Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Waldemar Popik is active.

Publication


Featured researches published by Waldemar Popik.


Journal of Virology | 2009

Exosomes Packaging APOBEC3G Confer Human Immunodeficiency Virus Resistance to Recipient Cells

Atanu K. Khatua; Harry E. Taylor; James E. K. Hildreth; Waldemar Popik

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.


Virology | 2010

Inhibition of LINE-1 and Alu retrotransposition by exosomes encapsidating APOBEC3G and APOBEC3F.

Atanu K. Khatua; Harry E. Taylor; James E. K. Hildreth; Waldemar Popik

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.


American Journal of Physiology-cell Physiology | 2015

Exon 4-encoded sequence is a major determinant of cytotoxicity of apolipoprotein L1

Atanu K. Khatua; Amber M. Cheatham; Etty Kruzel; Pravin C. Singhal; Karl Skorecki; Waldemar Popik

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.


Journal of Virology | 2011

Sterol Regulatory Element-Binding Protein 2 Couples HIV-1 Transcription to Cholesterol Homeostasis and T Cell Activation

Harry E. Taylor; Michael E. Linde; Atanu K. Khatua; Waldemar Popik; James E. K. Hildreth

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.


Virology | 2010

Non-productive HIV-1 infection of human glomerular and urinary podocytes.

Atanu K. Khatua; Harry E. Taylor; James E. K. Hildreth; Waldemar Popik

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.


American Journal of Pathology | 2016

HIV Promotes NLRP3 Inflammasome Complex Activation in Murine HIV-Associated Nephropathy

Shabirul Haque; Xiqian Lan; Hongxiu Wen; Rivka Lederman; Amrita Chawla; Mohamed Attia; Ramchandra P. Bongu; Mohammad Husain; Joanna Mikulak; Moin A. Saleem; Waldemar Popik; Ashwani Malhotra; Praveen N. Chander; Pravin C. Singhal

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 Txa0lymphocytes through induction of Nod-like receptor protein 3 (NLRP3) inflammasome complexes. We evaluated thexa0role of HIV in podocyte NLRP3 inflammasome formation both inxa0vivo and inxa0vitro. Renal cortical sections of HIV-transgenic mice (Tg26) displayed increased expression of NLRP3, ASC (axa0CARD protein), caspase-1, and IL-1β proteins, confirming NLRP3 inflammasome complex formation in podocytes ofxa0Tg26 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 (axa0superoxide dismutase-mimetic agent) and by glyburide (an inhibitor of potassium efflux). Thesexa0findings suggest that generation of reactive oxygen species and potassium efflux contribute to HIV-induced pyroptosis and NLRP3 inflammasome activation in podocytes.


Experimental and Molecular Pathology | 2015

Protein domains of APOL1 and its risk variants.

Xiqian Lan; Hongxiu Wen; Rivka Lederman; Ashwani Malhotra; Joanna Mikulak; Waldemar Popik; Karl Skorecki; Pravin C. Singhal

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.


PLOS Pathogens | 2015

Phospholipase D1 Couples CD4+ T Cell Activation to c-Myc-Dependent Deoxyribonucleotide Pool Expansion and HIV-1 Replication

Harry E. Taylor; Glenn E. Simmons; Thomas P. Mathews; Atanu K. Khatua; Waldemar Popik; Craig W. Lindsley; Richard T. D’Aquila; H. Alex Brown

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.


Bioscience Reports | 2017

Effect of APOL1 disease risk variants on APOL1 gene product

Shabirul Haque; Gauri Patil; Abheepsa Mishra; Xiqian Lan; Waldemar Popik; Ashwani Malhotra; Karl Skorecki; Pravin C. Singhal

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.


Virology | 2018

Phosphorodiamidate morpholino targeting the 5′ untranslated region of the ZIKV RNA inhibits virus replication

Waldemar Popik; Atanu K. Khatua; James E. K. Hildreth; Benjamin Lee; Donald J. Alcendor

BACKGROUNDnZika 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.nnnMETHODSnMorpholino 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.nnnRESULTSnPodocytes 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.nnnCONCLUSIONSnMorpholino DWK-1 targeting the ZIKV 5-UTR effectively inhibits ZIKV replication and suppresses ZIKV-induced proinflammatory gene expression.

Collaboration


Dive into the Waldemar Popik's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Pravin C. Singhal

The Feinstein Institute for Medical Research

View shared research outputs
Top Co-Authors

Avatar

Ashwani Malhotra

The Feinstein Institute for Medical Research

View shared research outputs
Top Co-Authors

Avatar

Xiqian Lan

The Feinstein Institute for Medical Research

View shared research outputs
Top Co-Authors

Avatar

Karl Skorecki

Technion – Israel Institute of Technology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Kamesh Ayasolla

The Feinstein Institute for Medical Research

View shared research outputs
Top Co-Authors

Avatar

Nitpriya Paliwal

The Feinstein Institute for Medical Research

View shared research outputs
Top Co-Authors

Avatar

Rukhsana Aslam

The Feinstein Institute for Medical Research

View shared research outputs
Researchain Logo
Decentralizing Knowledge