Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters










Publication year range
1.
BMC Genomics ; 25(1): 498, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38773419

ABSTRACT

BACKGROUND: The hair follicle development process is regulated by sophisticated genes and signaling networks, and the hair grows from the hair follicle. The Tianzhu white yak population exhibits differences in hair length, especially on the forehead and shoulder region. However, the genetic mechanism is still unclear. Isoform sequencing (Iso-seq) technology with advantages in long reads sequencing. Hence, we combined the Iso-seq and RNA-seq methods to investigate the transcript complexity and difference between long-haired yak (LHY) and normal-haired yak (NHY). RESULTS: The hair length measurement result showed a significant difference between LHY and NHY on the forehead and the shoulder (P-value < 0.001). The skin samples from the forehead and the shoulder of LHY and NHY were pooled for isoform sequencing (Iso-seq). We obtained numerous long transcripts, including novel isoforms, long non-coding RNA, alternative splicing events, and alternative polyadenylation events. Combined with RNA-seq data, we performed differential isoforms (DEIs) analysis between LHY and NHY. We found that some hair follicle and skin development-related DEIs, like BMP4, KRT2, IGF2R, and COL1A2 in the forehead skin; BMP1, KRT1, FGF5, COL2A1, and IGFBP5 in the shoulder skin. Enrichment analysis revealed that DEIs in both two comparable groups significantly participated in skin and hair follicle development-related pathways, like ECM-receptor interaction, focal adhesion, and PI3K-Akt signaling pathways. The results indicated that the hair follicle development of Tianzhu white yak may influence the hair length difference. Besides, the protein-protein interaction (PPI) network of DEIs showed COL2A1 and COL3A1 exhibited a high degree of centrality, and these two genes were suggested as potential candidates for the hair length growth of Tianzhu white yak. CONCLUSIONS: The results provided a comprehensive analysis of the transcriptome complexity and identified differential transcripts that enhance our understanding of the molecular mechanisms underlying the variation in hair length growth in Tianzhu white yak.


Subject(s)
Hair , Protein Isoforms , RNA-Seq , Skin , Transcriptome , Animals , Cattle/genetics , Skin/metabolism , Hair/metabolism , Hair/growth & development , Protein Isoforms/genetics , Protein Isoforms/metabolism , Hair Follicle/metabolism , Hair Follicle/growth & development , Gene Expression Profiling , Alternative Splicing , Sequence Analysis, RNA
3.
Cells ; 11(24)2022 12 09.
Article in English | MEDLINE | ID: mdl-36552749

ABSTRACT

The interaction between the dermal papilla cells (DPCs) and epidermal hair matrix cells (HMCs) of hair follicles (HFs) is crucial for the growth and development of HFs, but the molecular mechanism is complex and remains unclear. MicroRNAs (miRNAs) are the key signaling molecules for cellular communication. In this study, the DPCs and HMCs of yak were isolated and cultured, and the differentially expressed mRNA and miRNA were characterized to analyze the molecular basis of the interaction between DPCs and HMCs during hair follicle (HF) development in yak. The mRNA differential expression and functional enrichment analysis revealed that there were significant differences between DPCs and HMCs, and they showed the molecular functional characteristics of dermal cells and epidermal cells, respectively. Multiple KEGG pathways related to HF development were enriched in the highly expressed genes in DPCs, while the pathways associated with microbiota and immunity were significantly enriched in the highly expressed genes in HMCs. By combining analysis with our previous 10× genomics single-cell transcriptome data, 39 marker genes of DPCs of yak were identified. A total of 123 relatively specifically expressed miRNAs were screened; among these, the miRNAs associated with HF development such as miR-143, miR-214, miR-125b, miR-31, and miR-200 were presented. In conclusion, the large changes in yak DPCs and HMCs for both mRNA and miRNA expression were revealed, and numerous specifically expressed mRNAs and miRNAs in DPCs or HMCs were identified, which may contribute to the interaction and cellular communication between DPCs and HMCs during HF development in yak.


Subject(s)
Hair Follicle , MicroRNAs , Animals , Cattle , MicroRNAs/genetics , MicroRNAs/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Cells, Cultured , Signal Transduction
5.
Cancer Treat Res ; 177: 23-62, 2019.
Article in English | MEDLINE | ID: mdl-30523620

ABSTRACT

Discovered in 1994, Kaposi's sarcoma-associated herpesvirus (KSHV) has been associated with four human malignancies including Kaposi's sarcoma, primary effusion lymphoma, a subset of multicentric Castleman's disease, and KSHV inflammatory cytokine syndrome. These malignancies mostly occur in immunocompromised patients including patients with acquired immunodeficiency syndrome and often cause significant mortality because of the lack of effective therapies. Significant progresses have been made to understand the molecular basis of KSHV infection and KSHV-induced oncogenesis in the last two decades. This chapter provides an update on the recent advancements focusing on the molecular events of KSHV primary infection, the mechanisms regulating KSHV life cycle, innate and adaptive immunity, mechanism of KSHV-induced tumorigenesis and inflammation, and metabolic reprogramming in KSHV infection and KSHV-transformed cells.


Subject(s)
Acquired Immunodeficiency Syndrome/virology , Herpesviridae Infections/genetics , Herpesvirus 8, Human/genetics , Herpesvirus 8, Human/immunology , Neoplasms/virology , Acquired Immunodeficiency Syndrome/complications , Carcinogenesis/genetics , Carcinogenesis/immunology , Castleman Disease/physiopathology , Castleman Disease/virology , Coinfection/virology , Cytokines/immunology , HIV Infections/complications , HIV Infections/virology , Herpesviridae Infections/complications , Herpesviridae Infections/immunology , Herpesviridae Infections/virology , Herpesvirus 8, Human/physiology , Humans , Immunocompromised Host , Inflammation/immunology , Inflammation/physiopathology , Inflammation/virology , Lymphoma, Primary Effusion/physiopathology , Lymphoma, Primary Effusion/virology , Neoplasms/genetics , Neoplasms/immunology , Neoplasms/physiopathology , Sarcoma, Kaposi/genetics , Sarcoma, Kaposi/virology , Syndrome , Viremia/immunology , Viremia/physiopathology , Viremia/virology
6.
J Pathol ; 242(3): 309-321, 2017 07.
Article in English | MEDLINE | ID: mdl-28393364

ABSTRACT

Primary effusion lymphoma (PEL) is a rare and aggressive B-cell lymphoma with a dismal prognosis caused by infection of Kaposi's sarcoma-associated herpesvirus. Despite the findings that numerous viral genes and cellular pathways are essential for the proliferation and survival of PEL cells, there is currently no effective therapeutic treatment for PEL. Here, we report that the metabolic sensor SIRT1 is functionally required for sustaining the proliferation and survival of PEL cells. Knockdown of SIRT1 with specific shRNAs or inhibition of SIRT1 with an inhibitor (tenovin-6) induced cell cycle arrest and apoptosis in PEL cells. We detected high levels of AMPK activation in PEL cells, reflected in AMPKα1 phosphorylation at T174. Knockdown or inhibition of SIRT1 reduced AMPK activation, indicating that SIRT1 was required for AMPK activation. Interestingly, knockdown of AMPK with specific shRNAs or inhibition of AMPK with the inhibitor compound C recapitulated the phenotype of SIRT1, and induced cell cycle arrest and apoptosis, whereas overexpression of a constitutively active AMPK construct rescued the cytotoxic effect of SIRT1 knockdown. Remarkably, treatment with tenovin-6 effectively inhibited the initiation and progression of PEL, and significantly extended the survival of mice in a murine PEL model. Taken together, these results illustrate that the SIRT1-AMPK axis is essential for maintaining the proliferation and survival of PEL and identify SIRT1 and AMPK as potential therapeutic targets, and tenovin-6 as a candidate therapeutic agent for PEL patients. Copyright © 2017 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
AMP-Activated Protein Kinases/physiology , Lymphoma, Primary Effusion/physiopathology , Sirtuin 1/physiology , AMP-Activated Protein Kinases/metabolism , Animals , Antineoplastic Agents/pharmacology , Apoptosis/physiology , Benzamides/pharmacology , Cell Cycle Checkpoints/physiology , Cell Proliferation/physiology , Cell Survival/physiology , Dose-Response Relationship, Drug , Gene Knockdown Techniques , Lymphoma, Primary Effusion/enzymology , MAP Kinase Signaling System/physiology , Mice, Inbred NOD , Mice, SCID , Phosphorylation/physiology , Sirtuin 1/antagonists & inhibitors
7.
Oncotarget ; 8(9): 14912-14924, 2017 Feb 28.
Article in English | MEDLINE | ID: mdl-28118604

ABSTRACT

Diffuse large B-cell lymphoma (DLBCL) is one of the most aggressive non-Hodgkin lymphomas. It is curable but one-third of cases are refractory to therapy or relapse after initial response highlighting the urgent need for developing novel therapeutic approaches. Targeting sirtuins, particularly SIRT1 by genetic approaches or using pharmaceutical inhibitor tenovin-6, has shown promising therapeutic potential in various hematopoietic malignancies. However, it remains unknown whether these approaches are effective for DLBCL. In this study, we have found that tenovin-6 potently inhibits the proliferation and survival of DLBCL cells. Surprisingly, specific knockdown of SIRT1/2/3 has no effect on DLBCL. Mechanistically, tenovin-6 increases the level of microtubule-associated protein 1 light chain 3B (LC3B)-II in a SIRT1/2/3- and p53-independent manner in DLBCL cell lines. Tenovin-6-mediated increase of LC3B-II is through inhibition of classical autophagy pathway. Furthermore, inhibition of the autophagy pathway by using other inhibitors or by knocking down key genes in the pathway impairs cell proliferation and survival of DLBCL cells. These results indicate that targeting the autophagic pathway could be a novel therapeutic strategy for DLBCL and that precaution should be taken to interpret data where tenovin-6 was used as an inhibitor of sirtuins.


Subject(s)
Apoptosis/drug effects , Autophagy/drug effects , Benzamides/pharmacology , Biomarkers, Tumor/metabolism , Cell Proliferation/drug effects , Lymphoma, Large B-Cell, Diffuse/drug therapy , Microtubule-Associated Proteins/metabolism , Antineoplastic Agents/pharmacology , Humans , Lymphoma, Large B-Cell, Diffuse/metabolism , Lymphoma, Large B-Cell, Diffuse/pathology , Tumor Cells, Cultured
8.
Oncotarget ; 7(46): 75698-75711, 2016 11 15.
Article in English | MEDLINE | ID: mdl-27708228

ABSTRACT

Kaposi's sarcoma-associated herpesvirus (KSHV) is an oncogenic virus associated with Kaposi's sarcoma (KS), a malignancy commonly found in AIDS patients. Despite intensive studies in the last two decades, the mechanism of KSHV-induced cellular transformation and tumorigenesis remains unclear. In this study, we found that the expression of SIRT1, a metabolic sensor, was upregulated in a variety of KSHV-infected cells. In a model of KSHV-induced cellular transformation, SIRT1 knockdown with shRNAs or knockout by CRISPR/Cas9 gene editing dramatically suppressed cell proliferation and colony formation in soft agar of KSHV-transformed cells by inducing cell cycle arrest and contact inhibition. SIRT1 knockdown or knockout induced the expression of cyclin-dependent kinase inhibitor 1B (p27Kip1). Consequently, p27 knockdown rescued the inhibitory effect of SIRT1 knockdown or knockout on cell proliferation and colony formation. Furthermore, treatment of KSHV-transformed cells with a SIRT1 inhibitor, nicotinamide (NAM), had the same effect as SIRT1 knockdown and knockout. NAM significantly inhibited cell proliferation in culture and colony formation in soft agar, and induced cell cycle arrest. Significantly, NAM inhibited the progression of tumors and extended the survival of mice in a KSHV-induced tumor model. Collectively, these results demonstrate that SIRT1 suppression of p27 is required for KSHV-induced tumorigenesis and identify a potential therapeutic target for KS.


Subject(s)
Contact Inhibition/genetics , Cyclin-Dependent Kinase Inhibitor p27/genetics , Herpesvirus 8, Human/physiology , Sarcoma, Kaposi/etiology , Sarcoma, Kaposi/metabolism , Sirtuin 1/metabolism , Cell Line, Transformed , Cell Proliferation , Colony-Forming Units Assay , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Gene Knockout Techniques , Humans , Models, Biological , Sarcoma, Kaposi/pathology , Sirtuin 1/genetics
9.
J Virol ; 90(14): 6515-6525, 2016 07 15.
Article in English | MEDLINE | ID: mdl-27147746

ABSTRACT

UNLABELLED: The host intracellular antiviral restriction factors inhibit viral infection and replication. The 5'-AMP-activated protein kinase (AMPK) is a cellular energy sensor regulating metabolic homeostasis. Activated AMPK inhibits the replication of numerous RNA viruses but enhances the entry of vaccinia virus. However, the role of AMPK in herpesvirus infection is unclear. In this study, we showed that the constitutive AMPK activity restricted Kaposi's sarcoma-associated herpesvirus (KSHV) lytic replication in primary human umbilical vein endothelial cells while KSHV infection did not markedly affect the endogenous AMPK activity. Knockdown of the AMPKα1 considerably enhanced the expression of viral lytic genes and the production of infectious virions, while overexpression of a constitutively active AMPK had the opposite effects. Accordingly, an AMPK inhibitor, compound C, augmented viral lytic gene expressions and virion productions but an AMPK agonist, 5-aminoimidazole-4-carboxamide ribonucleotide (AICAR), suppressed both. Furthermore, a common diabetes drug, metformin, which carries an AMPK-agonistic activity, drastically inhibited the expression of viral lytic genes and the production of infectious virions, suggesting the use of metformin as a therapeutic agent for KSHV infection and replication. Together, these results identify the host AMPK as a KSHV restriction factor that can serve as a potential therapeutic target. IMPORTANCE: Host cells encode specific proteins to restrict viral infection and replication. Kaposi's sarcoma-associated herpesvirus (KSHV) is a human tumor virus associated with several cancers. In this study, we have identified 5'-AMP-activated protein kinase (AMPK), a cellular energy sensor, as a restriction factor of KSHV lytic replication during primary infection. Activation of AMPK suppresses, while inhibition of AMPK enhances, KSHV lytic replication by regulating the expression of viral genes. AICAR and metformin, both of which are AMPK agonists currently used in clinics for the treatment of conditions associated with metabolic disorders, inhibit KSHV lytic replication. Thus, our work has identified AMPK as a potential therapeutic target and AICAR and metformin as potential therapeutic agents for KSHV-associated cancers.


Subject(s)
AMP-Activated Protein Kinases/antagonists & inhibitors , Gene Expression Regulation, Enzymologic/drug effects , Herpesvirus 8, Human/drug effects , Sarcoma, Kaposi/drug therapy , Virus Activation/drug effects , Virus Latency/drug effects , Virus Replication/drug effects , AMP-Activated Protein Kinases/metabolism , Herpesvirus 8, Human/enzymology , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Hypoglycemic Agents/pharmacology , Metformin/pharmacology , Phosphorylation/drug effects , Sarcoma, Kaposi/metabolism , Sarcoma, Kaposi/virology
10.
PLoS Pathog ; 12(5): e1005648, 2016 05.
Article in English | MEDLINE | ID: mdl-27187079

ABSTRACT

Aerobic glycolysis is essential for supporting the fast growth of a variety of cancers. However, its role in the survival of cancer cells under stress conditions is unclear. We have previously reported an efficient model of gammaherpesvirus Kaposi's sarcoma-associated herpesvirus (KSHV)-induced cellular transformation of rat primary mesenchymal stem cells. KSHV-transformed cells efficiently induce tumors in nude mice with pathological features reminiscent of Kaposi's sarcoma tumors. Here, we report that KSHV promotes cell survival and cellular transformation by suppressing aerobic glycolysis and oxidative phosphorylation under nutrient stress. Specifically, KSHV microRNAs and vFLIP suppress glycolysis by activating the NF-κB pathway to downregulate glucose transporters GLUT1 and GLUT3. While overexpression of the transporters rescues the glycolytic activity, it induces apoptosis and reduces colony formation efficiency in softagar under glucose deprivation. Mechanistically, GLUT1 and GLUT3 inhibit constitutive activation of the AKT and NF-κB pro-survival pathways. Strikingly, GLUT1 and GLUT3 are significantly downregulated in KSHV-infected cells in human KS tumors. Furthermore, we have detected reduced levels of aerobic glycolysis in several KSHV-infected primary effusion lymphoma cell lines compared to a Burkitt's lymphoma cell line BJAB, and KSHV infection of BJAB cells reduced aerobic glycolysis. These results reveal a novel mechanism by which an oncogenic virus regulates a key metabolic pathway to adapt to stress in tumor microenvironment, and illustrate the importance of fine-tuning the metabolic pathways for sustaining the proliferation and survival of cancer cells, particularly under stress conditions.


Subject(s)
Adaptation, Physiological/physiology , Cell Transformation, Viral/physiology , Herpesviridae Infections/metabolism , Animals , Blotting, Western , Cell Line, Tumor , Cell Survival , Disease Models, Animal , Flow Cytometry , Fluorescent Antibody Technique , Gene Knockdown Techniques , Glycolysis/physiology , Herpesvirus 8, Human/metabolism , Humans , Lymphoma, Primary Effusion/metabolism , Lymphoma, Primary Effusion/virology , Microscopy, Confocal , Polymerase Chain Reaction , Rats
11.
Cell Cycle ; 13(21): 3328-30, 2014.
Article in English | MEDLINE | ID: mdl-25485577

ABSTRACT

Viruses often hijack cellular functions to facilitate their infection and replication. SIRT1, one of the most widely studied sirtuins, functions as both metabolic sensor and transcriptional regulator. SIRT1 has broad cellular functions including metabolic homeostasis, stress response, tumorigenesis and autophagy. The role of SIRT1 in the life cycle of viruses remains unclear. Like all herpesviruses, oncogenic gammaherpesvirus KSHV has both latent and lytic phases. In a recent study, we have shown that SIRT1 binds to the promoter and silence the expression of KSHV replication and transcription activator (RTA), a key activator of viral lytic replication. Chemical inhibition or knock down of SIRT1 is sufficient to initiate the lytic replication program by increasing active histone H3 trimethyl Lys4 (H3K4me3) mark and decreasing repressive histone H3 trimethyl Lys27 (H3K27me3) mark in the RTA promoter. SIRT1 also interacts with RTA and inhibits RTA transactivation of its own promoter and those of downstream target genes. Our findings reveal that SIRT1 regulates KSHV latency by inhibiting different stages of viral lytic replication, and link a metabolic sensor and transcriptional regulator SIRT1 to KSHV life cycle.


Subject(s)
Herpesvirus 8, Human/physiology , Sirtuin 1/metabolism , Caloric Restriction , Cell Line , DNA Replication , Herpesvirus 8, Human/genetics , Histones/metabolism , Humans , NAD/metabolism , Promoter Regions, Genetic , Sirtuin 1/antagonists & inhibitors , Trans-Activators/genetics , Trans-Activators/metabolism , Transcriptional Activation , Viral Proteins/genetics , Viral Proteins/metabolism , Virus Latency
12.
J Virol ; 88(11): 6355-67, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24672028

ABSTRACT

UNLABELLED: Kaposi's sarcoma-associated herpesvirus (KSHV) establishes persistent latent infection in immunocompetent hosts. Disruption of KSHV latency results in viral lytic replication, which promotes the development of KSHV-related malignancies in immunocompromised individuals. While inhibitors of classes I and II histone deacetylases (HDACs) potently reactivate KSHV from latency, the role of class III HDAC sirtuins (SIRTs) in KSHV latency remains unclear. Here, we examined the effects of inhibitors of SIRTs, nicotinamide (NAM) and sirtinol, on KSHV reactivation from latency. Treatment of latently KSHV-infected cells with NAM or sirtinol induced transcripts and proteins of the master lytic transactivator RTA (ORF50), early lytic genes ORF57 and ORF59, and late lytic gene ORF65 and increased the production of infectious virions. NAM increased the acetylation of histones H3 and H4 as well as the level of the active histone H3 trimethyl Lys4 (H3K4me3) mark but decreased the level of the repressive histone H3 trimethyl Lys27 (H3K27me3) mark in the RTA promoter. Consistent with these results, we detected SIRT1 binding to the RTA promoter. Importantly, knockdown of SIRT1 was sufficient to increase the expression of KSHV lytic genes. Accordingly, the level of the H3K4me3 mark in the RTA promoter was increased following SIRT1 knockdown, while that of the H3K27me3 mark was decreased. Furthermore, SIRT1 interacted with RTA and inhibited RTA transactivation of its own promoter and that of its downstream target, the viral interleukin-6 gene. These results indicate that SIRT1 regulates KSHV latency by inhibiting different stages of viral lytic replication and link the cellular metabolic state with the KSHV life cycle. IMPORTANCE: Kaposi's sarcoma-associated herpesvirus (KSHV) is the causal agent of several malignancies, including Kaposi's sarcoma, commonly found in immunocompromised patients. While latent infection is required for the development of KSHV-induced malignancies, viral lytic replication also promotes disease progression. However, the mechanism controlling KSHV latent versus lytic replication remains unclear. In this study, we found that class III histone deacetylases (HDACs), also known as SIRTs, whose activities are linked to the cellular metabolic state, mediate KSHV replication. Inhibitors of SIRTs can reactivate KSHV from latency. SIRTs mediate KSHV latency by epigenetically silencing a key KSHV lytic replication activator, RTA. We found that one of the SIRTs, SIRT1, binds to the RTA promoter to mediate KSHV latency. Knockdown of SIRT1 is sufficient to induce epigenetic remodeling and KSHV lytic replication. SIRT1 also interacts with RTA and inhibits RTA's transactivation function, preventing the expression of its downstream genes. Our results indicate that SIRTs regulate KSHV latency by inhibiting different stages of viral lytic replication and link the cellular metabolic state with the KSHV life cycle.


Subject(s)
Gene Expression Regulation, Viral/drug effects , Herpesvirus 8, Human/physiology , Sirtuin 1/antagonists & inhibitors , Virus Activation/drug effects , Benzamides/pharmacology , Blotting, Western , Cell Line, Tumor , Chromatin Immunoprecipitation , DNA Primers/genetics , Gene Knockdown Techniques , Humans , Immediate-Early Proteins/metabolism , Immunoprecipitation , Luciferases , Microscopy, Confocal , Naphthols/pharmacology , Niacinamide/pharmacology , RNA, Small Interfering/genetics , Reverse Transcriptase Polymerase Chain Reaction , Sirtuin 1/genetics , Trans-Activators/metabolism , Virus Activation/physiology , Virus Latency/drug effects
13.
Cancer Res ; 72(14): 3582-92, 2012 Jul 15.
Article in English | MEDLINE | ID: mdl-22593192

ABSTRACT

EZH2 is a component of the epigenetic regulator PRC2 that suppresses gene expression. Elevated expression of EZH2 is common in human cancers and is associated with tumor progression and poor prognosis. In this study, we show that EZH2 elevation is associated with epigenetic modifications of Kaposi sarcoma-associated herpesvirus (KSHV), an oncogenic virus that promotes the development of Kaposi sarcoma and other malignancies that occur in patients with chronic HIV infections. KSHV induction of EZH2 expression was essential for KSHV-induced angiogenesis. High expression of EZH2 was observed in Kaposi sarcoma tumors. In cell culture, latent KSHV infection upregulated the expression of EZH2 in human endothelial cells through the expression of vFLIP and LANA, two KSHV-latent genes that activate the NF-κB pathway. KSHV-mediated upregulation of EZH2 was required for the induction of Ephrin-B2, an essential proangiogenic factor that drives endothelial cell tubule formation. Taken together, our findings indicate that KSHV regulates the host epigenetic modifier EZH2 to promote angiogenesis.


Subject(s)
Angiogenesis Inducing Agents/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Ephrin-B2/genetics , Herpesviridae Infections/genetics , Herpesvirus 8, Human/genetics , Neovascularization, Pathologic/genetics , Sarcoma, Kaposi/blood supply , Sarcoma, Kaposi/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Antigens, Viral/metabolism , Cell Line , Endothelial Cells/metabolism , Endothelial Cells/virology , Enhancer of Zeste Homolog 2 Protein , Gene Expression Regulation , Herpesvirus 8, Human/physiology , Humans , NF-kappa B/metabolism , Nuclear Proteins/metabolism , Polycomb Repressive Complex 2 , Sarcoma, Kaposi/virology , Up-Regulation , Viral Proteins/metabolism , Virus Latency/genetics
14.
PLoS Pathog ; 8(5): e1002703, 2012.
Article in English | MEDLINE | ID: mdl-22615563

ABSTRACT

Ubiquitination, a post-translational modification, mediates diverse cellular functions including endocytic transport of molecules. Kaposi's sarcoma-associated herpesvirus (KSHV), an enveloped herpesvirus, enters endothelial cells primarily through clathrin-mediated endocytosis. Whether ubiquitination and proteasome activity regulates KSHV entry and endocytosis remains unknown. We showed that inhibition of proteasome activity reduced KSHV entry into endothelial cells and intracellular trafficking to nuclei, thus preventing KSHV infection of the cells. Three-dimensional (3-D) analyses revealed accumulation of KSHV particles in a cytoplasmic compartment identified as EEA1+ endosomal vesicles upon proteasome inhibition. KSHV particles are colocalized with ubiquitin-binding proteins epsin and eps15. Furthermore, ubiquitination mediates internalization of both KSHV and one of its receptors integrin ß1. KSHV particles are colocalized with activated forms of the E3 ligase c-Cbl. Knock-down of c-Cbl or inhibition of its phosphorylation reduced viral entry and intracellular trafficking, resulting in decreased KSHV infectivity. These results demonstrate that ubiquitination mediates internalization of both KSHV and one of its cognate receptors integrin ß1, and identify c-Cbl as a potential E3 ligase that facilitates this process.


Subject(s)
Endosomes/virology , Herpesvirus 8, Human/pathogenicity , Human Umbilical Vein Endothelial Cells/virology , Integrin beta1/metabolism , Proteasome Endopeptidase Complex/metabolism , Virus Internalization , Adaptor Proteins, Vesicular Transport/metabolism , Cell Line , Endocytosis , Endosomes/metabolism , Gene Expression Regulation, Viral , Guanine Nucleotide Exchange Factors/metabolism , Herpesvirus 8, Human/genetics , Herpesvirus 8, Human/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Phosphorylation , Proto-Oncogene Proteins c-cbl/genetics , Proto-Oncogene Proteins c-cbl/metabolism , RNA Interference , RNA, Small Interfering , Ubiquitin-Protein Ligases/metabolism , Ubiquitination
15.
Mol Carcinog ; 51(4): 339-51, 2012 Apr.
Article in English | MEDLINE | ID: mdl-21538577

ABSTRACT

Kaposi's sarcoma-associated herpesvirus (KSHV) is the etiologic agent of Kaposi's sarcoma, an angiogenic and inflammatory endothelial cell (EC) tumor that is common in areas of high KSHV prevalence. KSHV encodes a pro-angiogenic viral chemokine receptor (vGPCR) that promotes EC growth in vitro and KS-like tumors in mouse models. vGPCR is therefore considered a viral oncogene that plays a crucial role in the pathobiology of KS. In this study, we show that focal adhesion kinase (FAK) becomes activated upon vGPCR expression in primary ECs and that FAK is required for vGPCR-mediated activation of ERK1/2, NFκB, AP-1, and vGPCR-induced migration and inhibition of anoikis. FAK is crucial to cell motility and tumor invasiveness and is a potential therapeutic target in various malignancies. Our data show that via vGPCR, KSHV has evolved a way to constitutively activate FAK signaling.


Subject(s)
Focal Adhesion Kinase 1/metabolism , Herpesvirus 8, Human/metabolism , Receptors, G-Protein-Coupled/metabolism , Animals , Anoikis , Cell Movement , Culture Media, Conditioned/chemistry , Cytoskeleton/metabolism , Endothelial Cells/cytology , Fibroblasts/metabolism , HEK293 Cells , Humans , Integrins/metabolism , Mice , Neovascularization, Pathologic , Oncogenes/genetics , Signal Transduction
16.
Regen Med ; 6(6): 733-41, 2011 Nov.
Article in English | MEDLINE | ID: mdl-22050525

ABSTRACT

Nonunion and nerve injury are the most severe and common complications of bone fracture treatments. There is still no ideal therapy for these two complications. In this report, we first applied umbilical cord mesenchymal stem cell (UC-MSC) therapy to one patient with both nonunion and nerve injury, and observed the therapeutic effects. UC-MSCs were produced and expanded according to a clinical-grade technique using serum-free medium enriched in human platelet lysate. Flow cytometry was performed to evaluate the purity of UC-MSCs, which were then intravenously injected. At 60 days postinjection, clinical examinations were performed to evaluate the therapeutic effects. Compared with before treatment, the patient's nerve reflex was present, and their muscle tone and strength increased, and x-ray and electromyography analysis further showed that the fracture gap disappeared and the nerve conduction velocity increased with shorter latency and higher amplitude. Furthermore, the clinical evolution was favorable and no side effects were observed during the 1-year follow-up. Overall, this novel treatment might open up a new strategy for the treatment of bone fracture complications.


Subject(s)
Fractures, Ununited/therapy , Humeral Fractures/complications , Humeral Fractures/therapy , Malnutrition/complications , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/cytology , Radial Nerve/injuries , Electromyography , Female , Fractures, Ununited/complications , Fractures, Ununited/physiopathology , Humans , Humeral Fractures/physiopathology , Infusions, Intravenous , Malnutrition/physiopathology , Middle Aged , Osteogenesis , Phenotype , Quality Control , Radial Nerve/physiopathology , Umbilical Cord/cytology
17.
Acta Pharmacol Sin ; 31(6): 741-5, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20523344

ABSTRACT

AIM: To investigate the antiproliferative and apoptotic effects of gemcitabine combined with gum mastic and the underlying mechanisms in human pancreatic cancer cell lines. METHODS: Cell proliferation and apoptosis were examined using the methyl thiazolyl tetrazolium (MTT) assay and propidium iodine staining, respectively. The expression of Bcl-2, Bax, NF-kappaB p65 subunit, and IkappaBalpha protein was measured using Western blotting. RESULTS: Gemcitabine 0.01-100 microg/mL inhibited cell proliferation and induced apoptosis in both pancreatic cancer BxPC-3 and COLO 357 cells. Gum mastic 40 microg/mL significantly potentiated the antiproliferative and apoptotic effects of gemcitabine 10 microg/mL after 72-h treatment. When cells were treated with gemcitabine in combination with gum mastic, the IkappaBalpha level was increased, whereas NF-kappaB activation was blocked; the expression of Bax protein was substantially increased, but Bcl-2 protein was down-regulated. CONCLUSION: Gemcitabine combined with gum mastic causes potent apoptosis in pancreatic cancer cells. The combination may be an effective therapeutic strategy for pancreatic cancer.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Deoxycytidine/analogs & derivatives , Pancreatic Neoplasms/pathology , Resins, Plant/pharmacology , Antineoplastic Agents, Phytogenic/pharmacology , Antineoplastic Agents, Phytogenic/therapeutic use , Apoptosis/drug effects , Blotting, Western , Cell Line, Tumor , Deoxycytidine/pharmacology , Flow Cytometry , Genes, bcl-2 , Growth Inhibitors/therapeutic use , Humans , I-kappa B Proteins/metabolism , Mastic Resin , NF-kappa B/metabolism , Pancreatic Neoplasms/drug therapy , Resins, Plant/therapeutic use , bcl-2-Associated X Protein/metabolism , Gemcitabine
18.
Virology ; 397(2): 379-88, 2010 Feb 20.
Article in English | MEDLINE | ID: mdl-20004456

ABSTRACT

Kaposi's sarcoma-associated herpesvirus (KSHV) is the causative agent of Kaposi's sarcoma (KS), an AIDS-related endothelial cell malignancy that is the most common cancer in central and southern Africa. The KSHV viral G protein-coupled receptor (vGPCR) is a viral oncogene that conveys a survival advantage to endothelial cells and causes KS-like tumors in mouse models. In this study we investigate the role of Shp2, a protein tyrosine phosphatase in vGPCR signaling. Shp2 is vital to many cytokine-induced signaling pathways and is dysregulated in various infections and malignancies. It has also recently been implicated in angiogenesis. We find that vGPCR activity results in phosphorylation of regulatory tyrosines in Shp2 and that in turn, Shp2 is required for vGPCR-mediated activation of MEK, NFkappaB, and AP-1. Furthermore, both genetic and chemical inhibition of Shp2 abrogate vGPCR-induced enhancement of endothelial cell migration. This establishes Shp2 as an important point of convergence of KSHV vGPCR signaling and a potential molecular target in the design of an anti-KSHV therapeutic regimen.


Subject(s)
Endothelial Cells/virology , Herpesvirus 8, Human/physiology , Host-Pathogen Interactions , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism , Receptors, Chemokine/metabolism , Signal Transduction , Cell Line , Cell Movement , Cells, Cultured , Humans , MAP Kinase Kinase Kinases/metabolism , NF-kappa B/metabolism , Phosphorylation , Transcription Factor AP-1/metabolism
19.
Prostate ; 69(15): 1643-50, 2009 Nov 01.
Article in English | MEDLINE | ID: mdl-19623543

ABSTRACT

BACKGROUND: Hypermethylation of CpG islands is a common epigenetic alteration associated with cancer. Tumor suppressor genes retinoic acid receptor beta (RARbeta) and PDLIM4 are hypermethylated and silenced in prostate cancer (PCa) tissues and PCa cell lines compared to normal prostate cells. METHODS: In this study, a benign prostate epithelial cell line RWPE1 was used as a model to study the epigenetic regulation of Myc on the RARbeta and PDLIM4 promoters. Forced Myc overexpression inhibited the RARbeta and PDLIM4 expression. RESULTS: Pyrosequencing study showed that Myc overexpression increased methylation in several CpG sites of both promoters. A DNA methylation inhibitor 5-aza-2'-deoxycytidine reversed the epigenetic alteration effect of Myc on both RARbeta and PDLIM4. CONCLUSION: The epigenetic regulation of Myc may be related to its up-regulation of the DNA methyltransferase DNMT3a and DNMT3b.


Subject(s)
DNA-Binding Proteins/genetics , Genes, myc , Prostate/physiology , Receptors, Retinoic Acid/genetics , Blotting, Western , Cell Growth Processes/genetics , Cell Line , DNA/genetics , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methylation , DNA Methyltransferase 3A , Epigenesis, Genetic , Gene Expression Profiling , Humans , LIM Domain Proteins , Male , Polymerase Chain Reaction , Prostate/enzymology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Sequence Analysis, DNA , Transfection , DNA Methyltransferase 3B
20.
Mol Nutr Food Res ; 53(7): 878-86, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19437484

ABSTRACT

This study was undertaken to investigate the mechanism by which phenethyl isothiocyanate (PEITC), a natural compound from cruciferous vegetables, exhibits antitumor effect on prostate cancer cells. Cell proliferation, cell cycle, Western blot, gene transfer, and reporter assays were used to test the effects of PEITC on the growth and IL6/JAK/STAT3 pathway in prostate cancer. The result showed that PEITC significantly inhibited DU145 cell proliferation in a dose-dependent manner and induced the cell arrest at G2-M phase. PEITC inhibited both constitutive and IL-6-induced STAT3 activity in DU145 cells. IL-6-stimulated phosphorylation of JAK2, an STAT3 upstream kinase, was also attenuated by PEITC. Moreover, an antioxidant reagent, N-acetyl-L-cysteine (NAC) which suppresses reactive oxygen species (ROS) generation, reversed the early inhibitory effects of PEITC on cell proliferation, constitutive or IL-6-mediated JAK-STAT3 phosphorylation in PCa cells. Taken together, our data demonstrated that PEITC can inhibit the activation of the JAK-STAT3 signal-cascade in prostate cancer cells and the underlying mechanism may be partially involved with blocking cellular ROS production during the early stage of the signaling activation by IL-6.


Subject(s)
Anticarcinogenic Agents/pharmacology , Isothiocyanates/pharmacology , Prostatic Neoplasms/drug therapy , STAT3 Transcription Factor/antagonists & inhibitors , Acetylcysteine/pharmacology , Cell Division/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , G2 Phase/drug effects , Humans , Interleukin-6/pharmacology , Janus Kinase 2/antagonists & inhibitors , Janus Kinase 2/metabolism , Male , Phosphorylation , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Receptors, Androgen/genetics , STAT3 Transcription Factor/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...