inhibitor. This study aimed to evaluate the prognostic effect of CMTM6/PD-L1 coexpression and its relationship with inflammatory cells in HCC. We analyzed 619 HCC patients and tumors were classified into MTM and non-MTM HCC subtypes. The expression levels of CMTM6 and PD-L1 in tumor and inflammatory cells were evaluated by immunohistochemistry. The density of inflammatory cells in the cancer cell nest was calculated. Tumoral PD-L1 expression and inflammatory cell density were higher in the MTM type than in the non-MTM type. CMTM6-high expression was significantly associated with shorter OS and DFS than CMTM6-low expression in the whole HCC patient population and the MTM HCC patient population. Moreover, MTM HCC patients with CMTM6/PD-L1 coexpression experienced a higher risk of HCC progression and death. In addition, CMTM6/PD-L1 coexpression was shown to be related to a high density of inflammatory cells. Notably, a new immune classification, based on CMTM6/PD-L1 coexpression and inflammatory cells, successfully stratified OS and DFS in MTM HCC. CMTM6/PD-L1 coexpression has an adverse effect on the prognosis of HCC patients, especially MTM HCC patients. Our study provides evidence for the combination of immune status assessment with anti-CMTM6 and anti-PD-L1 therapy in MTM HCC patients.


Subject(s)
B7-H1 Antigen/biosynthesis , Carcinoma, Hepatocellular/immunology , Liver Neoplasms/immunology , MARVEL Domain-Containing Proteins/immunology , Myelin Proteins/immunology , Adolescent , Adult , Aged , B7-H1 Antigen/immunology , Carcinoma, Hepatocellular/pathology , Female , Humans , Immunophenotyping , MARVEL Domain-Containing Proteins/biosynthesis , Male , Middle Aged , Myelin Proteins/biosynthesis , Prognosis , Retrospective Studies , Survival Analysis , Young Adult
4.
Oncol Rep ; 42(4): 1459-1466, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31322270

ABSTRACT

The expression of CDR1­AS, a representative circular RNA, is closely linked with poor prognosis in gastrointestinal cancers, such as colon, liver, and pancreatic cancers. Although it is well known that CDR1­AS antagonizes microRNA­7 function through its sequence similarities in the brain, its biological function and link with the malignant potential of cancer cells remain unclear, partly due to the difficulties of ectopic expression of circular RNAs. In the present study, SW620, a colon cancer cell line that stably expresses CDR1­AS RNA circularized, was established using the laccase 2 gene cassette, and its biological function associated with malignant behavior was determined. In contrast to previous studies, cell growth or invasion ability was not altered by CDR1­AS expression. However, the expression levels of CMTM4 and CMTM6, which were recently recognized as critical regulators of PD­L1 protein expression at the cell surface, were significantly increased. Accordingly, the cell surface PD­L1 protein levels were increased in CDR1­AS­expressing cells. Notably, the effects were not canceled out by overexpressing microRNA­7, indicating that the increase in cell surface PD­L1 in CDR1­AS­expressing cells was not dependent on microRNA­7 function. These results indicated that expression of this circular RNA in cancer cells may lead to poor prognosis by increasing cell surface PD­L1 levels through microRNA­7­independent mechanisms.


Subject(s)
B7-H1 Antigen/biosynthesis , Colorectal Neoplasms/metabolism , RNA, Long Noncoding/biosynthesis , Animals , B7-H1 Antigen/genetics , Caco-2 Cells , Cell Growth Processes/physiology , Cell Line, Tumor , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , HEK293 Cells , Humans , Immunohistochemistry , MARVEL Domain-Containing Proteins/biosynthesis , MARVEL Domain-Containing Proteins/genetics , Male , Membrane Proteins/biosynthesis , Membrane Proteins/genetics , Mice , Mice, Inbred BALB C , Mice, Nude , MicroRNAs/antagonists & inhibitors , MicroRNAs/genetics , MicroRNAs/metabolism , Myelin Proteins , Neoplasm Invasiveness , Prognosis , RNA, Long Noncoding/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism
5.
Cancer Biol Ther ; 19(5): 436-444, 2018 05 04.
Article in English | MEDLINE | ID: mdl-29509055

ABSTRACT

OBJECTIVE: This study investigated miR-422a and PLP2 expressions in breast cancer cells and breast cancer stem cells (BCSCs). Besides, their influences on polymorphism changes were observed. METHODS: Flow cytometry and fluorescence-activated cell sorting was performed and CD24-/CD44+ cells were sorted from breast cancer cells and recognized as BCSCs. Microarray was applied to search for the differentially expressed miRNAs and mRNAs between MCF7 and BCSCs. The aberrant expression of miR-422a and PLP2 was further confirmed by RT-qPCR and the direct targeted relationship was verified by dual-luciferase reporter assay. After in vitro transfection, the expression of miR-422a and PLP2 were manipulated and biological functions of BMSCs were compared with CCK-8, colony formation and sphere formation assay. The tumorigenesis ability of transfected BMSCs was also investigated in NOD/SCID tumor mice models. RESULTS: BMSCs were successfully established from MCF7 cells and miR-422a expression was downregulated while PLP2 level decreased in BMSCs. MiR-422a directly targets the 3'UTR of PLP2 and suppressed its expression. Besides, the up-regulation of miR-422a contributed to weakened ability of proliferation and microsphere formation of BMSCs, while PLP2 overexpression facilitated those biological abilities. Tumorigenesis of BMSCs in mice models was impaired by either overexpression of miR-442a or silencing of PLP2. CONCLUSION: Up-regulation of miR-422a attenuated microsphere formation, proliferation and tumor formation of breast cancer stem cells via suppressing the PLP2 expression.


Subject(s)
Breast Neoplasms/pathology , MARVEL Domain-Containing Proteins/metabolism , MicroRNAs/metabolism , Neoplastic Stem Cells/pathology , Proteolipids/metabolism , Animals , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Line, Tumor , Female , Heterografts , Humans , MARVEL Domain-Containing Proteins/biosynthesis , MARVEL Domain-Containing Proteins/genetics , MCF-7 Cells , Mice, Inbred NOD , Mice, SCID , MicroRNAs/biosynthesis , MicroRNAs/genetics , Neoplastic Stem Cells/metabolism , Proteolipids/biosynthesis , Proteolipids/genetics , Transfection
6.
Oncol Res ; 25(2): 285-293, 2017 Jan 26.
Article in English | MEDLINE | ID: mdl-27629543

ABSTRACT

The CKLF-like MARVEL transmembrane domain-containing 3 (CMTM3), a member of the CMTM family, was found in several human tumors and plays an important role in the development and progression of tumors. However, the role of CMTM3 in hepatocellular carcinoma (HCC) remains largely unknown. Thus, in the present study, we explored its expression pattern in human HCC cell lines, as well as its functions in HCC cells. Our results demonstrated that the expression of CMTM3 is lowly expressed in HCC cell lines. In vitro, we found that overexpression of CMTM3 obviously inhibited the proliferation, invasion, and EMT process in HCC cells. Furthermore, overexpression of CMTM3 significantly downregulated the expression levels of phosphorylation of JAK2 and STAT3 in HepG2 cells. In vivo, overexpression of CMTM3 attenuated the tumor growth in Balb/c nude mice. In conclusion, we demonstrated that CMTM3 could play an important role in HCC metastasis by EMT induction via, at least partially, suppressing the JAK2/STAT3 signaling pathway. Therefore, CMTM3 may serve as a potential molecular target in the prevention and/or treatment of HCC invasion and metastasis.


Subject(s)
Carcinogenesis/metabolism , Carcinoma, Hepatocellular/metabolism , Cell Proliferation/physiology , Chemokines/biosynthesis , Liver Neoplasms/metabolism , MARVEL Domain-Containing Proteins/biosynthesis , Animals , Carcinogenesis/genetics , Carcinogenesis/pathology , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Chemokines/genetics , Female , Hep G2 Cells , Humans , Liver Neoplasms/genetics , Liver Neoplasms/pathology , MARVEL Domain-Containing Proteins/genetics , Mice , Mice, Inbred BALB C , Mice, Nude
7.
Tumour Biol ; 37(5): 6217-25, 2016 May.
Article in English | MEDLINE | ID: mdl-26615421

ABSTRACT

Previous research revealed that CMTM8 acts as a tumor suppressor gene in variety cancers. However, the role of CMTM8 in bladder cancer has never been reported. In this study, the expression profile of CMTM8 was examined in bladder cancer tissues and bladder cancer cell lines. The effects of CMTM8 on bladder cancer cell proliferation, apoptosis, migration, and invasion were examined. Bladder tumor tissues from 84 cases were examined for CMTM8 expression by immunohistochemistry. Disease-specific survival was investigated using a Kaplan-Meier analysis, and Cox proportional hazards analysis was assessed. Our results showed that upregulation of CMTM8 in the T24 cell line could suppress T24 cells proliferation, migration and invasion and enhance the sensitivity to Epirubicin. Kaplan-Meier analysis revealed that the expression of CMTM8 was correlated with the survival time of bladder cancer patients. Altogether, our data suggested that CMTM8 is an important tumor suppressor gene in human bladder cancer and qualified as a useful prognostic indicator for patients with bladder cancer.


Subject(s)
Biomarkers, Tumor/genetics , Chemokines/genetics , MARVEL Domain-Containing Proteins/genetics , Prognosis , Urinary Bladder Neoplasms/genetics , Adult , Aged , Apoptosis/genetics , Biomarkers, Tumor/biosynthesis , Cell Line, Tumor , Cell Proliferation/genetics , Chemokines/biosynthesis , Female , Gene Expression Regulation, Neoplastic , Humans , Kaplan-Meier Estimate , MARVEL Domain-Containing Proteins/biosynthesis , Male , Middle Aged , Tumor Suppressor Proteins/genetics , Urinary Bladder Neoplasms/pathology
8.
Oncol Rep ; 34(6): 2853-63, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26503336

ABSTRACT

Bladder cancer is the most common tumor of the urinary tract. The incidence of bladder cancer has increased in the last few decades, thus novel molecular markers for early diagnosis and more efficacious treatment are urgently needed. Chemokine­like factor (CKLF)­like MARVEL transmembrane domain containing 8 (CMTM8) is downregulated in several types of cancers and is associated with tumor progression. However, CMTM8 expression has been unexplored in bladder cancer to date. Our results revealed that the expression of CMTM8 was negative in 46 of 74 (62.2%) bladder cancer samples via immunohistochemistry assay. CMTM8 downregulation was associated with advancing tumor stage and tumor grade. CMTM8 was successfully overexpressed by lentivirus in EJ and T24 cells, and the CCK­8 and Transwell assays showed that CMTM8 overexpression decreased cell proliferation, migration and invasion in vitro. In tumor xenografts upregulation of CMTM8 inhibited tumor growth and lymph node metastasis in vivo. In conclusion, overexpression of CMTM8 in bladder cancer results in reduced malignant cell growth, migration and invasion, which could make it a potential therapeutic target in the treatment of bladder cancer.


Subject(s)
Carcinogenesis/genetics , Cell Proliferation/genetics , Chemokines/biosynthesis , MARVEL Domain-Containing Proteins/biosynthesis , Urinary Bladder Neoplasms/genetics , Adult , Aged , Animals , Apoptosis/genetics , Cell Line, Tumor , Chemokines/genetics , Disease Progression , Female , Gene Expression Regulation, Neoplastic , Humans , MARVEL Domain-Containing Proteins/genetics , Male , Mice , Middle Aged , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Urinary Bladder Neoplasms/pathology , Xenograft Model Antitumor Assays
9.
J Exp Clin Cancer Res ; 34: 122, 2015 Oct 16.
Article in English | MEDLINE | ID: mdl-26474560

ABSTRACT

BACKGROUND: Chemokine-like factor (CKLF)-like MARVEL transmembrane domain-containing family (CMTM) is a gene family involved in multiple malignancies. CMTM4 is a member of this family and is located at chromosome 16q22.1, a locus that harbours a number of tumour suppressor genes. It has been defined as a regulator of cell cycle and division in HeLa cells; however, its roles in tumourigenesis remain poorly studied. METHODS: An integrated bioinformatics analysis based on the array data from the GEO database was conducted to view the differential expression of CMTM4 across multiple cancers and their corresponding control tissues. Primary clear cell renal cell carcinoma (ccRCC) and the paired adjacent non-tumour tissues were then collected to examine the expression of CMTM4 by western blotting, immunohistochemistry, and quantitative RT-PCR. The ccRCC cell lines A498 and 786-O and the normal renal tubular epithelial cell line HK-2 were also tested for CMTM4 expression by western blotting. Cell Counting Kit-8 (CCK-8) and viable cell counting assays were used to delineate the growth curves of 786-O cells after CMTM4 overexpression or knockdown. Wound healing and transwell assays were performed to assess the cells' ability to migrate. The effects of CMTM4 on cellular apoptosis and cell cycle progression were analysed by flow cytometry, and cell cycle hallmarks were detected by western blotting and RT-PCR. The xenograft model in nude mice was used to elucidate the function of CMTM4 in tumourigenesis ex vivo. RESULTS: By omic data analysis, we found a substantial downregulation of CMTM4 in ccRCC. Western blotting then confirmed that CMTM4 was dramatically reduced in 86.9 % (53/61) of ccRCC tissues compared with the paired adjacent non-tumour tissues, as well as in the 786-O and A498 ccRCC cell lines. Restoration of CMTM4 significantly suppressed 786-O cell growth by inducing G2/M cell cycle arrest and p21 upregulation, and cell migration was also inhibited. However, knockdown of CMTM4 led to a completely opposite effect on these cell behaviours. Overexpression of CMTM4 also markedly inhibited the tumour xenograft growth in nude mice. CONCLUSIONS: CMTM4 is downregulated and exhibits tumour-suppressor activities in ccRCC, and could be exploited as a target for ccRCC treatment.


Subject(s)
Carcinoma, Renal Cell/genetics , Genes, Tumor Suppressor , Kidney Neoplasms/genetics , MARVEL Domain-Containing Proteins/genetics , Tumor Suppressor Proteins/genetics , Adult , Aged , Animals , Apoptosis/genetics , Carcinoma, Renal Cell/pathology , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Cell Transformation, Neoplastic/genetics , Cyclin-Dependent Kinase Inhibitor p21/biosynthesis , Down-Regulation , Female , G2 Phase Cell Cycle Checkpoints/genetics , Humans , Kidney Neoplasms/pathology , MARVEL Domain-Containing Proteins/biosynthesis , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Middle Aged , Neoplasm Transplantation , RNA Interference , RNA, Small Interfering , Transplantation, Heterologous , Young Adult
10.
Sci Rep ; 5: 13014, 2015 Aug 11.
Article in English | MEDLINE | ID: mdl-26260329

ABSTRACT

A leucine-rich protein, ARR19 (androgen receptor corepressor-19 kDa), is highly expressed in male reproductive organs and moderately in others. Previously, we have reported that ARR19 is differentially expressed in adult Leydig cells during the testis development and inhibits steroidogenesis by reducing the expression of steroidogenic enzymes. Whereas in prostate, ARR19 represses the transcriptional activity of AR (androgen receptor), it is important for male sexual differentiation and maturation in prostate and epididymis, through the recruitment of HDAC4. In this study we show that long term adenovirus mediated overexpression of ARR19 in mice testis has the potential of inhibiting the differentiation of testicular and prostatic cells by reducing the size of testis and prostate but has no effect on the growth of seminal vesicles. Further, it reduces the level of progesterone and testosterone by reducing the steroidogenic enzymes such as 3HSD, P450c17 and StAR. This is the first study reporting a time-course analysis of the implications of long term overexpression of ARR19 in mice testis and its effect on other organs such as prostate and seminal vesicles. Taken together, these results suggest that ARR19 may play an important role in the differentiation of male reproductive organs such as testis and prostate.


Subject(s)
Cell Differentiation/genetics , MARVEL Domain-Containing Proteins/biosynthesis , Prostate/growth & development , Repressor Proteins/biosynthesis , Testis/growth & development , Adenoviridae/genetics , Animals , Epididymis/growth & development , Epididymis/metabolism , Gene Expression Regulation, Developmental , Histone Deacetylases/genetics , Humans , MARVEL Domain-Containing Proteins/genetics , Male , Mice , Progesterone/metabolism , Prostate/metabolism , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , Repressor Proteins/genetics , Testis/metabolism , Testosterone/metabolism
11.
Dev Biol ; 401(2): 299-309, 2015 May 15.
Article in English | MEDLINE | ID: mdl-25797154

ABSTRACT

In Drosophila, myoblast fusion is a conserved process in which founder cells (FCs) and fusion competent myoblasts (FCMs) fuse to form a syncytial muscle fiber. Mutants for the myogenic regulator Myocyte enhancer factor-2 (MEF2) show a failure of myoblast fusion, indicating that MEF2 regulates the fusion process. Indeed, chromatin immunoprecipitation studies show that several genes involved in myoblast fusion are bound by MEF2 during embryogenesis. Of these, the MARVEL domain gene singles bar (sing), is down-regulated in MEF2 knockdown pupae, and has five consensus MEF2 binding sites within a 9000-bp region. To determine if MEF2 is an essential and direct regulator of sing during pupal muscle development, we identified a 315-bp myoblast enhancer of sing. This enhancer was active during myoblast fusion, and mutation of two MEF2 sites significantly decreased enhancer activity. We show that lack of sing expression resulted in adult lethality and muscle loss, due to a failure of fusion during the pupal stage. Additionally, we sought to determine if sing was required in either FCs or FCMs to support fusion. Interestingly, knockdown of sing in either population did not significantly affect fusion, however, knockdown in both FCs and FCMs resulted in muscles with significantly reduced nuclei numbers, provisionally indicating that sing function is required in either cell type, but not both. Finally, we found that MEF2 regulated sing expression at the embryonic stage through the same 315-bp enhancer, indicating that sing is a MEF2 target at both critical stages of myoblast fusion. Our studies define for the first time how MEF2 directly controls fusion at multiple stages of the life cycle, and provide further evidence that the mechanisms of fusion characterized in Drosophila embryos is also used in the formation of the more complex adult muscles.


Subject(s)
Drosophila Proteins/genetics , Drosophila/embryology , Membrane Proteins/genetics , Myoblasts/cytology , Myogenic Regulatory Factors/genetics , Transcriptional Activation/genetics , Animals , Animals, Genetically Modified , Base Sequence , Binding Sites/genetics , Cell Fusion , Cell Nucleus/genetics , Chromatin Immunoprecipitation , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/genetics , Electrophoretic Mobility Shift Assay , Gene Expression Regulation, Developmental , Giant Cells/cytology , MARVEL Domain-Containing Proteins/biosynthesis , MARVEL Domain-Containing Proteins/genetics , Molecular Sequence Data , Muscle Development/genetics , Muscle Fibers, Skeletal , RNA Interference , RNA, Small Interfering
12.
Clin Transl Oncol ; 17(6): 431-7, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25387568

ABSTRACT

PURPOSE: A novel tumor suppressor CKLF-like MARVEL transmembrane domain-containing member 5 (CMTM5) is reduced or undetectable in many kinds of cancers and inhibits tumor cells' malignant features. To explore its role in prostate cancer (PCa), we detected its expression patterns in prostate tissues and PCa cells, and determined its anti-proliferation functions in PCa cells in vitro and in vivo. METHODS: The expression of CMTM5 in prostate tissue microarray, specimens and cell lines was evaluated by immunohistochemistry and Western blot, respectively. After being transfected with CMTM5 adenovirus or vector, the proliferation and migration of DU145 cells were detected by MTT assay and transwell assay, respectively. Furthermore, the effects of CMTM5 on tumor growth were performed in nude mice xenograft in vivo. RESULTS: We found CMTM5 was reduced in PCa tissues and cells compared with BPH tissues, and its expression in PCa tissues was related to the Gleason score. Moreover, after being transfected with adenovirus, ectopic expression of CMTM5-v1 in DU145 cells led to significant inhibition of cell proliferation and migration compared with the control, which may be attributed to decreased Akt activity. Finally, restoration of CMTM5 significantly suppressed tumor growth in vivo. CONCLUSIONS: These results indicate that CMTM5 is down-regulated in PCa and exhibit tumor suppressor activities in androgen-independent PCa cells. Loss of CMTM5 protein may be contributed to the development of PCa and it is a potential therapeutic target for castration-resistant prostate cancer.


Subject(s)
Chemokines/biosynthesis , MARVEL Domain-Containing Proteins/biosynthesis , Prostatic Neoplasms/pathology , Tumor Suppressor Proteins/biosynthesis , Aged , Animals , Blotting, Western , Cell Proliferation , Down-Regulation , Heterografts , Humans , Immunohistochemistry , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Middle Aged , Prostatic Neoplasms/metabolism , Tissue Array Analysis
13.
PLoS Pathog ; 9(11): e1003772, 2013.
Article in English | MEDLINE | ID: mdl-24278019

ABSTRACT

The Kaposi's sarcoma-associated herpesvirus gene products K3 and K5 are viral ubiquitin E3 ligases which downregulate MHC-I and additional cell surface immunoreceptors. To identify novel cellular genes required for K5 function we performed a forward genetic screen in near-haploid human KBM7 cells. The screen identified proteolipid protein 2 (PLP2), a MARVEL domain protein of unknown function, as essential for K5 activity. Genetic loss of PLP2 traps the viral ligase in the endoplasmic reticulum, where it is unable to ubiquitinate and degrade its substrates. Subsequent analysis of the plasma membrane proteome of K5-expressing KBM7 cells in the presence and absence of PLP2 revealed a wide range of novel K5 targets, all of which required PLP2 for their K5-mediated downregulation. This work ascribes a critical function to PLP2 for viral ligase activity and underlines the power of non-lethal haploid genetic screens in human cells to identify the genes involved in pathogen manipulation of the host immune system.


Subject(s)
Cell Membrane/metabolism , Down-Regulation , Herpesvirus 8, Human/enzymology , Immediate-Early Proteins/biosynthesis , MARVEL Domain-Containing Proteins/biosynthesis , Proteolipids/biosynthesis , Ubiquitin-Protein Ligases/biosynthesis , Viral Proteins/biosynthesis , Cell Membrane/genetics , Cell Membrane/immunology , Genetic Testing , HeLa Cells , Hep G2 Cells , Herpesvirus 8, Human/genetics , Herpesvirus 8, Human/immunology , Humans , Immediate-Early Proteins/genetics , MARVEL Domain-Containing Proteins/genetics , MARVEL Domain-Containing Proteins/immunology , Proteolipids/genetics , Proteolipids/immunology , Sarcoma, Kaposi/genetics , Sarcoma, Kaposi/immunology , Sarcoma, Kaposi/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/immunology , Viral Proteins/genetics , Viral Proteins/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...