70 results on '"Popik W"'
Search Results
2. Inhibition of human immunodeficiency virus type 1 replication by a Tat-activated, transduced interferon gene: targeted expression to human immunodeficiency virus type 1-infected cells
- Author
-
Su, Y, primary, Popik, W, additional, and Pitha, P M, additional
- Published
- 1995
- Full Text
- View/download PDF
3. Evidence that levels of the dimeric cellular transcription factor CP2 play little role in the activation of the HIV-1 long terminal repeat in vivo or following superinfection with herpes simplex virus type 1.
- Author
-
Zhong, F., primary, Swendeman, S.L., additional, Popik, W., additional, Pitha, P.M., additional, and Sheffery, M., additional
- Published
- 1994
- Full Text
- View/download PDF
4. The presence of tat protein or tumor necrosis factor alpha is critical for herpes simplex virus type 1-induced expression of human immunodeficiency virus type 1
- Author
-
Popik, W, primary and Pitha, P M, additional
- Published
- 1994
- Full Text
- View/download PDF
5. Role of tumor necrosis factor alpha in activation and replication of the tat-defective human immunodeficiency virus type 1
- Author
-
Popik, W, primary and Pitha, P M, additional
- Published
- 1993
- Full Text
- View/download PDF
6. Inhibition by interferon of herpes simplex virus type 1-activated transcription of tat-defective provirus.
- Author
-
Popik, W, primary and Pitha, P M, additional
- Published
- 1991
- Full Text
- View/download PDF
7. Transforming Growth Factor-μ Inhibits the Antiviral Action of Interferons in Human Embryonic Fibroblasts
- Author
-
ZIELIŃSKA-JENCZYLIK, J., primary, INGLOT, A.D., additional, INGL0T, O., additional, and POPIK, W., additional
- Published
- 1991
- Full Text
- View/download PDF
8. Binding of human immunodeficiency virus type 1 to CD4 induces association of Lck and Raf-1 and activates Raf-1 by a Ras-independent pathway
- Author
-
Popik, W and Pitha, P M
- Abstract
We have analyzed CD4-mediated signaling during the early stages of human immunodeficiency virus type 1 (HIV-1) infection. Binding of purified HIV-1 virions or recombinant HIV-1 glycoprotein gp120 to CD4 receptors resulted in association and tyrosine phosphorylation and activation of tyrosine kinase Lck and serine/threonine kinase Raf-1. The association between Lck and Raf-1 was mediated by stimulation of the CD4 receptors, since it was abolished by preincubation of the virus with soluble CD4 and was not detected in CD4-negative A201 T cells. However, the Lck-Raf-1 association was restored in A201 cells permanently transfected with human CD4 cDNA and stimulated with anti-CD4 antibodies. In addition, a catalytically active Lck was required for the association of Lck and Raf-1. Surprisingly, the CD4-mediated signaling, induced by the HIV-1 binding, did not result in stimulation of the Ras GTP-binding activity or its association with Raf-1, indicating that the signaling pathway generated by the HIV-1 binding is not identical to the classical Ras/Raf-1 pathway. Furthermore, overexpression of activated Raf-1 in Jurkat T cells stimulated the HIV long terminal repeat promoter activity and significantly enhanced HIV-1 replication. This suggests that the Lck-Raf-1 pathway, rapidly stimulated by the binding of HIV-1 or gp120 to CD4 receptors, may play an essential role in the transcriptional activation of the integrated HIV-1 provirus as well as in its pathogenicity.
- Published
- 1996
- Full Text
- View/download PDF
9. A few distinct ?molecular sandwiches? are basis for structural and functional similarities of subspecies of interferon ? and of families of growth-promoting hormones
- Author
-
Inglot, A. D., primary, Popik, W., additional, Piasecki, E., additional, and Czyrski, J., additional
- Published
- 1986
- Full Text
- View/download PDF
10. Effect of tumor necrosis factor-alpha and herpes simplex virus type 1 on the Tat-targeted inhibition of human immunodeficiency virus type 1 replication
- Author
-
Popik, W. and Pitha, P.M.
- Subjects
HIV (Viruses) -- Reproduction ,Tumor necrosis factor -- Physiological aspects ,Herpes simplex virus -- Physiological aspects - Abstract
SOURCE: Virology, August 1, 1994;202(2):521-529. According to the authors' abstract of an article published in Virology, "In this study, we have examined whether the Tat antagonist can inhibit human immunodeficiency [...]
- Published
- 1994
11. The effect of interferons at the early stage of HIV replication
- Author
-
Popik, W., Rodrigues-Ortega, M., Bednarik, D.P., and Pitha, P.M.
- Subjects
HIV (Viruses) -- Reproduction ,Interferon alpha -- Physiological aspects ,Business ,Health care industry - Abstract
AUTHORS: W. Popik, M. Rodrigues-Ortega, D.P. Bednarik, and P.M. Pitha. The Johns Hopkins University Oncology Center, Baltimore, Md. According to the authors' abstract of a presentation to the 81st annual [...]
- Published
- 1990
12. APOL1 Modulates Renin-Angiotensin System.
- Author
-
Kumar V, Kaur P, Ayasolla K, Jha A, Wiqas A, Vashistha H, Saleem MA, Popik W, Malhotra A, Gebeshuber CA, Skorecki K, and Singhal PC
- Subjects
- Humans, Animals, Mice, MicroRNAs genetics, MicroRNAs metabolism, Receptor, Angiotensin, Type 1 metabolism, Receptor, Angiotensin, Type 1 genetics, Angiotensinogen metabolism, Angiotensinogen genetics, Glomerulosclerosis, Focal Segmental metabolism, Glomerulosclerosis, Focal Segmental genetics, Glomerulosclerosis, Focal Segmental pathology, Podocytes metabolism, Apolipoprotein L1 genetics, Apolipoprotein L1 metabolism, Renin-Angiotensin System genetics, Receptors, Calcitriol metabolism, Receptors, Calcitriol genetics, Renin metabolism, Renin genetics
- Abstract
Patients carrying APOL1 risk alleles (G1 and G2) have a higher risk of developing Focal Segmental Glomerulosclerosis (FSGS); we hypothesized that escalated levels of miR193a contribute to kidney injury by activating renin-angiotensin system (RAS) in the APOL1 milieus. Differentiated podocytes (DPDs) stably expressing vector (V/DPD), G0 (G0/DPDs), G1 (G1/DPDs), and G2 (G2/DPDs) were evaluated for renin, Vitamin D receptor (VDR), and podocyte molecular markers (PDMMs, including WT1, Podocalyxin, Nephrin, and Cluster of Differentiation [CD]2 associated protein [AP]). G0/DPDs displayed attenuated renin but an enhanced expression of VDR and Wilms Tumor [WT]1, including other PDMMs; in contrast, G1/DPDs and G2/DPDs exhibited enhanced expression of renin but decreased expression of VDR and WT1, as well as other PDMMs (at both the protein and mRNA levels). G1/DPDs and G2/DPDs also showed increased mRNA expression for Angiotensinogen and Angiotensin II Type 1 (AT1R) and 2 (AT2R) receptors. Protein concentrations of Brain Acid-Soluble Protein [BASP]1, Enhancer of Zeste Homolog [EZH]2, Histone Deacetylase [HDAC]1, and Histone 3 Lysine27 trimethylated [H3K27me3] in WT1-IP (immunoprecipitated proteins with WT1 antibody) fractions were significantly higher in G0/DPDs vs. G1/DPD and G2/DPDs. Moreover, DPD-silenced BASP1 displayed an increased expression of renin. Notably, VDR agonist-treated DPDs showed escalated levels of VDR and a higher expression of PDMMs, but an attenuated expression of renin. Human Embryonic Kidney (HEK) cells transfected with increasing APOL1(G0) plasmid concentrations showed a corresponding reduction in renin mRNA expression. Bioinformatics studies predicted the miR193a target sites in the VDR 3'UTR (untranslated region), and the luciferase assay confirmed the predicted sites. As expected, podocytes transfected with miR193a plasmid displayed a reduced VDR and an enhanced expression of renin. Renal cortical section immunolabeling in miR193a transgenic (Tr) mice showed renin-expressing podocytes. Kidney tissue extracts from miR193aTr mice also showed reduced expression of VDR and PDMMs, but enhanced expression of Renin. Blood Ang II levels were higher in miR193aTr, APOLG1, and APOL1G1/G2 mice when compared to control mice. Based on these findings, miR193a regulates the activation of RAS and podocyte molecular markers through modulation of VDR and WT1 in the APOL1 milieu.
- Published
- 2024
- Full Text
- View/download PDF
13. IFI16 Is Indispensable for Promoting HIF-1α-Mediated APOL1 Expression in Human Podocytes under Hypoxic Conditions.
- Author
-
Randle RK, Amara VR, and Popik W
- Subjects
- Humans, Apolipoprotein L1 genetics, Apolipoprotein L1 metabolism, Cell Hypoxia genetics, Chromatin Immunoprecipitation, Hypoxia metabolism, Hypoxia-Inducible Factor 1, alpha Subunit genetics, Hypoxia-Inducible Factor 1, alpha Subunit metabolism, Nuclear Proteins genetics, Nuclear Proteins metabolism, Phosphoproteins genetics, Phosphoproteins metabolism, Podocytes metabolism, Renal Insufficiency, Chronic metabolism
- Abstract
Genetic variants in the protein-coding regions of APOL1 are associated with an increased risk and progression of chronic kidney disease (CKD) in African Americans. Hypoxia exacerbates CKD progression by stabilizing HIF-1α, which induces APOL1 transcription in kidney podocytes. However, the contribution of additional mediators to regulating APOL1 expression under hypoxia in podocytes is unknown. Here, we report that a transient accumulation of HIF-1α in hypoxia is sufficient to upregulate APOL1 expression in podocytes through a cGAS/STING/IRF3-independent pathway. Notably, IFI16 ablation impedes hypoxia-driven APOL1 expression despite the nuclear accumulation of HIF-1α. Co-immunoprecipitation assays indicate no direct interaction between IFI16 and HIF-1α. Our studies identify hypoxia response elements (HREs) in the APOL1 gene enhancer/promoter region, showing increased HIF-1α binding to HREs located in the APOL1 gene enhancer. Luciferase reporter assays confirm the role of these HREs in transcriptional activation. Chromatin immunoprecipitation (ChIP)-qPCR assays demonstrate that IFI16 is not recruited to HREs, and IFI16 deletion reduces HIF-1α binding to APOL1 HREs. RT-qPCR analysis indicates that IFI16 selectively affects APOL1 expression, with a negligible impact on other hypoxia-responsive genes in podocytes. These findings highlight the unique contribution of IFI16 to hypoxia-driven APOL1 gene expression and suggest alternative IFI16-dependent mechanisms regulating APOL1 gene expression under hypoxic conditions.
- Published
- 2024
- Full Text
- View/download PDF
14. Endoplasmic reticulum-translocation is essential for APOL1 cellular toxicity.
- Author
-
Kruzel-Davila E, Bavli-Kertselli I, Ofir A, Cheatham AM, Shemer R, Zaknoun E, Chornyy S, Tabachnikov O, Davis SE, Khatua AK, Skorecki K, and Popik W
- Abstract
Two variants at the APOL1 gene, encoding apolipoprotein L1, account for more than 70% of the increased risk for chronic kidney disease in individuals of African ancestry. While the initiating event for APOL1 risk variant cell injury remains to be clarified, we explored the possibility of blocking APOL1 toxicity at a more upstream level. We demonstrate that deletion of the first six amino acids of exon 4 abrogates APOL1 cytotoxicity by impairing APOL1 translocation to the lumen of ER and splicing of the signal peptide. Likewise, in orthologous systems, APOL1 lethality was partially abrogated in yeast strains and flies with reduced dosage of genes encoding ER translocon proteins. An inhibitor of ER to Golgi trafficking reduced lethality as well. We suggest that targeting the MSALFL sequence or exon 4 skipping may serve as potential therapeutic approaches to mitigate the risk of CKD caused by APOL1 renal risk variants., Competing Interests: KS and RS are inventors on Patent No.: US 10,927,414 B2. KS is an Associate Editor for The Kidney, 11th Edition, Elsevier 2020., (© 2022 The Authors.)
- Published
- 2021
- Full Text
- View/download PDF
15. Nucleosomal dsDNA Stimulates APOL1 Expression in Human Cultured Podocytes by Activating the cGAS/IFI16-STING Signaling Pathway.
- Author
-
Davis SE, Khatua AK, and Popik W
- Subjects
- Apolipoprotein L1 genetics, Cell Line, Transformed, Humans, Apolipoprotein L1 metabolism, DNA metabolism, Membrane Proteins metabolism, Nuclear Proteins metabolism, Nucleosomes metabolism, Nucleotidyltransferases metabolism, Phosphoproteins metabolism, Podocytes metabolism, Signal Transduction
- Abstract
APOL1 alleles G1 and G2 are associated with faster progression to lupus nephritis (LN)-associated end-stage renal disease (LN-ESRD) in African Americans. Increased levels of type I interferons (IFNs) and nucleosome-associated double-stranded DNA (dsDNA) fragments (nsDNA) are the hallmark of this disease. Here, we identify cyclic GMP-AMP synthase (cGAS) and interferon-inducible protein 16 (IFI16) as the major DNA sensors in human immortalized podocytes. We also show that nsDNA triggers the expression of APOL1 and IFNβ via IRF3 activation through the cGAS/IFI16-STING pathway. We demonstrate that maximal APOL1 expression also requires the activation of type I IFN receptor (IFNAR) and STAT1 signaling triggered by IFNβ produced in response to nsDNA, or by exogenous IFNβ. Finally, we show that STAT1 activation is sufficient to upregulate IFI16, subsequently boosting APOL1 expression through a positive feedback mechanism. Collectively, we find that nsDNA-induced APOL1 expression is mediated by both IFNβ-independent and dependent signaling pathways triggered by activation of the cGAS/IFI16-STING pathway. We propose that simultaneous inhibition of STING and the IFNAR-STAT1 pathway may attenuate IFI16 expression, reduce IFI16-cGAS cross-talk, and prevent excessive APOL1 expression in human podocytes in response to nsDNA.
- Published
- 2019
- Full Text
- View/download PDF
16. BK Virus Replication in the Glomerular Vascular Unit: Implications for BK Virus Associated Nephropathy.
- Author
-
Popik W, Khatua AK, Fabre NF, Hildreth JEK, and Alcendor DJ
- Subjects
- BK Virus genetics, Cytokines genetics, Cytokines immunology, Humans, Kidney Diseases genetics, Kidney Diseases immunology, Kidney Glomerulus immunology, Kidney Transplantation adverse effects, Polyomavirus Infections genetics, Polyomavirus Infections immunology, Postoperative Complications etiology, Postoperative Complications genetics, Postoperative Complications immunology, Virus Activation, Virus Replication, BK Virus physiology, Kidney Diseases virology, Kidney Glomerulus virology, Polyomavirus Infections virology, Postoperative Complications virology
- Abstract
Background: BK polyomavirus (BKV) reactivates from latency after immunosuppression in renal transplant patients, resulting in BKV-associated nephropathy (BKVAN). BKVAN has emerged as an important cause of graft dysfunction and graft loss among transplant patients. BKV infection in kidney transplant patients has increased over recent decades which correlates with the use of more potent immunosuppressive therapies. BKV infection of the Glomerular Vascular Unit (GVU) consisting of podocytes, mesangial cells, and glomerular endothelial cells could lead to glomerular inflammation and contribute to renal fibrosis. The effects of BKV on GVU infectivity have not been reported., Methods: We infected GVU cells with the Dunlop strain of BKV. Viral infectivity was analyzed by microscopy, immunofluorescence, Western blot analysis, and quantitative RT-PCR (qRT-PCR). The expression of specific proinflammatory cytokines induced by BKV was analyzed by qRT-PCR., Results: BKV infection of podocytes, mesangial cells, and glomerular endothelial cells was confirmed by qRT-PCR and positive staining with antibodies to the BKV VP1 major capsid protein, or the SV40 Large T-Antigen. The increased transcriptional expression of interferon gamma-induced protein 10 (CXCL10/IP-10) and interferon beta (IFNβ) was detected in podocytes and mesangial cells at 96 h post-infection., Conclusions: All cellular components of the GVU are permissive for BKV replication. Cytopathic effects induced by BKV in podocytes and glomerular endothelial cells and the expression of CXCL10 and IFNβ genes by podocytes and mesangial cells may together contribute to glomerular inflammation and cytopathology in BKVAN.
- Published
- 2019
- Full Text
- View/download PDF
17. Mesangial cells, specialized renal pericytes and cytomegalovirus infectivity: Implications for HCMV pathology in the glomerular vascular unit and post-transplant renal disease.
- Author
-
Popik W, Correa H, Khatua A, Aronoff DM, and Alcendor DJ
- Abstract
Background: Human Cytomegalovirus (HCMV) infection is problematic after kidney transplantation. Human mesangial cells along with human glomerular endothelial cells and podocytes constitute the renal glomerular vascular unit (GVU). HCMV infection of the GVU is poorly understood., Methods: GVU cells infectivity was analysed by microscopy and immunofluorescence. Cytokines profiles were measured by Luminex assays. Renal tissue analysis for HCMV infection was performed by immunohistochemistry., Results: Mesangial cells and glomerular endothelial cells but not podocytes were permissive for both lab adapted and clinical strains of HCMV. Luminex analysis of cytokines expressed by mesangial cells exposed to the SBCMV clinical strain was examined. A Tricell infection model of the GVU maintains >90% viability with a unique cytokine profile. Finally, we show αSMA stained mesangial cells permissive for HCMV in renal tissue from a transplant patient., Conclusions: HCMV infection of mesangial cells induces angiogenic and proinflammatory cytokines that could contribute to glomerular inflammation., Competing Interests: Competing interests The authors declare that they have no competing interests.
- Published
- 2019
- Full Text
- View/download PDF
18. Role of Apolipoprotein L1 in Human Parietal Epithelial Cell Transition.
- Author
-
Kumar V, Vashistha H, Lan X, Chandel N, Ayasolla K, Shoshtari SSM, Aslam R, Paliwal N, Abbruscato F, Mikulak J, Popik W, Atta MG, Chander PN, Malhotra A, Meyer-Schwesinger C, Skorecki K, and Singhal PC
- Subjects
- AIDS-Associated Nephropathy metabolism, AIDS-Associated Nephropathy virology, Animals, Apolipoprotein L1 genetics, Case-Control Studies, Epithelial Cells metabolism, HEK293 Cells, Hep G2 Cells, Humans, Kidney Glomerulus metabolism, Mice, Mice, Transgenic, AIDS-Associated Nephropathy pathology, Apolipoprotein L1 metabolism, Epithelial Cells pathology, Gene Expression Regulation, Kidney Glomerulus pathology, MicroRNAs genetics
- Abstract
Human parietal epithelial cells (PECs) are progenitor cells that sustain podocyte homeostasis. We hypothesized that the lack of apolipoprotein (APO) L1 ensures the PEC phenotype, but its induction initiates PEC transition (expression of podocyte markers). APOL1 expression and down-regulation of miR193a coincided with the expression of podocyte markers during the transition. The induction of APOL1 also stimulated transition markers in human embryonic kidney cells (cells with undetectable APOL1 protein expression). APOL1 silencing in PECs up-regulated miR193a expression, suggesting the possibility of a reciprocal feedback relationship between APOL1 and miR193a. HIV, interferon-γ, and vitamin D receptor agonist down-regulated miR193a expression and induced APOL1 expression along with transition markers in PECs. Luciferase assay suggested a putative interaction between miR193a and APOL1. Since silencing of APOL1 attenuated HIV-, vitamin D receptor agonist-, miR193a inhibitor-, and interferon-γ-induced expression of transition markers, APOL1 appears to be a critical functional constituent of the miR193a- APOL1 axis in PECs. This notion was confirmed by further enhanced expression of PEC markers in APOL1 mRNA-silenced PECs. In vivo studies, glomeruli in patients with HIV, and HIV/APOL1 transgenic mice had foci of PECs expressing synaptopodin, a transition marker. APOL1 likely regulates PEC molecular phenotype through modulation of miR193a expression, and APOL1 and miR193a share a reciprocal feedback relationship., (Copyright © 2018 American Society for Investigative Pathology. Published by Elsevier Inc. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
19. Blocking the 5' splice site of exon 4 by a morpholino oligomer triggers APOL1 protein isoform switch.
- Author
-
Cheatham AM, Davis SE, Khatua AK, and Popik W
- Subjects
- Apolipoprotein L1 genetics, HEK293 Cells, Heterogeneous Nuclear Ribonucleoprotein A1 genetics, Heterogeneous Nuclear Ribonucleoprotein A1 metabolism, Humans, Kidney Diseases genetics, Kidney Diseases metabolism, Kidney Diseases pathology, Podocytes pathology, Protein Isoforms biosynthesis, Protein Isoforms genetics, Alternative Splicing, Apolipoprotein L1 biosynthesis, Exons, Morpholinos pharmacology, Podocytes metabolism, RNA Splice Sites
- Abstract
APOL1 risk alleles G1 or G2 are associated with a kidney disease phenotype exclusively in people of recent African ancestry. Here we show that exon 4 encoding a part of the APOL1 signal peptide is constitutively spliced in major APOL1 transcripts expressed in kidney glomerular and tubular cells. We demonstrate that constitutive splicing of exon 4 results from a suboptimal hnRNP A1 binding motif found in exon 4. Accordingly, a robust binding of hnRNP A1 protein to a consensus hnRNP A1 cis-acting element in exon 4 results in almost complete exclusion of exon 4 from the APOL1 minigene transcripts. Blocking the 5' splice site at the exon 4/intron boundary with a specific antisense morpholino oligonucleotide excludes exon 4 from the splicing pattern of endogenous APOL1 transcripts. These transcripts are fully functional and produce APOL1 protein isoform that is not normally detectable in podocytes. Together with our previous data showing no cytotoxicity of overexpressed APOL1 isoform lacking exon 4, we propose that morpholino-induced APOL1 isoform switch may provide a new tool to identify in vivo molecular mechanism(s) by which risk alleles promote or mediate the kidney disease phenotype.
- Published
- 2018
- Full Text
- View/download PDF
20. Phosphorodiamidate morpholino targeting the 5' untranslated region of the ZIKV RNA inhibits virus replication.
- Author
-
Popik W, Khatua A, Hildreth JEK, Lee B, and Alcendor DJ
- Subjects
- Chemokine CCL5 genetics, Cytokines genetics, Fluorescent Antibody Technique, Humans, Interferon-beta genetics, Podocytes virology, Real-Time Polymerase Chain Reaction, Zika Virus genetics, Zika Virus physiology, 5' Untranslated Regions drug effects, Morpholinos pharmacology, Virus Replication drug effects, Zika Virus drug effects
- Abstract
Background: Zika virus (ZIKV) infection has been associated with microcephaly in infants. Currently there is no treatment or vaccine. Here we explore the use of a morpholino oligonucleotide targeted to the 5' untranslated region (5'-UTR) of the ZIKV RNA to prevent ZIKV replication., Methods: Morpholino DWK-1 inhibition of ZIKV replication in human glomerular podocytes was examined by qRT-PCR, reduction in ZIKV genome copy number, western blot analysis, immunofluorescence and proinflammatory cytokine gene expression., Results: Podocytes pretreated with DWK-1 showed reduced levels of both viral mRNA and ZIKV E protein expression compared to controls. We observed suppression in proinflammatory gene expression for IFN-β (interferon β) RANTES (regulated on activation, normal T cell expressed and secreted), MIP-1α (macrophage inflammatory protein-1α), TNF-α (tumor necrosis factor-α) and IL1-α (interleukin 1-α) in ZIKV-infected podocytes pretreated with DWK-1., Conclusions: Morpholino DWK-1 targeting the ZIKV 5'-UTR effectively inhibits ZIKV replication and suppresses ZIKV-induced proinflammatory gene expression., (Copyright © 2018 Elsevier Inc. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
21. Modulation of apolipoprotein L1-microRNA-193a axis prevents podocyte dedifferentiation in high-glucose milieu.
- Author
-
Mishra A, Ayasolla K, Kumar V, Lan X, Vashistha H, Aslam R, Hussain A, Chowdhary S, Marashi Shoshtari S, Paliwal N, Popik W, Saleem MA, Malhotra A, Meggs LG, Skorecki K, and Singhal PC
- Subjects
- Apolipoprotein L1 genetics, Calcitriol analogs & derivatives, Calcitriol pharmacology, Cell Line, Transformed, DNA (Cytosine-5-)-Methyltransferase 1 genetics, DNA (Cytosine-5-)-Methyltransferase 1 metabolism, Enhancer of Zeste Homolog 2 Protein genetics, Enhancer of Zeste Homolog 2 Protein metabolism, Gene Expression Regulation drug effects, Humans, MicroRNAs genetics, PAX2 Transcription Factor genetics, PAX2 Transcription Factor metabolism, Phenotype, Podocytes metabolism, Podocytes pathology, Polycomb-Group Proteins genetics, Polycomb-Group Proteins metabolism, Receptors, Calcitriol agonists, Receptors, Calcitriol metabolism, Signal Transduction drug effects, WT1 Proteins genetics, WT1 Proteins metabolism, Apolipoprotein L1 metabolism, Cell Dedifferentiation drug effects, Glucose toxicity, MicroRNAs metabolism, Podocytes drug effects
- Abstract
The loss of podocyte (PD) molecular phenotype is an important feature of diabetic podocytopathy. We hypothesized that high glucose (HG) induces dedifferentiation in differentiated podocytes (DPDs) through alterations in the apolipoprotein (APO) L1-microRNA (miR) 193a axis. HG-induced DPD dedifferentiation manifested in the form of downregulation of Wilms' tumor 1 (WT1) and upregulation of paired box 2 (PAX2) expression. WT1-silenced DPDs displayed enhanced expression of PAX2. Immunoprecipitation of DPD cellular lysates with anti-WT1 antibody revealed formation of WT1 repressor complexes containing Polycomb group proteins, enhancer of zeste homolog 2, menin, and DNA methyltransferase (DNMT1), whereas silencing of either WT1 or DNMT1 disrupted this complex with enhanced expression of PAX2. HG-induced DPD dedifferentiation was associated with a higher expression of miR193a, whereas inhibition of miR193a prevented DPD dedifferentiation in HG milieu. HG downregulated DPD expression of APOL1. miR193a-overexpressing DPDs displayed downregulation of APOL1 and enhanced expression of dedifferentiating markers; conversely, silencing of miR193a enhanced the expression of APOL1 and preserved DPD phenotype. Moreover, stably APOL1G0-overexpressing DPDs displayed the enhanced expression of WT1 but attenuated expression of miR193a; nonetheless, silencing of APOL1 reversed these effects. Since silencing of APOL1 enhanced miR193a expression as well as dedifferentiation in DPDs, it appears that downregulation of APOL1 contributed to dedifferentiation of DPDs through enhanced miR193a expression in HG milieu. Vitamin D receptor agonist downregulated miR193a, upregulated APOL1 expression, and prevented dedifferentiation of DPDs in HG milieu. These findings suggest that modulation of the APOL1-miR193a axis carries a potential to preserve DPD molecular phenotype in HG milieu.
- Published
- 2018
- Full Text
- View/download PDF
22. Effect of APOL1 disease risk variants on APOL1 gene product.
- Author
-
Haque S, Patil G, Mishra A, Lan X, Popik W, Malhotra A, Skorecki K, and Singhal PC
- Subjects
- Amino Acid Substitution, Cell Differentiation, Cell Line, Cell Proliferation, Endoplasmic Reticulum Chaperone BiP, Gene Deletion, Gene Expression Regulation, HEK293 Cells, Heat-Shock Proteins genetics, Humans, Podocytes cytology, Podocytes metabolism, Protein Biosynthesis, RNA, Messenger genetics, Transcription, Genetic, Apolipoprotein L1 genetics, Genetic Variation, Kidney Diseases genetics
- Abstract
Gene sequence mutations may alter mRNA transcription, transcript stability, protein translation, protein stability and protein folding. Apolipoprotein L1 (APOL1) has two sets of sequence variants that are risk factors for kidney disease development, APOL1G1 (substitution mutation) and APOL1G2 (deletion mutation). Our present study focuses on the impact of these variants on APOL1 mRNA transcription and translation. APOL1 plasmids (EV, G0, G1 and G2) were transfected into human embryonic kidney (HEK) 293T cells. APOL1 variant expression was observed to be significantly lower than that of APOL1G0. Podocyte cell lines stably expressing APOL1 transgenes also showed lower levels of APOL1 expression of APOL1 variants (G1 and G2) compared with APOL1G0 by Western blotting and FACS analysis. The enhanced expression of GRP78 by podocytes expressing APOL1 variants would indicate endoplasmic reticulum (ER) stress. Bioinformatics evaluation using two different programs (MUPro and I-Mutant 2.0) predicted that APOL1 variants were less stable than APOL1G0. Concomitant with protein levels, APOL1 mRNA levels were also depressed following induction of APOL1 variant compared with APOL1G0 in both proliferating and differentiated podocytes. APOL1 mRNA transcript stability was tested after actinomycin D pulsing; APOL1G1 and APOL1G2 mRNAs transcript decayed 10-15% and 15-20% (within a period of 0.5-3 h) respectively. Our data suggest that down-regulated APOL1 protein expression in APOL1 variants is due to compromised transcription and decay of the APOL1 variant transcripts., (© 2017 The Author(s).)
- Published
- 2017
- Full Text
- View/download PDF
23. HIV Promotes NLRP3 Inflammasome Complex Activation in Murine HIV-Associated Nephropathy.
- Author
-
Haque S, Lan X, Wen H, Lederman R, Chawla A, Attia M, Bongu RP, Husain M, Mikulak J, Saleem MA, Popik W, Malhotra A, Chander PN, and Singhal PC
- Subjects
- Animals, Apoptosis physiology, Mice, NLR Family, Pyrin Domain-Containing 3 Protein, Podocytes virology, AIDS-Associated Nephropathy metabolism, Apoptosis Regulatory Proteins metabolism, Carrier Proteins metabolism, Inflammasomes metabolism, Podocytes metabolism, Reactive Oxygen Species metabolism
- Abstract
Dysregulated growth and loss of podocytes are important features of HIV-associated nephropathy. Recently, HIV was reported to induce a new type of programed cell death, pyroptosis, in T lymphocytes through induction of Nod-like receptor protein 3 (NLRP3) inflammasome complexes. We evaluated the role of HIV in podocyte NLRP3 inflammasome formation both in vivo and in vitro. Renal cortical sections of HIV-transgenic mice (Tg26) displayed increased expression of NLRP3, ASC (a CARD protein), caspase-1, and IL-1β proteins, confirming NLRP3 inflammasome complex formation in podocytes of Tg26 mice. Renal tissues of Tg26 mice also displayed enhanced mRNA levels and protein expressions of inflammasome markers (NLRP3, ASC, and caspase-1, and IL-1β). Serum of Tg26 mice also showed elevated concentrations of IL-1β cytokine compared with FVBN mice. HIV induced pyroptosis in a dose- and time-dependent manner within podocytes, a phenotype of inflammasome activation. Caspase-1 inhibitor not only attenuated podocyte expression of caspase-1 and IL-1β but also provided protection against pyroptosis, suggesting that HIV-induced podocyte injury was mediated by caspase-1 activation. Interestingly, HIV-induced podocyte pyroptosis could be partially inhibited by Tempol (a superoxide dismutase-mimetic agent) and by glyburide (an inhibitor of potassium efflux). These findings suggest that generation of reactive oxygen species and potassium efflux contribute to HIV-induced pyroptosis and NLRP3 inflammasome activation in podocytes., (Copyright © 2016 American Society for Investigative Pathology. Published by Elsevier Inc. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
24. Protein domains of APOL1 and its risk variants.
- Author
-
Lan X, Wen H, Lederman R, Malhotra A, Mikulak J, Popik W, Skorecki K, and Singhal PC
- Subjects
- Black or African American genetics, Apolipoprotein L1, Apolipoproteins metabolism, Genotype, HEK293 Cells, Humans, Lipoproteins, HDL metabolism, Protein Structure, Tertiary, Risk Factors, Apolipoproteins genetics, Genetic Variation, Kidney Diseases genetics, Lipoproteins, HDL genetics
- Abstract
Increasing lines of evidence have demonstrated that the development of higher rates of non-diabetic glomerulosclerosis (GS) in African Americans can be attributed to two coding sequence variants (G1 and G2) in the Apolipoprotein L1 (APOL) gene. Recent studies indicate that the gene products of these APOL1 risk variants have augmented toxicity to kidney cells. However, the biological characteristics of APOL1 and its risk variants are not well elucidated. The APOL1 protein can be divided into several functional domains, including signal peptide (SP), pore forming domain (PFD), membrane address domain (MAD), and SRA-interacting domain. To investigate the relative contribution of each domain to cell injury, we constructed a serial expression vectors to delete or express each domain. These vectors were transfected into the human embryonic kidney cell line 293T, and then compared the cytotoxicity. In addition, we conducted studies in which APOL1 wild type (G0) was co-transfected in combination with G1 or G2 to see whether G0 could counteract the toxicity of the risk variants. The results showed that deleting the SP did not abolish the toxicity of APOL1, though deletion of 26 amino acid residues of the mature peptide at the N-terminal partially decreased the toxicity. Deleting PFD or MAD or SRA-interacting domain abolished toxicity, while, overexpressing each domain alone could not cause toxicity to the host cells. Deletion of the G2 sites while retaining G1 sites in the risk state resulted in persistent toxicity. Either deletion or exchanging the BH3 domain in the PFD led to complete loss of the toxicity in this experimental platform. Adding G0 to either G1 or G2 did not attenuate the toxicity of the either moiety. These results indicate that the integrity of the mature APOL1 protein is indispensable for its toxicity. Our study not only reveals the contribution of each domain of the APOL1 protein to cell injury, but also highlights some potential suggested targets for drug design to prevent or treat APOL1-associated nephropathy., (Copyright © 2015 Elsevier Inc. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
25. Exon 4-encoded sequence is a major determinant of cytotoxicity of apolipoprotein L1.
- Author
-
Khatua AK, Cheatham AM, Kruzel ED, Singhal PC, Skorecki K, and Popik W
- Subjects
- Active Transport, Cell Nucleus, Alternative Splicing, Amino Acid Sequence, Apolipoprotein L1, Apolipoproteins chemistry, Base Sequence, Basic Helix-Loop-Helix Leucine Zipper Transcription Factors genetics, Basic Helix-Loop-Helix Leucine Zipper Transcription Factors metabolism, Gene Expression Regulation, HEK293 Cells, Humans, Lipoproteins, HDL chemistry, Molecular Sequence Data, Podocytes pathology, RNA Interference, RNA, Messenger metabolism, Signal Transduction, Structure-Activity Relationship, Transcription, Genetic, Transfection, Apolipoproteins genetics, Apolipoproteins metabolism, Autophagy, Exons, Lipoproteins, HDL genetics, Lipoproteins, HDL metabolism, Podocytes metabolism
- Abstract
The apolipoprotein L1 (APOL1) gene (APOL1) product is toxic to kidney cells, and its G1 and G2 alleles are strongly associated with increased risk for kidney disease progression in African Americans. Variable penetrance of the G1 and G2 risk alleles highlights the significance of additional factors that trigger or modify the progression of disease. In this regard, the effect of alternative splicing in the absence or presence of G1 or G2 alleles is unknown. In this study we investigated whether alternative splicing of non-G1, non-G2 APOL1 (APOL1 G0) affects its biological activity. Among seven APOL1 exons, exons 2 and 4 are differentially expressed in major transcripts. We found that, in contrast to APOL1 splice variants B3 or C, variants A and B1 demonstrate strong toxicity in human embryonic kidney (HEK293T) cells. Subsequently, we established that exon 4 is a major determinant of toxicity of variants A and B1 and that extracellular release of these variants is dispensable for their cytotoxicity. Although only variants A and B1 induced nuclear translocation of transcription factor EB (TFEB), a master regulator of lysosomal biogenesis and autophagy, exon 4-positive and -negative APOL1 variants stimulated perinuclear accumulation of unprocessed autophagosomes. Knockdown of endogenous TFEB did not attenuate APOL1 cytotoxicity, indicating that nuclear translocation of TFEB is dispensable for APOL1 toxicity. Our findings that a human podocyte cell line expresses exon 4-positive and -negative APOL1 transcripts suggest that these variants may play a differential role in podocyte pathology. In summary, we have identified exon 4 as a major determinant of APOL1 G0 cytotoxicity., (Copyright © 2015 the American Physiological Society.)
- Published
- 2015
- Full Text
- View/download PDF
26. Phospholipase D1 Couples CD4+ T Cell Activation to c-Myc-Dependent Deoxyribonucleotide Pool Expansion and HIV-1 Replication.
- Author
-
Taylor HE, Simmons GE Jr, Mathews TP, Khatua AK, Popik W, Lindsley CW, D'Aquila RT, and Brown HA
- Subjects
- Apoptosis, Blotting, Western, CD4-Positive T-Lymphocytes metabolism, Cell Proliferation, Cells, Cultured, DNA Replication, HIV Infections immunology, HIV Infections metabolism, Humans, Lymphocyte Activation, RNA, Messenger genetics, Real-Time Polymerase Chain Reaction, Reverse Transcriptase Polymerase Chain Reaction, CD4-Positive T-Lymphocytes virology, Deoxyribonucleotides metabolism, HIV Infections virology, HIV-1 physiology, Phospholipase D metabolism, Proto-Oncogene Proteins c-myc metabolism, Virus Replication
- Abstract
Quiescent CD4+ T cells restrict human immunodeficiency virus type 1 (HIV-1) infection at early steps of virus replication. Low levels of both deoxyribonucleotide triphosphates (dNTPs) and the biosynthetic enzymes required for their de novo synthesis provide one barrier to infection. CD4+ T cell activation induces metabolic reprogramming that reverses this block and facilitates HIV-1 replication. Here, we show that phospholipase D1 (PLD1) links T cell activation signals to increased HIV-1 permissivity by triggering a c-Myc-dependent transcriptional program that coordinates glucose uptake and nucleotide biosynthesis. Decreasing PLD1 activity pharmacologically or by RNA interference diminished c-Myc-dependent expression during T cell activation at the RNA and protein levels. PLD1 inhibition of HIV-1 infection was partially rescued by adding exogenous deoxyribonucleosides that bypass the need for de novo dNTP synthesis. Moreover, the data indicate that low dNTP levels that impact HIV-1 restriction involve decreased synthesis, and not only increased catabolism of these nucleotides. These findings uncover a unique mechanism of action for PLD1 inhibitors and support their further development as part of a therapeutic combination for HIV-1 and other viral infections dependent on host nucleotide biosynthesis.
- Published
- 2015
- Full Text
- View/download PDF
27. Complement protective epitopes and CD55-microtubule complexes facilitate the invasion and intracellular persistence of uropathogenic Escherichia coli.
- Author
-
Rana T, Hasan RJ, Nowicki S, Venkatarajan MS, Singh R, Urvil PT, Popov V, Braun WA, Popik W, Goodwin JS, and Nowicki BJ
- Subjects
- Adhesins, Escherichia coli immunology, Adhesins, Escherichia coli metabolism, Animals, CD55 Antigens genetics, CHO Cells, Cricetulus, Microscopy, Confocal, Microscopy, Electron, Mutant Proteins genetics, Mutant Proteins metabolism, Protein Conformation, CD55 Antigens metabolism, Complement System Proteins immunology, Endocytosis, Microtubules metabolism, Uropathogenic Escherichia coli immunology, Uropathogenic Escherichia coli physiology
- Abstract
Background: Escherichia coli-bearing Dr-adhesins (Dr+ E. coli) cause chronic pyelonephritis in pregnant women and animal models. This chronic renal infection correlates with the capacity of bacteria to invade epithelial cells expressing CD55. The mechanism of infection remains unknown., Methods: CD55 amino acids in the vicinity of binding pocket-Ser155 for Dr-adhesin were mutated to alanine and subjected to temporal gentamicin-invasion/gentamicin-survival assay in Chinese hamster ovary cells. CD55/microtubule (MT) responses were studied using confocal/electron microscopy, and 3-dimensional structure analysis., Results: Mutant analysis revealed that complement-protective CD55-Ser165 and CD55-Phe154 epitopes control E. coli invasion by coregulating CD55-MT complex expression. Single-point CD55 mutations changed E. coli to either a minimally invasive (Ser165Ala) or a hypervirulent pathogen (Phe154Ala). Thus, single amino acid modifications with no impact on CD55 structure and bacterial attachment can have a profound impact on E. coli virulence. While CD55-Ser165Ala decreased E. coli invasion and led to dormant intracellular persistence, intracellular E. coli in CD55-Phe154Ala developed elongated forms (multiplying within vacuoles), upregulated CD55-MT complexes, acquired CD55 coat, and escaped phagolysosomal fusion., Conclusions: E. coli target complement-protective CD55 epitopes for invasion and exploit CD55-MT complexes to escape phagolysosomal fusion, leading to a nondestructive parasitism that allows bacteria to persist intracellularly.
- Published
- 2014
- Full Text
- View/download PDF
28. The innate immune factor apolipoprotein L1 restricts HIV-1 infection.
- Author
-
Taylor HE, Khatua AK, and Popik W
- Subjects
- Apolipoprotein L1, Cell Differentiation, Cell Line, Endocytosis, Flow Cytometry, Gene Products, gag immunology, HIV-1 physiology, Humans, Interferons immunology, Macrophages immunology, Virus Replication, Apolipoproteins physiology, HIV Infections immunology, HIV-1 isolation & purification, Immunity, Innate, Lipoproteins, HDL physiology
- Abstract
Apolipoprotein L1 (APOL1) is a major component of the human innate immune response against African trypanosomes. Although the mechanism of the trypanolytic activity of circulating APOL1 has been recently clarified, the intracellular function(s) of APOL1 in human cells remains poorly defined. Like that of many genes linked to host immunity, APOL1 expression is induced by proinflammatory cytokines gamma interferon (IFN-γ) and tumor necrosis factor alpha (TNF-α). Additionally, IFN-γ-polarized macrophages that potently restrict HIV-1 replication express APOL1, which suggests that APOL1 may contribute to HIV-1 suppression. Here, we report that APOL1 inhibits HIV-1 replication by multiple mechanisms. We found that APOL1 protein targeted HIV-1 Gag for degradation by the endolysosomal pathway. Interestingly, we found that APOL1 stimulated both endocytosis and lysosomal biogenesis by promoting nuclear localization of transcription factor EB (TFEB) and expression of TFEB target genes. Moreover, we demonstrated that APOL1 depletes cellular viral accessory protein Vif, which counteracts the host restriction factor APOBEC3G, via a pathway involving degradation of Vif in lysosomes and by secretion of Vif in microvesicles. As a result of Vif depletion by APOL1, APOBEC3G was not degraded and reduced infectivity of progeny virions. In support of this model, we also showed that endogenous expression of APOL1 in differentiated U937 monocytic cells stimulated with IFN-γ resulted in a reduced production of virus particles. This finding supports the hypothesis that induction of APOL1 contributes to HIV-1 suppression in differentiated monocytes. Deciphering the precise mechanism of APOL1-mediated HIV-1 restriction may facilitate the design of unique therapeutics to target HIV-1 replication.
- Published
- 2014
- Full Text
- View/download PDF
29. Sterol regulatory element-binding protein 2 couples HIV-1 transcription to cholesterol homeostasis and T cell activation.
- Author
-
Taylor HE, Linde ME, Khatua AK, Popik W, and Hildreth JE
- Subjects
- Base Sequence, Cell Line, DNA Primers, Flow Cytometry, Humans, Protein Binding, Sterol Regulatory Element Binding Protein 2 metabolism, Transcription Factors, TFII genetics, Cholesterol metabolism, HIV-1 genetics, Homeostasis physiology, Lymphocyte Activation, Sterol Regulatory Element Binding Protein 2 physiology, T-Lymphocytes immunology, Transcription, Genetic physiology
- Abstract
Cholesterol plays an essential role in the life cycle of several enveloped viruses. Many of these viruses manipulate host cholesterol metabolism to facilitate their replication. HIV-1 infection of CD4(+) T cells activates the sterol regulatory element-binding protein 2 (SREBP2) transcriptional program, which includes genes involved in cholesterol homeostasis. However, the role of SREBP2-dependent transcription in HIV-1 biology has not been fully examined. Here, we identify TFII-I, a gene critical for HIV-1 transcription in activated T cells, as a novel SREBP2 target gene. We found TFII-I expression increased after HIV-1 infection or activation of human primary CD4(+) T cells. We show that inhibition of SREBP2 activity reduced TFII-I induction in response to these stimuli. More importantly, small interfering RNA (siRNA)-mediated gene silencing of either SREBP2 or TFII-I significantly reduced HIV-1 production in CD4(+) T cells. We also found that TFII-I potentiates Tat-dependent viral gene expression, consistent with a role at the level of HIV-1 transcription. Collectively, our results demonstrate for the first time that HIV-1 transcription in T cells is linked to cholesterol homeostasis through control of TFII-I expression by SREBP2.
- Published
- 2011
- Full Text
- View/download PDF
30. Non-productive HIV-1 infection of human glomerular and urinary podocytes.
- Author
-
Khatua AK, Taylor HE, Hildreth JE, and Popik W
- Subjects
- Cell Line, Endocytosis, HIV-1 growth & development, HIV-1 pathogenicity, Humans, Receptors, HIV genetics, Virus Release, HIV-1 physiology, Podocytes virology, Receptors, HIV biosynthesis, Virus Internalization, Virus Replication
- Abstract
Podocyte damage induced by HIV-1 is critical to the pathogenesis of HIV-1 associated nephropathy (HIVAN) and is believed to result from productive replication of the virus. Here we demonstrate that HIV-1 readily enters human podocytes by a dynamin-mediated endocytosis but does not establish productive infection. We provide evidence suggesting that viral nucleic acids and proteins detected in podocytes are delivered by viral particles internalized by the cells. Endocytosed HIV-1 is only transiently harbored by podocytes and is subsequently released to the extracellular milieu as fully infectious virus. Similarly, primary podocytes established from normal human urine do not support productive infection by HIV-1 but sustain replication of VSV-G pseudotyped virus that bypasses HIV-1 entry receptors. Moreover, transfected podocytes expressing CD4 and CXCR4 receptors support productive replication of HIV-1. This further confirms that lack of HIV-1 entry receptors is the major barrier preventing productive infection of podocytes in vitro., (Copyright © 2010 Elsevier Inc. All rights reserved.)
- Published
- 2010
- Full Text
- View/download PDF
31. Inhibition of LINE-1 and Alu retrotransposition by exosomes encapsidating APOBEC3G and APOBEC3F.
- Author
-
Khatua AK, Taylor HE, Hildreth JE, and Popik W
- Subjects
- APOBEC-3G Deaminase, CD4-Positive T-Lymphocytes metabolism, Cell Line, Dendritic Cells metabolism, Humans, Plasmids genetics, RNA, Messenger genetics, RNA, Messenger metabolism, Retroviridae genetics, Alu Elements, Cytidine Deaminase metabolism, Cytosine Deaminase metabolism, Exosomes enzymology, Exosomes genetics, Long Interspersed Nucleotide Elements
- Abstract
Human cytidine deaminases, including APOBEC3G (A3G) and A3F, are part of a cellular defense system against retroviruses and retroelements including non-LTR retrotransposons LINE-1 (L1) and Alu. Expression of cellular A3 proteins is sufficient for inhibition of L1 and Alu retrotransposition, but the effect of A3 proteins transferred in exosomes on retroelement mobilization is unknown. Here, we demonstrate for the first time that exosomes secreted by CD4(+)H9 T cells and mature monocyte-derived dendritic cells encapsidate A3G and A3F and inhibit L1 and Alu retrotransposition. A3G is the major contributor to the inhibitory activity of exosomes, however, the contribution of A3F in H9 exosomes cannot be excluded. Additionally, we show that exosomes encapsidate mRNAs coding for A3 proteins. A3G mRNA, and less so A3F, was enriched in exosomes secreted by H9 cells. Exosomal A3G mRNA was functional in vitro. Whether exosomes inhibit retrotransposons in vivo requires further investigation., (Copyright 2010 Elsevier Inc. All rights reserved.)
- Published
- 2010
- Full Text
- View/download PDF
32. Exosomes packaging APOBEC3G confer human immunodeficiency virus resistance to recipient cells.
- Author
-
Khatua AK, Taylor HE, Hildreth JE, and Popik W
- Subjects
- APOBEC-3G Deaminase, Cell Line, DNA, Viral genetics, DNA, Viral metabolism, Humans, Polymerase Chain Reaction, Viral Proteins biosynthesis, Virus Replication immunology, Cytidine Deaminase immunology, Exosomes immunology, HIV-1 immunology
- Abstract
The human cytidine deaminase APOBEC3G (A3G) is a part of a cellular defense system against human immunodeficiency virus type 1 (HIV-1) and other retroviruses. Antiretroviral activity of A3G can be severely blunted in the presence of the HIV-1 protein Vif. However, in some cells expressing the enzymatically active low-molecular-mass form of A3G, HIV-1 replication is restricted at preintegration steps, before accumulation of Vif. Here, we show that A3G can be secreted by cells in exosomes that confer resistance to both vif-defective and wild-type HIV-1 in exosome recipient cells. Our results also suggest that A3G is the major exosomal component responsible for the anti-HIV-1 activity of exosomes. However, enzymatic activity of encapsidated A3G does not correlate with the observed limited cytidine deamination in HIV-1 DNA, suggesting that A3G-laden exosomes restrict HIV-1 through a nonenzymatic mechanism. Real-time PCR quantitation demonstrated that A3G exosomes reduce accumulation of HIV-1 reverse transcription products and steady-state levels of HIV-1 Gag and Vif proteins. Our findings suggest that A3G exosomes could be developed into a novel class of anti-HIV-1 therapeutics.
- Published
- 2009
- Full Text
- View/download PDF
33. Cocaine increases human immunodeficiency virus type 1 neuroinvasion through remodeling brain microvascular endothelial cells.
- Author
-
Fiala M, Eshleman AJ, Cashman J, Lin J, Lossinsky AS, Suarez V, Yang W, Zhang J, Popik W, Singer E, Chiappelli F, Carro E, Weinand M, Witte M, and Arthos J
- Subjects
- Blood-Brain Barrier virology, Brain blood supply, Cells, Cultured, Cocaine metabolism, Dose-Response Relationship, Drug, Electric Impedance, Endothelium, Vascular physiology, Endothelium, Vascular virology, Humans, Permeability, Pinocytosis, Receptors, Estrogen metabolism, Receptors, Muscarinic metabolism, Signal Transduction, Time Factors, Virus Replication drug effects, Cocaine pharmacology, HIV-1 physiology
- Abstract
Cocaine is a suspected cofactor in human immunodeficiency virus (HIV)-associated dementia but cocaine's effects are not clear. Herein the authors describe investigations of the mechanisms by which cocaine increases HIV-1 invasion through brain microvascular endothelial cells (BMVECs). Cocaine binds to a site on BMVECs, which is not a biogenic amine transporter, a binding site for estrogen, or a muscarinic receptor and for which benztropine and tamoxifen have the highest affinity. Cocaine treatment of BMVECs disrupts intercellular junctions and induces cell ruffling, which could account for their increased permeability and decreased electrical resistance. HIV-1 enters BMVECs by macropinocytosis and is transported to lysosomes and inactivated. In cocaine-treated BMVECs, the virus enters and persists in large cytoplasmic "lakes." Cocaine exposure of BMVECs up-regulates transcription of genes important in cytoskeleton organization, signal transduction, cell swelling, vesicular trafficking, and cell adhesion. The toxicity of cocaine for the blood-brain barrier may lead to increased virus neuroinvasion and neurovascular complications of cocaine abuse.
- Published
- 2005
- Full Text
- View/download PDF
34. APOBEC3G is incorporated into virus-like particles by a direct interaction with HIV-1 Gag nucleocapsid protein.
- Author
-
Alce TM and Popik W
- Subjects
- APOBEC-3G Deaminase, Anti-Retroviral Agents pharmacology, Blotting, Western, Cell Line, Cytidine Deaminase, DNA, Complementary metabolism, Endosomes metabolism, Gene Products, vif metabolism, Glutathione Transferase metabolism, Humans, Lentivirus genetics, Lipids chemistry, Membrane Microdomains, Microscopy, Confocal, Microscopy, Fluorescence, Nucleocapsid metabolism, Nucleoside Deaminases, Precipitin Tests, Protein Binding, Protein Structure, Tertiary, RNA metabolism, Recombinant Proteins chemistry, Repressor Proteins, Retroviridae genetics, Transcription, Genetic, Transfection, Virus Assembly, Virus Replication, vif Gene Products, Human Immunodeficiency Virus, Gene Products, gag metabolism, HIV-1 metabolism, Nucleocapsid Proteins metabolism, Proteins chemistry
- Abstract
APOBEC3G belongs to the family of cellular cytidine deaminase-editing enzymes with a potent antiretroviral activity, which is counteracted by the Vif protein expressed by lentiviruses. Antiretroviral activity of APOBEC3G requires its packaging into assembling virions, presumably to ensure its close association with nascent retroviral cDNA. Here, we demonstrate that APOBEC3G is encapsidated through a direct interaction with the HIV-1 Gag polyprotein which likely takes place on the membranes of the multivesicular bodies (MVB)/late endosomal compartments. This interaction is mediated by the Gag nucleocapsid protein NC, and the N-terminal part of NC is most critical for this interaction. Binding to the NC domain would ensure that APOBEC3G will be concentrated in the viral core of mature HIV-1, in close proximity to the reverse transcription complex.
- Published
- 2004
- Full Text
- View/download PDF
35. CD4 receptor localized to non-raft membrane microdomains supports HIV-1 entry. Identification of a novel raft localization marker in CD4.
- Author
-
Popik W and Alce TM
- Subjects
- Amino Acid Sequence, Base Sequence, CD4 Antigens analysis, CD4 Antigens genetics, Cell Line, Cells, Cultured, DNA Primers, Glycosylation, Green Fluorescent Proteins, Humans, Luminescent Proteins immunology, Oligosaccharides pharmacology, Palmitic Acid metabolism, Peptide Fragments chemistry, Receptors, HIV analysis, Recombinant Fusion Proteins immunology, B-Lymphocytes immunology, CD4 Antigens immunology, Cell Membrane immunology, HIV-1 physiology, Membrane Microdomains immunology, T-Lymphocytes immunology
- Abstract
Despite the preferential localization of CD4 to lipid rafts, the significance and role of these microdomains in HIV-1 entry is still controversial. The possibility that CD4, when localized to non-raft domains, might be able to support virus entry cannot be excluded. Because disintegration of rafts by extraction of cellular cholesterol with methyl-beta-cyclodextrin suffers from various adverse effects, we investigated molecular determinants controlling raft localization of the CD4 receptor. Extensive mutagenesis of the receptor showed that a raft-localizing marker, consisting of a short sequence of positively charged amino acid residues, RHRRR, was present in the membrane-proximal cytoplasmic domain of CD4. Substitution of the RHRRR sequence with alanine residues abolished raft localization of the CD4 mutant, RA5, as determined biochemically using solubilization in nonionic detergents and by confocal microscopy. The possible inhibitory effect of the introduced mutations on the adjacent CVRC palmitoylation site was ruled out because wild type (wt) CD4 and RA5, but not a palmitoylation-deficient mutant, were efficiently palmitoylated. Nonetheless, the RA5 mutant supported productive virus entry to levels equivalent to that of wild type (wt) CD4. Sucrose gradient analysis of Triton X-100 virus lysates showed that Gag and envelope gp120 proteins accumulated in low buoyant, high-density fractions. This pattern was changed after virus incubation with cells. Whereas Gag proteins localized to lipid rafts in cells expressing wt CD4 and RA5, gp120 accumulated in rafts in cells expressing wt CD4 but not RA5. We propose that raft localization of CD4 is not required for virus entry, however, post-binding fusion/entry steps may require lipid raft assembly.
- Published
- 2004
- Full Text
- View/download PDF
36. HIV-1 induces cardiomyopathyby cardiomyocyte invasion and gp120, Tat, and cytokine apoptotic signaling.
- Author
-
Fiala M, Popik W, Qiao JH, Lossinsky AS, Alce T, Tran K, Yang W, Roos KP, and Arthos J
- Subjects
- Animals, Animals, Newborn, Cardiomyopathies metabolism, Cardiomyopathies pathology, Caspase 3, Caspases metabolism, Cells, Cultured, Coronary Vessels metabolism, Coronary Vessels pathology, Coronary Vessels ultrastructure, DNA, Viral metabolism, Endothelial Cells metabolism, Endothelial Cells pathology, Endothelial Cells ultrastructure, Extracellular Signal-Regulated MAP Kinases metabolism, Humans, Immunohistochemistry, In Situ Hybridization, In Situ Nick-End Labeling, Macrophages virology, Myocytes, Cardiac metabolism, Myocytes, Cardiac ultrastructure, Phosphorylation, Polymerase Chain Reaction, RNA, Viral metabolism, Rats, Signal Transduction, tat Gene Products, Human Immunodeficiency Virus, Apoptosis, Cardiomyopathies etiology, Cytokines physiology, Gene Products, tat physiology, HIV Envelope Protein gp120 physiology, HIV Infections complications, HIV-1, Myocytes, Cardiac pathology
- Abstract
We examined heart tissues of AIDS patients with or without HIV cardiomyopathy (HIVCM) by immunohistochemistry, in situ polymerase chain reaction, in situ riboprobe hybridization, and the TUNEL technique for apoptosis. In HIVCM tissues, only inflammatory cells, but not endothelial cells or cardiomyocytes, displayed HIV-1 DNA and RNA. However, macrophages, lymphocytes, and--in a patchy fashion--cardiomyocytes and endothelial cells exhibited virus envelope protein gp120. Macrophages infiltrated the myocardium in a perivascular fashion and expressed tumor necrosis factor family ligands; adjacent cardiomyocytes suffered apoptosis. In vitro HIV-1 strongly invaded neonatal rat ventricular myocytes (NRVMs) and coronary artery endothelial cells (CAECs) and induced microvilli but did not replicate. HIV-1, gp120, or Tat induced Erk 1/2 phosphorylation, activation of caspase-3, and apoptosis of NRVMs and CAECs; all of these were inhibited by a MAPK/ERK-kinase (MEK) inhibitor U0126. The pathogenesis of HIVCM involves HIV-1 replication in inflammatory cells and induction of cardiomyocyte apoptosis by (1) the extrinsic pathway through apoptotic ligands and (2) the intrinsic pathway through direct virus entry and gp120- and Tat-proapoptotic signaling.
- Published
- 2004
- Full Text
- View/download PDF
37. Cardiomyocytes undergo apoptosis in human immunodeficiency virus cardiomyopathy through mitochondrion- and death receptor-controlled pathways.
- Author
-
Twu C, Liu NQ, Popik W, Bukrinsky M, Sayre J, Roberts J, Rania S, Bramhandam V, Roos KP, MacLellan WR, and Fiala M
- Subjects
- Acquired Immunodeficiency Syndrome complications, Acquired Immunodeficiency Syndrome pathology, Animals, Cardiomyopathies complications, Cardiomyopathies pathology, Cells, Cultured, Chemokine CCL5 metabolism, Cholesterol metabolism, Fas Ligand Protein, G(M1) Ganglioside metabolism, HIV Envelope Protein gp120 metabolism, Heparin metabolism, Humans, Macrophages, Pinocytosis, Rats, Rats, Sprague-Dawley, Acquired Immunodeficiency Syndrome metabolism, Apoptosis, Cardiomyopathies metabolism, Chemokine CCL5 analogs & derivatives, HIV-1 metabolism, Membrane Glycoproteins metabolism, Mitochondria metabolism, Myocardium cytology, Tumor Necrosis Factor-alpha metabolism
- Abstract
We investigated 18 AIDS hearts (5 with and 13 without cardiomyopathy) by using immunocytochemistry and computerized image analysis regarding the roles of HIV-1 proteins and tumor necrosis factor ligands in HIV cardiomyopathy (HIVCM). HIVCM and cardiomyocyte apoptosis were significantly related to each other and to the expression by inflammatory cells of gp120 and tumor necrosis factor-alpha. In HIVCM heart, active caspase 9, a component of the mitochondrion-controlled apoptotic pathway, and the elements of the death receptor-mediated pathway, tumor necrosis factor-alpha and Fas ligand, were expressed strongly on macrophages and weakly on cardiomyocytes. HIVCM showed significantly greater macrophage infiltration and cardiomyocyte apoptosis rate compared with non-HIVCM. HIV-1 entered cultured neonatal rat ventricular myocytes by macropinocytosis but did not replicate. HIV-1- or gp120-induced apoptosis of rat myocytes through a mitochondrion-controlled pathway, which was inhibited by heparin, AOP-RANTES, or pertussis toxin, suggesting that cardiomyocyte apoptosis is induced by signaling through chemokine receptors. In conclusion, in patients with HIVCM, cardiomyocytes die through both mitochondrion- and death receptor-controlled apoptotic pathways.
- Published
- 2002
- Full Text
- View/download PDF
38. Human immunodeficiency virus type 1 enters brain microvascular endothelia by macropinocytosis dependent on lipid rafts and the mitogen-activated protein kinase signaling pathway.
- Author
-
Liu NQ, Lossinsky AS, Popik W, Li X, Gujuluva C, Kriederman B, Roberts J, Pushkarsky T, Bukrinsky M, Witte M, Weinand M, and Fiala M
- Subjects
- Blood-Brain Barrier, Cells, Cultured, HIV Infections virology, Humans, Microcirculation, Microscopy, Confocal, Mitogen-Activated Protein Kinases metabolism, Models, Biological, Signal Transduction, Brain blood supply, Endothelium, Vascular virology, HIV-1 physiology, MAP Kinase Signaling System, Membrane Microdomains metabolism, Pinocytosis
- Abstract
Brain microvascular endothelial cells (BMVECs) present an incomplete barrier to human immunodeficiency virus type 1 (HIV-1) neuroinvasion. In order to clarify the mechanisms of HIV-1 invasion, we have examined HIV-1 uptake and transcellular penetration in an in vitro BMVEC model. No evidence of productive infection was observed by luciferase, PCR, and reverse transcriptase assays. Approximately 1% of viral RNA and 1% of infectious virus penetrated the BMVEC barrier without disruption of tight junctions. The virus upregulated ICAM-1 on plasma membranes and in cytoplasmic vesiculotubular structures. HIV-1 virions were entangled by microvilli and were taken into cytoplasmic vesicles through surface invaginations without fusion of the virus envelope with the plasma membrane. Subsequently, the cytoplasmic vesicles fused with lysosomes, the virions were lysed, and the vesicles diminished in size. Upon cell entry, HIV-1 colocalized with cholera toxin B, which targets lipid raft-associated GM1 ganglioside. Cholesterol-extracting agents, cyclodextrin and nystatin, and polyanion heparin significantly inhibited virus entry. Anti-CD4 had no effect and the chemokine AOP-RANTES had only a slight inhibitory effect on virus entry. HIV-1 activated the mitogen-activated protein kinase (MAPK) pathway, and inhibition of MAPK/Erk kinase inhibited virus entry. Entry was also blocked by dimethylamiloride, indicating that HIV-1 enters endothelial cells by macropinocytosis. Therefore, HIV-1 penetrates BMVECs in ICAM-1-lined macropinosomes by a mechanism involving lipid rafts, MAPK signaling, and glycosylaminoglycans, while CD4 and chemokine receptors play limited roles in this process.
- Published
- 2002
- Full Text
- View/download PDF
39. Human immunodeficiency virus type 1 uses lipid raft-colocalized CD4 and chemokine receptors for productive entry into CD4(+) T cells.
- Author
-
Popik W, Alce TM, and Au WC
- Subjects
- CD4-Positive T-Lymphocytes chemistry, CD4-Positive T-Lymphocytes immunology, Cell Line, Cholesterol analysis, Cholesterol deficiency, Cyclodextrins pharmacology, G(M1) Ganglioside metabolism, HIV Envelope Protein gp120 metabolism, HIV-1 metabolism, Humans, Jurkat Cells, Membrane Lipids chemistry, Membrane Lipids isolation & purification, Membrane Microdomains immunology, Membrane Microdomains metabolism, Microscopy, Confocal, Receptors, CCR5 metabolism, Receptors, CXCR4 metabolism, Receptors, Chemokine analysis, Virus Replication drug effects, CD4 Antigens metabolism, CD4-Positive T-Lymphocytes virology, HIV-1 physiology, Membrane Lipids metabolism, Membrane Microdomains virology, Receptors, Chemokine metabolism, beta-Cyclodextrins
- Abstract
In this report, we describe a crucial role of lipid raft-colocalized receptors in the entry of human immunodeficiency virus type 1 (HIV-1) into CD4(+) T cells. We show that biochemically isolated detergent-resistant fractions have characteristics of lipid rafts. Lipid raft integrity was required for productive HIV-1 entry as determined by (i) semiquantitative PCR analysis and (ii) single-cycle infectivity assay using HIV-1 expressing the luciferase reporter gene and pseudotyped with HIV-1 HXB2 envelope or vesicular stomatitis virus envelope glycoprotein (VSV-G). Depletion of plasma membrane cholesterol with methyl-beta-cyclodextrin (MbetaCD) relocalized raft-resident markers to a nonraft environment but did not significantly change the surface expression of HIV-1 receptors. MbetaCD treatment inhibited productive infection of HIV-1 by 95% as determined by luciferase activity in cells infected with HXB2 envelope-pseudotyped virus. In contrast, infection with VSV-G-pseudotyped virus, which enters the cells through an endocytic pathway, was not suppressed. Biochemical fractionation and confocal imaging of HIV-1 receptor distribution in live cells demonstrated that CD4, CCR5, and CXCR4 colocalized with raft-resident markers, ganglioside GM1, and glycosylphosphatidylinositol-anchored CD48. While confocal microscopy analysis revealed that HIV-1 receptors localized most likely to the same lipid microdomains, sucrose gradient analysis of the receptor localization showed that, in contrast to CD4 and CCR5, CXCR4 was associated preferentially with the nonraft membrane fraction. The binding of HIV-1 envelope gp120 to lipid rafts in the presence, but not in the absence, of cholesterol strongly supports our hypothesis that raft-colocalized receptors are directly involved in virus entry. Dramatic changes in lipid raft and HIV-1 receptor redistribution were observed upon binding of HIV-1 NL4-3 to PM1 T cells. Colocalization of CCR5 with GM1 and gp120 upon engagement of CD4 and CXCR4 by HIV-1 further supports our observation that HIV-1 receptors localize to the same lipid rafts in PM1 T cells.
- Published
- 2002
- Full Text
- View/download PDF
40. HIV-1 penetrates coronary artery endothelial cells by transcytosis.
- Author
-
Gujuluva C, Burns AR, Pushkarsky T, Popik W, Berger O, Bukrinsky M, Graves MC, and Fiala M
- Subjects
- Arteries ultrastructure, Base Sequence, Cells, Cultured, Chemokines physiology, Coronary Vessels ultrastructure, DNA Primers, Gene Products, pol genetics, HIV Long Terminal Repeat, Microscopy, Electron, Reverse Transcriptase Polymerase Chain Reaction, Transcription, Genetic, Virus Replication, Arteries virology, Coronary Vessels virology, Endocytosis, HIV-1 physiology
- Abstract
Background: The pathogenesis of HIV-1-related cardiomyopathy is poorly understood, but HIV-1 has been detected in cardiomyocytes. Whether HIV-1 penetrates into the myocardium by infection of coronary artery endothelial cells (CAEC) or using transcellular or paracellular routes across CAEC has not been resolved., Materials and Methods: A model of the CAEC barrier was constructed with primary CAEC (derived from human coronary vessels). Polymerase chain reaction (PCR) assay, infectious assay, and immunofluorescence were employed to show abortive nature of HIV-1 infection of CAEC. Tight junction (TJ) and cell adhesion proteins were visualized by immunofluorescence. The time course of HIV-1 invasion was measured by HIV-1 RNA assay. Inulin permeability assay determined paracellular leakage. Transmission electron microscopy demonstrated virus-induced endothelial vacuolization., Results: Despite a strong display on CAEC of CXCR4 and a lesser expression of CCR3 and CCR5, HIV-1 did not productively replicate in CAEC, as shown by infectious assay, immunofluorescence, and electron microscopy. HIV-1 infection of CAEC was abortive with minimal reverse transcription of strong stop DNA and pol but not full-length or two LTR DNA circles. Upon infection of the model with 1 million RNA copies of HIV-1JR-FL, virus penetration 2 hr postinfection (PI) was negligible but increased by 1,750% 24 hr PI. The paracellular permeability increased during this period by only 25%. Neither AOP-RANTES nor v-MIPII significantly reduced HIV-1JR-FL invasion. Virus infection did not alter the integral TJ protein occludin and the TJ-associated protein ZO-1. HIV-1 exposed CAEC and brain microvascular endothelial cells (BMVEC) developed extensive cytoplasmic vacuolization with retroviral-like particles in the vacuoles., Conclusions: The endothelium is not an impenetrable barrier to HIV-1. The virus opens a transcellular route across coronary and brain endothelia in cytoplasmic vacuoles.
- Published
- 2001
41. Exploitation of cellular signaling by HIV-1: unwelcome guests with master keys that signal their entry.
- Author
-
Popik W and Pitha PM
- Subjects
- CD4 Antigens physiology, Glycosaminoglycans physiology, HIV Envelope Protein gp120 physiology, Humans, Mitogen-Activated Protein Kinases physiology, Receptors, Chemokine physiology, HIV-1 physiology
- Published
- 2000
- Full Text
- View/download PDF
42. Inhibition of CD3/CD28-mediated activation of the MEK/ERK signaling pathway represses replication of X4 but not R5 human immunodeficiency virus type 1 in peripheral blood CD4(+) T lymphocytes.
- Author
-
Popik W and Pitha PM
- Subjects
- Biological Transport, Butadienes pharmacology, CD4 Antigens metabolism, CD4-Positive T-Lymphocytes cytology, Cell Nucleus metabolism, Cells, Cultured, DNA, Viral metabolism, Down-Regulation, Enzyme Activation, Enzyme Inhibitors pharmacology, HIV-1 drug effects, HIV-1 isolation & purification, Humans, Mitogen-Activated Protein Kinase Kinases antagonists & inhibitors, Nitriles pharmacology, Proviruses genetics, Receptors, CXCR4 metabolism, Transcription, Genetic, Virus Integration, CD28 Antigens metabolism, CD3 Complex metabolism, CD4-Positive T-Lymphocytes virology, HIV-1 physiology, MAP Kinase Signaling System, Mitogen-Activated Protein Kinase Kinases metabolism, Virus Replication drug effects
- Abstract
Binding of human immunodeficiency virus type 1 (HIV-1) to CD4 receptors induces multiple cellular signaling pathways, including the MEK/ERK cascade. While the interaction of X4 HIV-1 with CXCR4 does not seem to activate this pathway, viruses using CCR5 for entry efficiently activate MEK/ERK kinases (W. Popik, J. E. Hesselgesser, and P. M. Pitha, J. Virol. 72:6406-6413, 1998; W. Popik and P. M. Pitha, Virology 252:210-217, 1998). Since the importance of MEK/ERK in the initial steps of viral replication is poorly understood, we have examined the role of MEK/ERK signaling in the CD3- and CD28 (CD3/CD28)-mediated activation of HIV-1 replication in resting peripheral blood CD4(+) T lymphocytes infected with X4 or R5 HIV-1. We have found that the MEK/ERK inhibitor U0126 selectively inhibited CD3/CD28-stimulated replication of X4 HIV-1, while it did not affect the replication of R5 HIV-1. Inhibition of the CD3/CD28-stimulated MEK/ERK pathway did not affect the formation of the early proviral transcripts in cells infected with either X4 or R5 HIV-1, indicating that virus reverse transcription is not affected in the absence of MEK/ERK signaling. In contrast, the levels of nuclear provirus in cells infected with X4 HIV-1, detected by the formation of circular proviral DNA, was significantly lower in cells stimulated in the presence of MEK/ERK inhibitor than in the absence of the inhibitor. However, in cells infected with R5 HIV-1, the inhibition of the MEK/ERK pathway did not affect nuclear localization of the proviral DNA. These data suggest that the nuclear import of X4, but not R5, HIV-1 is dependent on a CD3/CD28-stimulated MEK/ERK pathway.
- Published
- 2000
- Full Text
- View/download PDF
43. Early activation of mitogen-activated protein kinase kinase, extracellular signal-regulated kinase, p38 mitogen-activated protein kinase, and c-Jun N-terminal kinase in response to binding of simian immunodeficiency virus to Jurkat T cells expressing CCR5 receptor.
- Author
-
Popik W and Pitha PM
- Subjects
- CD4-Positive T-Lymphocytes metabolism, Enzyme Activation, HIV-1 pathogenicity, Humans, JNK Mitogen-Activated Protein Kinases, Jurkat Cells, Mitogen-Activated Protein Kinase 1, Mitogen-Activated Protein Kinase 3, Mitogen-Activated Protein Kinase Kinases, Receptors, CXCR4 biosynthesis, Signal Transduction, p38 Mitogen-Activated Protein Kinases, CD4-Positive T-Lymphocytes enzymology, Calcium-Calmodulin-Dependent Protein Kinases metabolism, Mitogen-Activated Protein Kinases, Protein Kinases metabolism, Receptors, CCR5 biosynthesis, Simian Immunodeficiency Virus metabolism
- Abstract
We have shown that the binding of simian immunodeficiency virus (SIV) to Jurkat T cells expressing CD4 receptor strongly induces mitogen-activated protein (MAP) kinase kinase (MEK) and extracellular signal-regulated kinases 1 and 2 (ERK1/2) and only weakly induces p38 MAP kinase and c-Jun N-terminal kinase (JNK). Similarly, T-tropic NL4-3 virus, which uses both CD4 and CXCR4 receptors for entry, stimulated in these cells the MEK/ERK MAP kinase (MAPK) pathway in a CD4 receptor-dependent manner (Popik and Pitha, 1998). In contrast, both macrophage-tropic SIVmac316 and T cell-tropic SIVmac239, which in addition to CD4 require CCR5 coreceptor for entry, significantly enhanced early MEK/ERK, p38 MAPK, and JNK signaling in Jurkat cells expressing constitutively or transiently the CCR5 receptor. Together, this study provides the evidence that viruses using CXCR4 or CCR5 receptors for entry may differentially use signaling properties of their specific coreceptors to stimulate MAP kinase cascades. In addition, although SIVmac239 and SIVmac316 use different structural domains of the CCR5 receptor for entry, both viruses stimulate early phosphorylation of MEK, ERK, p38, and JNK independently of their tropism and replication.
- Published
- 1998
- Full Text
- View/download PDF
44. Binding of human immunodeficiency virus type 1 to CD4 and CXCR4 receptors differentially regulates expression of inflammatory genes and activates the MEK/ERK signaling pathway.
- Author
-
Popik W, Hesselgesser JE, and Pitha PM
- Subjects
- Antibodies, Monoclonal metabolism, Binding Sites, CCAAT-Enhancer-Binding Proteins, CD4-Positive T-Lymphocytes metabolism, Calcium-Calmodulin-Dependent Protein Kinases metabolism, Chemokine CCL4, Chemokine CXCL12, Chemokines, CXC metabolism, Cytoplasm, DNA-Binding Proteins metabolism, Enzyme Activation, HIV Envelope Protein gp120 metabolism, Humans, Jurkat Cells, Lymphocyte Specific Protein Tyrosine Kinase p56(lck) metabolism, MAP Kinase Kinase 1, MAP Kinase Kinase 2, Macrophage Inflammatory Proteins genetics, Macrophages virology, Mitogen-Activated Protein Kinase 1, Mitogen-Activated Protein Kinase 3, NF-kappa B metabolism, Nuclear Proteins metabolism, Transcription Factor AP-1 metabolism, CD4 Antigens metabolism, Gene Expression Regulation, Viral, HIV-1 metabolism, Interferon-gamma genetics, Mitogen-Activated Protein Kinase Kinases, Mitogen-Activated Protein Kinases, Protein Serine-Threonine Kinases metabolism, Protein-Tyrosine Kinases metabolism, Receptors, CXCR4 metabolism, Signal Transduction, Tumor Necrosis Factor-alpha genetics
- Abstract
We have previously shown that binding of human immunodeficiency virus type 1 (HIV-1) virions to CD4 receptors stimulates association of Lck with Raf-1 and results in the activation of Raf-1 kinase in a Ras-independent manner. In the present study, we demonstrate that HIV-1 envelope glycoproteins of both T-cell-tropic and macrophagetropic strains rapidly activate the ERK/mitogen-activated protein (MAP) kinase pathway and the binding of nuclear transcription factors (AP-1, NF-kappaB, and C/EBP) and stimulate expression of cytokine and chemokine genes. The activation of this signaling pathway requires functional CD4 receptors and is independent of binding to CXCR4. Binding of the natural ligand stromal cell-derived factor 1 (SDF-1) to CXCR4, which inhibits entry of T-cell-tropic HIV-1, activates also the ERK/MAP kinase pathway. However, SDF-1 did not affect the CD4-mediated expression of cytokine and chemokine genes. These results provide firm molecular evidence that binding of HIV-1 envelope glycoproteins to CD4 receptor initiates a signaling pathway(s) independent of the binding to the chemokine receptor that leads to the aberrant expression of inflammatory genes and may contribute significantly to HIV-1 replication as well as to deregulation of the immune system.
- Published
- 1998
- Full Text
- View/download PDF
45. Modulation of interferon-mediated inhibition of human immunodeficiency virus type 1 by Tat.
- Author
-
Shirazi Y, Popik W, and Pitha PM
- Subjects
- HIV-1 genetics, HeLa Cells, Humans, RNA Processing, Post-Transcriptional, tat Gene Products, Human Immunodeficiency Virus, Gene Products, tat biosynthesis, HIV-1 drug effects, Interferons pharmacology, Virus Replication drug effects
- Abstract
Recently, we have shown that in acutely infected T cells interferons (IFNs) effectively inhibit the human immunodeficiency type 1 (HIV-1) proviral DNA synthesis during a single replication cycle. In the present study, we have evaluated the relative effectiveness of IFNs in restricting HIV-1 expression at post-transcriptional level. Treatment of HeLa cells with IFNs A* and B (up to 1,000 U/ml) did not result in a reduction in HIV-1 RNA and protein synthesis encoded by the transfected HIV-1 proviral clone. Interestingly, IFN treatment reduced significantly the HIV-1 mRNA levels encoded by the transfected tat-defective HIV-1 provirus, and this inhibition could be overcome by transfection with Tat- and Rev-expressing plasmids. These results suggest that HIV-1-encoded Tat and Rev can overcome the inhibitory effects of IFNs on HIV-1 replication.
- Published
- 1994
- Full Text
- View/download PDF
46. Differential effect of tumor necrosis factor-alpha and herpes simplex virus type 1 on the Tat-targeted inhibition of human immunodeficiency virus type 1 replication.
- Author
-
Popik W and Pitha PM
- Subjects
- Gene Expression Regulation, Viral drug effects, Humans, In Vitro Techniques, Lymphocytes microbiology, RNA, Viral metabolism, Retroviridae Proteins metabolism, tat Gene Products, Human Immunodeficiency Virus, Antiviral Agents pharmacology, Benzodiazepinones pharmacology, Gene Products, tat antagonists & inhibitors, Genes, tat, HIV-1 growth & development, Herpesvirus 1, Human genetics, Pyrroles pharmacology, Tumor Necrosis Factor-alpha pharmacology, Virus Replication drug effects
- Abstract
In this study, we have examined whether the Tat antagonist can inhibit human immunodeficiency virus type 1 (HIV-1) replication in the presence of cofactors that can activate transcription of HIV-1 provirus by an NF-kappa B-mediated mechanism, such as tumor necrosis factor-alpha (TNF-alpha) or herpes simplex virus type 1 (HSV-1) infection. As a prototype, we have chosen a low-molecular-weight Tat inhibitor, Ro5-3335, and analyzed its effect on HIV-1 replication in the presence of TNF-alpha and HSV-1 infection in acutely infected peripheral blood lymphocytes (PBLs) and T cells. Ro5-3335 inhibited HIV-1 replication both in CEM-174 cells and in PBLs, but the magnitude of the inhibition was inversely related to viral inoculum and the inhibition was only temporary; viral replication resumed at later times postinfection in spite of the continuous presence of the drug. In contrast, Ro5-3335 suppressed TNF-alpha-induced activation of HIV-1 replication in chronically infected T cells and monocytes that both expressed only low levels of HIV-1 constitutively, while its effect in high-expressing OM-10.1 cells was negligible in the presence of TNF-alpha. The inhibition of HIV-1 replication by Ro5-3335 was specific for the Tat-mediated effect and this drug was not able to inhibit the TNF-alpha-induced expression of the tat-defective HIV-1 provirus. In contrast to TNF-alpha, HSV-1-stimulated HIV-1 expression in the ACH-2 cells was effectively inhibited in the presence of Ro5-3335. These results demonstrate that Tat plays an essential role in HSV-1-mediated activation of HIV-1 provirus, while the TNF-alpha complementation of Tat shows cell-type specificity. These observations suggest that inhibition of the Tat function alone may not be sufficient for an effective anti-HIV-1 inhibition.
- Published
- 1994
- Full Text
- View/download PDF
47. Transcriptional activation of the tat-defective human immunodeficiency virus type-1 provirus: effect of interferon.
- Author
-
Popik W and Pitha PM
- Subjects
- Gene Expression, Genes, tat, HeLa Cells, Humans, Interferon Type I genetics, NF-kappa B metabolism, Proviruses genetics, Recombinant Proteins, Tetradecanoylphorbol Acetate pharmacology, Transcription, Genetic, Transcriptional Activation, tat Gene Products, Human Immunodeficiency Virus, Gene Expression Regulation, Viral, Gene Products, tat physiology, HIV-1 genetics, Interferon Type I pharmacology, Virus Replication drug effects
- Abstract
The effect of human interferon-alpha 2 (HuIFN-alpha 2) on the activation of HIV-1 provirus was studied in cell lines containing either an integrated tat-defective HIV-1 provirus (HIV-1 (-tat)) (HNHIVdt4 cells) or the HIV-1 (-tat) provirus and a plasmid in which the expression of HuIFN-alpha 2 was under the control of HIV LTR (HNHIV alpha 1 cells). In both cell lines, the expression of HIV-1 RNA was below the limit of detection, but transcription of the HIV-1 (-tat) provirus could be induced either by transfection with Tat-expressing plasmid or by treatment with TPA and cycloheximide (CHX). By contrast, stimulation with TPA alone induced HIV-1 transcription only in HNHIVdt4 cells, but not in HNHIV alpha 1 cells that produced low levels of IFN-alpha constitutively. Similarly in a transient expression assay, TPA upregulated transcription of the transfected HIV-1 CAT plasmid only in HNHIVdt4 cells, but not in HNHIV alpha 1 cells. UV-crosslinking analysis of NF-kappa B-specific proteins induced in TPA-treated cells showed the presence of 45 and 55 kDa NF-kappa B-binding protein in TPA-induced HNHIVdt4 cells while, in HNHIV alpha 1 cells, we detected only 55-, 110-, and 200-kDa proteins, but no 45-kDa protein. The transcriptional effects of IFN could not, however, be seen in the presence of Tat protein, suggesting that the virus developed a mechanism to overcome the IFN-mediated restrictions.
- Published
- 1992
- Full Text
- View/download PDF
48. Transforming growth factor-beta inhibits the antiviral action of interferons in human embryonic fibroblasts.
- Author
-
Zielińska-Jenczylik J, Inglot AD, Inglot O, and Popik W
- Subjects
- Embryo, Mammalian cytology, Fibroblasts drug effects, Fibroblasts microbiology, Humans, Encephalomyocarditis virus drug effects, Interferons antagonists & inhibitors, Transforming Growth Factor beta pharmacology, Vesicular stomatitis Indiana virus drug effects, Virus Replication drug effects
- Abstract
Transforming growth factor-beta (TGF-beta) at concentration of 10 ng/ml inhibited the development of the interferon-alpha- (IFN-alpha) or IFN-gamma-induced antiviral state in quiescent human embryonic fibroblasts. The action of the cytokines was dose-related; TGF-beta had no direct effect on the replication of either vesicular stomatitis virus (VSV) or encephalomyocarditis virus (EMCV) used as the challenge viruses in the IFN assays. We suggest that despite the fact that TGF-beta acts mainly as a "negative" growth factor, its interactions with IFNs in the antiviral assays resemble known effects of the typical "positive" peptide growth factors.
- Published
- 1991
49. Combined action of interferons and transforming growth factor beta on the proliferation of human fibroblasts.
- Author
-
Popik W and Inglot AD
- Subjects
- Cell Division drug effects, Dose-Response Relationship, Drug, Drug Interactions, Epidermal Growth Factor administration & dosage, Fibroblasts cytology, Humans, In Vitro Techniques, Fibroblasts drug effects, Interferons administration & dosage, Transforming Growth Factor beta administration & dosage
- Abstract
The effects of interferons (IFNs) and transforming growth factor-beta (TGF-beta) used alone and in combination on the multiplication of human embryonic diploid fibroblasts were studied. The experimental conditions were standardized and medium containing 2% of a single batch of fetal calf serum, which was almost minimal dose required for good the cell attachment and growth, was used. The observed effects were dose related. IFNs (types alpha, beta, or gamma) at concentration of 1 to 100 U/ml or TGF-beta at concentration of 0.1 to 1.0 ng/ml stimulated the cell multiplication by 20 to 25% when compared to the control cultures incubated without the factors. At higher doses both IFNs (10(2) to 10(5) U/ml) or TGF-beta (1 to 10 ng/ml) inhibited the proliferation of the cells. The antimitogenic action of IFNs and TGF-beta was synergistic. We suggest that IFNs as well as TGF-beta are bifunctional growth regulators although their antimitotic action dominates over their possible growth stimulatory activity. The latter action may be a secondary phenomenon due to interaction of IFNs or TGF-beta with other factors present in cell culture. We have shown that epidermal growth factor (EGF) may increase the growth stimulatory action of low doses of IFN-gamma or TGF-beta.
- Published
- 1991
50. Participation of polypeptide proteinase inhibitors in proliferation of fibroblasts.
- Author
-
Popik W, Inglot AD, Wilusz T, and Polanowski A
- Subjects
- Animals, Cells, Cultured, Growth Substances metabolism, Humans, In Vitro Techniques, Mice, Mice, Inbred BALB C, Rabbits, Trypsin genetics, Fibroblasts drug effects, Mitogens, Protease Inhibitors pharmacology
- Abstract
The polypeptide proteinase inhibitors (PI) isolated from Cucurbitaceae plants, bovine pancreas, lima bean kale seeds and some other sources were assayed in mouse, rat and human cell cultures for possible mitogenic effects. PI stimulated the proliferation of the spare cultures of Balb/c 3T3 and NRK-49F fibroblasts pretreated with trypsin. PI had no effect on [3H]thymidine incorporation by the monolayer cultures of the mouse or rat cells. Also certain human cell lines were unresponsive to PI. The results suggest that PI have growth factor-like activities which are cell and/or species specific.
- Published
- 1988
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.