Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
Add more filters










Publication year range
1.
Biochim Biophys Acta Mol Basis Dis ; 1870(5): 167155, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38579939

ABSTRACT

Tubular proteinuria is a common feature in COVID-19 patients, even in the absence of established acute kidney injury. SARS-CoV-2 spike protein (S protein) was shown to inhibit megalin-mediated albumin endocytosis in proximal tubule epithelial cells (PTECs). Angiotensin-converting enzyme type 2 (ACE2) was not directly involved. Since Toll-like receptor 4 (TLR4) mediates S protein effects in various cell types, we hypothesized that TLR4 could be participating in the inhibition of PTECs albumin endocytosis elicited by S protein. Two different models of PTECs were used: porcine proximal tubule cells (LLC-PK1) and human embryonic kidney cells (HEK-293). S protein reduced Akt activity by specifically inhibiting of threonine 308 (Thr308) phosphorylation, a process mediated by phosphoinositide-dependent kinase 1 (PDK1). GSK2334470, a PDK1 inhibitor, decreased albumin endocytosis and megalin expression mimicking S protein effect. S protein did not change total TLR4 expression but decreased its surface expression. LPS-RS, a TLR4 antagonist, also counteracted the effects of the S protein on Akt phosphorylation at Thr308, albumin endocytosis, and megalin expression. Conversely, these effects of the S protein were replicated by LPS, an agonist of TLR4. Incubation of PTECs with a pseudovirus containing S protein inhibited albumin endocytosis. Null or VSV-G pseudovirus, used as control, had no effect. LPS-RS prevented the inhibitory impact of pseudovirus containing the S protein on albumin endocytosis but had no influence on virus internalization. Our findings demonstrate that the inhibitory effect of the S protein on albumin endocytosis in PTECs is mediated through TLR4, resulting from a reduction in megalin expression.


Subject(s)
Endocytosis , Kidney Tubules, Proximal , SARS-CoV-2 , Spike Glycoprotein, Coronavirus , Toll-Like Receptor 4 , Toll-Like Receptor 4/metabolism , Endocytosis/drug effects , Humans , Kidney Tubules, Proximal/metabolism , Kidney Tubules, Proximal/virology , Animals , Spike Glycoprotein, Coronavirus/metabolism , SARS-CoV-2/metabolism , HEK293 Cells , Swine , Proto-Oncogene Proteins c-akt/metabolism , Phosphorylation , COVID-19/metabolism , COVID-19/virology , COVID-19/pathology , Albumins/metabolism , LLC-PK1 Cells , Epithelial Cells/metabolism , Epithelial Cells/virology
2.
J Virol ; 98(3): e0180223, 2024 Mar 19.
Article in English | MEDLINE | ID: mdl-38334329

ABSTRACT

With a high incidence of acute kidney injury among hospitalized COVID-19 patients, considerable attention has been focussed on whether SARS-CoV-2 specifically targets kidney cells to directly impact renal function, or whether renal damage is primarily an indirect outcome. To date, several studies have utilized kidney organoids to understand the pathogenesis of COVID-19, revealing the ability for SARS-CoV-2 to predominantly infect cells of the proximal tubule (PT), with reduced infectivity following administration of soluble ACE2. However, the immaturity of standard human kidney organoids represents a significant hurdle, leaving the preferred SARS-CoV-2 processing pathway, existence of alternate viral receptors, and the effect of common hypertensive medications on the expression of ACE2 in the context of SARS-CoV-2 exposure incompletely understood. Utilizing a novel kidney organoid model with enhanced PT maturity, genetic- and drug-mediated inhibition of viral entry and processing factors confirmed the requirement for ACE2 for SARS-CoV-2 entry but showed that the virus can utilize dual viral spike protein processing pathways downstream of ACE2 receptor binding. These include TMPRSS- and CTSL/CTSB-mediated non-endosomal and endocytic pathways, with TMPRSS10 likely playing a more significant role in the non-endosomal pathway in renal cells than TMPRSS2. Finally, treatment with the antihypertensive ACE inhibitor, lisinopril, showed negligible impact on receptor expression or susceptibility of renal cells to infection. This study represents the first in-depth characterization of viral entry in stem cell-derived human kidney organoids with enhanced PTs, providing deeper insight into the renal implications of the ongoing COVID-19 pandemic. IMPORTANCE: Utilizing a human iPSC-derived kidney organoid model with improved proximal tubule (PT) maturity, we identified the mechanism of SARS-CoV-2 entry in renal cells, confirming ACE2 as the sole receptor and revealing redundancy in downstream cell surface TMPRSS- and endocytic Cathepsin-mediated pathways. In addition, these data address the implications of SARS-CoV-2 exposure in the setting of the commonly prescribed ACE-inhibitor, lisinopril, confirming its negligible impact on infection of kidney cells. Taken together, these results provide valuable insight into the mechanism of viral infection in the human kidney.


Subject(s)
Angiotensin-Converting Enzyme 2 , Kidney , Organoids , SARS-CoV-2 , Virus Internalization , Humans , Angiotensin-Converting Enzyme 2/metabolism , COVID-19/complications , COVID-19/virology , Kidney/cytology , Kidney/drug effects , Kidney/metabolism , Kidney/virology , Lisinopril/pharmacology , Lisinopril/metabolism , Organoids/cytology , Organoids/drug effects , Organoids/metabolism , Organoids/virology , Pandemics , SARS-CoV-2/metabolism , SARS-CoV-2/pathogenicity , Spike Glycoprotein, Coronavirus/metabolism , Virus Internalization/drug effects , Peptidyl-Dipeptidase A/metabolism , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Acute Kidney Injury/etiology , Acute Kidney Injury/metabolism , Acute Kidney Injury/virology , Kidney Tubules, Proximal/cytology , Kidney Tubules, Proximal/drug effects , Kidney Tubules, Proximal/metabolism , Kidney Tubules, Proximal/virology , Receptors, Coronavirus/metabolism , Models, Biological , Serine Endopeptidases/metabolism , Endosomes/drug effects , Endosomes/metabolism , Endosomes/virology , Gene Expression Regulation/drug effects , Stem Cells/cytology
3.
JCI Insight ; 6(24)2021 12 22.
Article in English | MEDLINE | ID: mdl-34767537

ABSTRACT

Kidneys are critical target organs of COVID-19, but susceptibility and responses to infection remain poorly understood. Here, we combine SARS-CoV-2 variants with genome-edited kidney organoids and clinical data to investigate tropism, mechanism, and therapeutics. SARS-CoV-2 specifically infects organoid proximal tubules among diverse cell types. Infections produce replicating virus, apoptosis, and disrupted cell morphology, features of which are revealed in the context of polycystic kidney disease. Cross-validation of gene expression patterns in organoids reflects proteomic signatures of COVID-19 in the urine of critically ill patients indicating interferon pathway upregulation. SARS-CoV-2 viral variants alpha, beta, gamma, kappa, and delta exhibit comparable levels of infection in organoids. Infection is ameliorated in ACE2-/- organoids and blocked via treatment with de novo-designed spike binder peptides. Collectively, these studies clarify the impact of kidney infection in COVID-19 as reflected in organoids and clinical populations, enabling assessment of viral fitness and emerging therapies.


Subject(s)
Acute Kidney Injury/urine , COVID-19/urine , Kidney Tubules, Proximal/virology , Kidney/virology , Organoids/virology , SARS-CoV-2/pathogenicity , Acute Kidney Injury/etiology , Adult , Aged , Angiotensin-Converting Enzyme 2/genetics , Animals , Apoptosis , Bowman Capsule/cytology , Bowman Capsule/virology , COVID-19/complications , Chlorocebus aethiops , Female , Gene Knockout Techniques , Hospital Mortality , Hospitalization , Humans , Kidney/metabolism , Kidney/pathology , Kidney Tubules, Proximal/metabolism , Kidney Tubules, Proximal/pathology , Male , Middle Aged , Organoids/metabolism , Podocytes/virology , Polycystic Kidney Diseases , Protein Kinase D2/genetics , Proteome , Receptors, Coronavirus/genetics , Reproducibility of Results , Transcriptome , Vero Cells , Viral Tropism , Virus Replication
4.
Eur J Pharmacol ; 888: 173487, 2020 Dec 05.
Article in English | MEDLINE | ID: mdl-32805256

ABSTRACT

Acute kidney injury (AKI) is an important complication of COVID-19 encompassing a wide range of presentations. SARS-CoV-2 is proposed to cause AKI in the patients through various mechanisms. We are, nevertheless, far from a comprehensive understanding of the underlying pathophysiological mechanisms of the kidney injury in this infection. AKI has been shown to be a marker of disease severity and also a negative prognostic factor for survival. Unfortunately, no effective preventive strategy to decrease the risk of kidney damage in these patients has yet been identified. In this hypothesis, we highlight the potential protective effects of acetazolamide, a carbonic anhydrase inhibitor, in preventing the proximal tubular damage caused by the virus through disrupting the virus-endosome fusion and also interfering with the lysosomal proteases. Our proposed mechanisms could pave the way for further in vitro studies and subsequent clinical trials.


Subject(s)
Acetazolamide/therapeutic use , Acute Kidney Injury/etiology , Acute Kidney Injury/prevention & control , Carbonic Anhydrase Inhibitors/therapeutic use , Coronavirus Infections/complications , Pneumonia, Viral/complications , Angiotensin-Converting Enzyme 2 , COVID-19 , Humans , Kidney Tubules, Proximal/cytology , Kidney Tubules, Proximal/virology , Pandemics , Peptidyl-Dipeptidase A/metabolism , Treatment Outcome
5.
Mol Genet Genomic Med ; 8(10): e1442, 2020 10.
Article in English | MEDLINE | ID: mdl-32744436

ABSTRACT

BACKGROUND: A novel coronavirus called SARS-Cov-2, which shared 82% similarity of genome sequence with SARS-CoV, was found in Wuhan in late December of 2019, causing an epidemic outbreak of novel coronavirus-induced pneumonia with dramatically increasing number of cases. Several organs are vulnerable to COVID-19 infection. Acute kidney injury (AKI) was reported in parts of case-studies reporting characteristics of COVID-19 patients. This study aimed at analyzing the potential route of SARS-Cov-2 entry and mechanism at cellular level. METHOD: Single-cell RNA sequencing (scRNA-seq) technology was used to obtain evidence of potential route and ACE2 expressing cell in renal system for underlying pathogenesis of kidney injury caused by COVID-19. The whole process was performed under R with Seurat packages. Canonical marker genes were used to annotate different types of cells. RESULTS: Ten different clusters were identified and ACE2 was mainly expressed in proximal tubule and glomerular parietal epithelial cells. From Gene Ontology (GO) & KEGG enrichment analysis, imbalance of ACE2 expression, renin-angiotensin system (RAS) activation, and neutrophil-related processes were the main issue of COVID-19 leading kidney injury. CONCLUSION: Our study provided the cellular evidence that SARS-Cov-2 invaded human kidney tissue via proximal convoluted tubule, proximal tubule, proximal straight tubule cells, and glomerular parietal cells by means of ACE2-related pathway and used their cellular protease TMPRSS2 for priming.


Subject(s)
Acute Kidney Injury/virology , Angiotensin-Converting Enzyme 2/metabolism , COVID-19/pathology , Kidney Glomerulus/metabolism , Kidney Tubules, Proximal/metabolism , Receptors, Virus/genetics , Acute Kidney Injury/pathology , Angiotensin-Converting Enzyme 2/genetics , Base Sequence , Humans , Kidney Glomerulus/pathology , Kidney Glomerulus/virology , Kidney Tubules, Proximal/pathology , Kidney Tubules, Proximal/virology , Principal Component Analysis , SARS-CoV-2/metabolism , Sequence Analysis, RNA , Serine Endopeptidases/metabolism , Single-Cell Analysis
6.
Int J Mol Sci ; 21(9)2020 May 05.
Article in English | MEDLINE | ID: mdl-32380787

ABSTRACT

Coronaviruses (CoVs), including Severe Acute Respiratory Syndrome (SARS), Middle East Respiratory Syndrome (MERS), and the novel coronavirus disease-2 (SARS-CoV-2) are a group of enveloped RNA viruses that cause a severe respiratory infection which is associated with a high mortality [...].


Subject(s)
Acute Kidney Injury/mortality , Acute Kidney Injury/virology , Betacoronavirus/pathogenicity , Coronavirus Infections/virology , Kidney Tubules, Proximal/metabolism , Kidney Tubules, Proximal/virology , Pneumonia, Viral/virology , Acute Kidney Injury/physiopathology , Acute Kidney Injury/prevention & control , Angiotensin II/pharmacology , Angiotensin-Converting Enzyme 2 , COVID-19 , Cathepsins/metabolism , Cell Death/drug effects , Coronavirus Infections/mortality , Coronavirus Infections/pathology , Coronavirus Infections/physiopathology , Creatinine/blood , Critical Illness/mortality , Endosomes/drug effects , Endosomes/enzymology , Endosomes/metabolism , Humans , Hydrogen-Ion Concentration , Incidence , Kidney Tubules, Proximal/physiopathology , Lysosomes/drug effects , Lysosomes/enzymology , Lysosomes/metabolism , Pandemics , Peptidyl-Dipeptidase A/metabolism , Pneumonia, Viral/mortality , Pneumonia, Viral/pathology , Pneumonia, Viral/physiopathology , Receptors, Virus/metabolism , SARS-CoV-2 , Severe Acute Respiratory Syndrome/blood , Severe Acute Respiratory Syndrome/mortality , Severe Acute Respiratory Syndrome/physiopathology , Virus Internalization , Virus Replication
7.
Biomaterials ; 219: 119367, 2019 10.
Article in English | MEDLINE | ID: mdl-31344514

ABSTRACT

Renal dysfunctions usually happen in viral infections and many viruses specially infect distal renal tubules, however the pathogenesis remains unknown. Here, in order to explore the pathogenesis of virus-related renal dysfunctions, a Pseudorabies Virus (PrV) induced kidney disease model was built on a distal tubule-on-a-chip (DTC), for the first time. The barrier structure and Na reabsorption of distal renal tubules were successfully reconstituted in DTCs. After PrV infection, results showed electrolyte regulation dysfunction in Na reabsorption for the disordered Na transporters, the broken reabsorption barrier, and the transformed microvilli. And it would lead to virus induced serum electrolyte abnormalities. This work brought us a new cognition about the advantages of organ-on-a-chip (OOC) in virus research, for it had given us a better insight into the pathogenesis of virus induced dysfunctions, based on its unique ability in function reproduction.


Subject(s)
Herpesvirus 1, Suid/physiology , Kidney Diseases/virology , Kidney/physiopathology , Kidney/virology , Lab-On-A-Chip Devices , Absorption, Physiological/drug effects , Angiotensin II/pharmacology , Animals , Cell Polarity/drug effects , Disease Models, Animal , Dogs , Herpesvirus 1, Suid/drug effects , Kidney/drug effects , Kidney/pathology , Kidney Tubules, Proximal/drug effects , Kidney Tubules, Proximal/pathology , Kidney Tubules, Proximal/physiopathology , Kidney Tubules, Proximal/virology , Madin Darby Canine Kidney Cells , Microfluidics , Sodium/metabolism
8.
Kidney Int ; 93(2): 355-364, 2018 02.
Article in English | MEDLINE | ID: mdl-29061332

ABSTRACT

Plasmacytoid dendritic cells (pDCs) are antigen presenting cells specialized in viral recognition through Toll-like receptor (TLR)7 and TLR9, and produce vast amounts of interferon alpha upon ligation of these TLRs. We had previously demonstrated a strong influx of pDCs in the tubulointerstitium of renal biopsies at the time of acute rejection. However, the role of human pDCs in mediating acute or chronic allograft rejection remains elusive. pDCs are thought to have a limited capacity to ingest apoptotic cells, critical for inducing CD4+ T cell activation via indirect antigen presentation and subsequent activation of antibody producing B cells. Here we tested whether the function of pDCs is affected by their presence within the graft. Maturation and interferon alpha production by pDCs was enhanced when cells were activated in the presence of viable HK2 renal epithelial cells. Importantly, soluble factors produced by cytomegalovirus-infected (primary) epithelial or endothelial cells enhanced pDC activation and induced their capacity to phagocytose apoptotic cells. Phagocytosis was not induced by free virus or soluble factors from non-infected cells. Activated pDCs showed an enhanced CD4+ and CD8+ T cell allostimulatory capacity as well as a potent indirect alloantigen presentation. Granulocyte Macrophage-Colony Stimulating Factor is one of the soluble factors produced by renal epithelial cells that, combined with TLR9 ligation, induced this functional capacity. Thus, pDCs present in the rejecting allograft can contribute to alloimmunity and potentially act as important orchestrators in the manifestation of acute and chronic rejection.


Subject(s)
Dendritic Cells/metabolism , Epithelial Cells/metabolism , Graft Rejection/metabolism , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Kidney Transplantation/adverse effects , Kidney Tubules, Proximal/metabolism , Paracrine Communication , Phagocytosis , Toll-Like Receptor 9/metabolism , Antigen Presentation , Apoptosis , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Line , Coculture Techniques , Cytomegalovirus/immunology , Cytomegalovirus/pathogenicity , Dendritic Cells/immunology , Epithelial Cells/immunology , Epithelial Cells/pathology , Epithelial Cells/virology , Graft Rejection/immunology , Graft Rejection/pathology , Graft Rejection/virology , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Host-Pathogen Interactions , Humans , Interferon-alpha/metabolism , Isoantigens/immunology , Isoantigens/metabolism , Kidney Tubules, Proximal/immunology , Kidney Tubules, Proximal/pathology , Kidney Tubules, Proximal/virology , Lymphocyte Activation , Phenotype , Signal Transduction , Toll-Like Receptor 9/immunology
9.
J Innate Immun ; 9(6): 574-586, 2017.
Article in English | MEDLINE | ID: mdl-28877527

ABSTRACT

Recognition of viral pathogen-associated molecular patterns by pattern recognition receptors (PRRs) is the first step in the initiation of a host innate immune response. As a PRR, RIG-I detects either viral RNA or replication transcripts. Avoiding RIG-I recognition is a strategy employed by viruses for immune evasion. Epstein-Barr virus (EBV) infects the majority of the human population worldwide. During the latent infection period there are only a few EBV proteins expressed, whereas EBV-encoded microRNAs, such as BART microRNAs, are highly expressed. BART microRNAs regulate both EBV and the host's gene expression, modulating virus proliferation and the immune response. Here, through gene expression profiling, we found that EBV miR-BART6-3ps inhibited genes of RIG-I-like receptor signaling and the type I interferon (IFN) response. We demonstrated that miR-BART6-3p rather than other BARTs specifically suppressed RIG-I-like receptor signaling-mediated IFN-ß production. RNA-seq was used to analyze the global transcriptome change upon EBV infection and miR-BART6-3p mimics transfection, which revealed that EBV infection-triggered immune response signaling can be repressed by miR-BART6-3p overexpression. Furthermore, miR-BART6-3p inhibited the EBV-triggered IFN-ß response and facilitated EBV infection through targeting the 3'UTR of RIG-I mRNA. These findings provide new insights into the mechanism underlying the strategies employed by EBV to evade immune surveillance.


Subject(s)
Epstein-Barr Virus Infections/immunology , Herpesvirus 4, Human/genetics , Kidney Tubules, Proximal/virology , MicroRNAs/genetics , RNA, Viral/genetics , Cell Line , Host-Pathogen Interactions , Humans , Immune Evasion , Immunity, Innate , Kidney Tubules, Proximal/immunology , Pathogen-Associated Molecular Pattern Molecules/immunology , Receptors, Pattern Recognition/metabolism , Receptors, Retinoic Acid/metabolism , Signal Transduction
10.
Emerg Microbes Infect ; 6(8): e77, 2017 Aug 23.
Article in English | MEDLINE | ID: mdl-28831192

ABSTRACT

Zika virus (ZIKV) infection can cause fetal developmental abnormalities and Guillain-Barré syndrome in adults. Although progress has been made in understanding the link between ZIKV infection and microcephaly, the pathology of ZIKV, particularly the viral reservoirs in human, remains poorly understood. Several studies have shown that compared to serum samples, patients' urine samples often have a longer duration of ZIKV persistency and higher viral load. This finding suggests that an independent viral reservoir may exist in the human urinary system. Despite the clinical observations, the host cells of ZIKV in the human urinary system are poorly characterized. In this study, we demonstrate that ZIKV can infect renal proximal tubular epithelial cells (RPTEpiCs) in immunodeficient mice in vivo and in both immortalized and primary human renal proximal tubular epithelial cells (hRPTEpiCs) in vitro. Importantly, ZIKV infection in mouse kidneys caused caspase-3-mediated apoptosis of renal cells. Similarly, in vitro infection of immortalized and primary hRPTEpiCs resulted in notable cytopathic effects. Consistent with the clinical observations, we found that ZIKV infection can persist with prolonged duration in hRPTEpiCs. RNA-Seq analyses of infected hRPTEpiCs revealed a large number of transcriptional changes in response to ZIKV infection, including type I interferon signaling genes and anti-viral response genes. Our results suggest that hRPTEpiCs are a potential reservoir of ZIKV in the human urinary system, providing a possible explanation for the prolonged persistency of ZIKV in patients' urine.


Subject(s)
Kidney Tubules, Proximal/pathology , Kidney Tubules, Proximal/virology , Urothelium/virology , Zika Virus Infection/virology , Zika Virus/physiology , Animals , Apoptosis , Cell Line, Tumor , Cytopathogenic Effect, Viral , Disease Models, Animal , Disease Reservoirs/virology , High-Throughput Nucleotide Sequencing , Humans , Interferon Type I/genetics , Interferon Type I/metabolism , Kidney/pathology , Kidney/virology , Kidney Tubules, Proximal/cytology , Mice , Mice, Inbred C57BL , Urine/virology , Urothelium/cytology , Viral Load , Virus Replication , Zika Virus/isolation & purification , Zika Virus Infection/pathology
11.
Virology ; 492: 66-72, 2016 May.
Article in English | MEDLINE | ID: mdl-26901486

ABSTRACT

BK polyomavirus (BKPyV) is a human pathogen that causes polyomavirus-associated nephropathy and hemorrhagic cystitis in transplant patients. Gangliosides and caveolin proteins have previously been reported to be required for BKPyV infection in animal cell models. Recent studies from our lab and others, however, have indicated that the identity of the cells used for infection studies can greatly influence the behavior of the virus. We therefore wished to re-examine BKPyV entry in a physiologically relevant primary cell culture model, human renal proximal tubule epithelial cells. Using siRNA knockdowns, we interfered with expression of UDP-glucose ceramide glucosyltransferase (UGCG), and the endocytic vesicle coat proteins caveolin 1, caveolin 2, and clathrin heavy chain. The results demonstrate that while BKPyV does require gangliosides for efficient infection, it can enter its natural host cells via a caveolin- and clathrin-independent pathway. The results emphasize the importance of studying viruses in a relevant cell culture model.


Subject(s)
BK Virus/drug effects , Caveolin 1/genetics , Caveolin 2/genetics , Clathrin Heavy Chains/genetics , Epithelial Cells/drug effects , Host-Pathogen Interactions , BK Virus/genetics , BK Virus/metabolism , Caveolin 1/antagonists & inhibitors , Caveolin 1/metabolism , Caveolin 2/antagonists & inhibitors , Caveolin 2/metabolism , Cell Line , Clathrin Heavy Chains/antagonists & inhibitors , Clathrin Heavy Chains/metabolism , Epithelial Cells/virology , G(M1) Ganglioside/pharmacology , Gangliosides/pharmacology , Gene Expression Regulation , Humans , Kidney Tubules, Proximal/drug effects , Kidney Tubules, Proximal/virology , MicroRNAs/genetics , MicroRNAs/metabolism , Monosaccharide Transport Proteins/antagonists & inhibitors , Monosaccharide Transport Proteins/genetics , Monosaccharide Transport Proteins/metabolism , Primary Cell Culture , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Virus Internalization/drug effects
12.
J Proteome Res ; 14(10): 4413-24, 2015 Oct 02.
Article in English | MEDLINE | ID: mdl-26354146

ABSTRACT

Polyomaviruses are a family of small DNA viruses that are associated with a number of severe human diseases, particularly in immunocompromised individuals. The detailed virus-host interactions during lytic polyomavirus infection are not fully understood. Here, we report the first nuclear proteomic study with BK polyomavirus (BKPyV) in a primary renal proximal tubule epithelial cell culture system using stable isotope labeling by amino acids in cell culture (SILAC) proteomic profiling coupled with liquid chromatography-tandem mass spectrometry. We demonstrated the feasibility of SILAC labeling in these primary cells and subsequently performed reciprocal labeling-infection experiments to identify proteins that are altered by BKPyV infection. Our analyses revealed specific proteins that are significantly up- or down-regulated in the infected nuclear proteome. The genes encoding many of these proteins were not identified in a previous microarray study, suggesting that differential regulation of these proteins may be independent of transcriptional control. Western blotting experiments verified the SILAC proteomic findings. Finally, pathway and network analyses indicated that the host cell DNA damage response signaling and DNA repair pathways are among the cellular processes most affected at the protein level during polyomavirus infection. Our study provides a comprehensive view of the host nuclear proteomic changes during polyomavirus lytic infection and suggests potential novel host factors required for a productive polyomavirus infection.


Subject(s)
BK Virus/physiology , Cell Nucleus/metabolism , DNA Repair , Epithelial Cells/metabolism , Proteome/metabolism , Cell Nucleus/chemistry , Cell Nucleus/pathology , Cell Nucleus/virology , Chromatography, Liquid , DNA Damage , Epithelial Cells/pathology , Epithelial Cells/virology , Gene Expression Regulation , Gene Regulatory Networks , Host-Pathogen Interactions , Humans , Isotope Labeling , Kidney Tubules, Proximal/metabolism , Kidney Tubules, Proximal/pathology , Kidney Tubules, Proximal/virology , Molecular Sequence Annotation , Primary Cell Culture , Proteome/genetics , Proteome/isolation & purification , Signal Transduction , Tandem Mass Spectrometry , Transcription, Genetic
13.
Virology ; 474: 110-6, 2015 Jan 01.
Article in English | MEDLINE | ID: mdl-25463609

ABSTRACT

BK Polyomavirus (BKPyV) is a ubiquitous nonenveloped human virus that can cause severe disease in immunocompromised populations. After internalization into renal proximal tubule epithelial cells, BKPyV traffics through the ER and enters the cytosol. However, it is unclear how the virus enters the nucleus. In this study, we elucidate a role for the nuclear localization signal located on the minor capsid proteins VP2 and VP3 during infection. Site-directed mutagenesis of a single lysine in the basic region of the C-terminus of the minor capsid proteins abrogated their nuclear localization, and the analogous genomic mutation reduced infectivity. Additionally, through use of the inhibitor ivermectin and knockdown of importin ß1, we found that the importin α/ß pathway is involved during infection. Overall these data are the first to show the significance of the NLS of the BKPyV minor capsid proteins during infection in a natural host cell.


Subject(s)
BK Virus/physiology , Capsid Proteins/physiology , Nuclear Localization Signals/physiology , Active Transport, Cell Nucleus/drug effects , Active Transport, Cell Nucleus/physiology , Amino Acid Sequence , Amino Acid Substitution , BK Virus/genetics , BK Virus/pathogenicity , Capsid Proteins/chemistry , Capsid Proteins/genetics , Cells, Cultured , Gene Knockdown Techniques , Host-Pathogen Interactions , Humans , Ivermectin/pharmacology , Kidney Tubules, Proximal/virology , Lysine/chemistry , Molecular Sequence Data , Mutagenesis, Site-Directed , Nuclear Localization Signals/chemistry , Nuclear Localization Signals/genetics , Sequence Homology, Amino Acid , Virus Internalization , beta Karyopherins/antagonists & inhibitors , beta Karyopherins/genetics , beta Karyopherins/physiology
14.
Int J Mol Med ; 31(5): 1017-29, 2013 May.
Article in English | MEDLINE | ID: mdl-23483208

ABSTRACT

Hepatitis B virus X protein (HBx) is a multifunctional protein, and it activates multiple signal transduction pathways in multiple types of cells and regulates the process of cell apoptosis. In the present study, we mainly investigated the correlation between HBx and renal tubular epithelial cell apoptosis in hepatitis B virus-associated glomerulonephritis (HBVGN) and the possible signaling mechanism. Cell apoptosis in nephridial tissues of patients with HBVGN were determined by the TUNEL method. HBx, p-STAT3 and STAT3 levels in nephridial tissues were determined by immunohistochemical assay, and a correlation analysis between HBx expression levels and apoptosis index in nephridial tissues was conducted. The activation of the JAK2/STAT3 signaling pathway in HK-2 cells and the expression of the apoptosis-related proteins Bax and Bcl-2 were determined by western blot analysis following transfection with the HBx eukaryotic expression vector. Cellular proliferation activity was determined by the CCK­8 method, and cell apoptosis was determined with HO33342 staining using transmission electron microscopy and Annexin V/PI double staining flow cytometry. The results revealed that the apoptosis index in nephridial tissues of patients with HBVGN was significantly higher when compared to that of the control group, and p-STAT3 expression levels in HBVGN nephridial tissues were significantly increased. In the control group, no HBx expression was observed in the nephridial tissues, whereas HBx expression was found in the nephridial tissues of 86% of the patients with HBVGN. The HBx expression levels had a linear correlation with the apoptosis index in the nephridial tissues. After target gene HBx infection, expression levels of both p-JAK2 and p-STAT3 in human proximal HK-2 cells were significantly increased, and the Bax/Bcl-2 ratio was also significantly increased. At the same time, cellular proliferation of HK-2 cells was significantly inhibited, and the rate of apoptosis was increased. After incubation with AG490, the JAK2/STAT3 signaling pathway was partially blocked, which caused a decrease in the Bax/Bcl-2 ratio and reduced cell apoptosis caused by HBx. In conclusion, HBx upregulates the Bax/Bcl-2 ratio by activating the JAK2/STAT3 signaling pathway to cause renal tubular epithelial cell apoptosis, and it is possibly involved in the pathogenic mechanism of nephridial tissue damage caused by HBV.


Subject(s)
Apoptosis , Janus Kinase 2/metabolism , Kidney Tubules, Proximal/metabolism , Kidney Tubules, Proximal/virology , STAT3 Transcription Factor/metabolism , Signal Transduction , Trans-Activators/metabolism , Adolescent , Adult , Blotting, Western , Cell Nucleus/metabolism , Cell Nucleus/ultrastructure , Cell Proliferation , Cell Survival , Fluorescent Antibody Technique , Glomerulonephritis/metabolism , Glomerulonephritis/pathology , Glomerulonephritis/virology , Humans , Kidney Tubules, Proximal/pathology , Kidney Tubules, Proximal/ultrastructure , Middle Aged , Phosphorylation , Transfection , Viral Regulatory and Accessory Proteins , Young Adult , bcl-2-Associated X Protein/metabolism
15.
Antiviral Res ; 92(1): 115-23, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21798289

ABSTRACT

Reactivation of human polyomavirus BK (BKV) may cause polyomavirus-associated nephropathy or polyomavirus-associated hemorrhagic cystitis in renal- or bone marrow-transplant patients, respectively. Lack of treatment options has led to exploration of fluoroquinolones that inhibit topoisomerase II and IV in prokaryotes and possibly large T-antigen (LT-ag) helicase activity in polyomavirus. We characterized the effects of ofloxacin and levofloxacin on BKV replication in the natural host cells - primary human renal proximal tubular epithelial cells (RPTECs). Ofloxacin and levofloxacin inhibited BKV load in a dose-dependent manner yielding a ∼90% inhibition at 150 µg/ml. Ofloxacin at 150 µg/ml inhibited LT-ag mRNA and protein expression from 24h post infection (hpi). BKV genome replication was 77% reduced at 48 hpi and a similar reduction was found in VP1 and agnoprotein expression. At 72 hpi, the reduction in genome replication and protein expression was less pronounced. A dose-dependent cytostatic effect was noted. In infected cells, 150 µg/ml ofloxacin led to a 26% and 6% inhibition of cellular DNA replication and total metabolic activity, respectively while 150 µg/ml levofloxacin affected this slightly more, particularly in uninfected cells. Cell counting and xCELLigence results revealed that cell numbers were not reduced. In conclusion, ofloxacin and levofloxacin inhibit but do not eradicate BKV replication in RPTECs. At a concentration of ofloxacin giving ∼90% inhibition in BKV load, no significant cytotoxicity was observed. This concentration can be achieved in urine and possibly in the kidneys. Our results support a mechanism involving inhibition of LT-ag expression or functions but also suggest inhibition of cellular enzymes.


Subject(s)
Antiviral Agents/pharmacology , BK Virus/drug effects , BK Virus/physiology , Down-Regulation/drug effects , Fluoroquinolones/pharmacology , Polyomavirus Infections/virology , Virus Replication/drug effects , BK Virus/genetics , Cell Line , Cells, Cultured , Epithelial Cells/virology , Gene Expression Regulation, Viral/drug effects , Humans , Kidney Tubules, Proximal/cytology , Kidney Tubules, Proximal/virology
16.
J Am Soc Nephrol ; 22(3): 496-507, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21335514

ABSTRACT

In animal models of HIV-associated nephropathy, the expression of HIV regulatory genes in epithelial cells is sufficient to cause disease, but how the CD4-negative epithelial cells come to express HIV genes is unknown. Here, we co-cultured T cells infected with fluorescently tagged HIV with renal tubular epithelial cells and observed efficient virus transfer between these cells. The quantity of HIV transferred was much greater than that achieved by exposure to large amounts of cell-free virus and occurred without a requirement for CD4 or Env. The transfer required stable cell-cell adhesion, which could be blocked by sulfated polysaccharides or poly-anionic compounds. We found that the internalization of virus could lead to de novo synthesis of viral protein from incoming viral RNAs even in the presence of a reverse transcriptase inhibitor. These results illustrate an interaction between infected T cells and nonimmune cells, supporting the presence of virological synapses between HIV-harboring T cells and renal tubular epithelial cells, allowing viral uptake and gene expression in epithelial cells.


Subject(s)
Cell Communication/physiology , Epithelial Cells/virology , Gene Expression Regulation, Viral/physiology , HIV-1/physiology , Kidney Tubules, Proximal/virology , AIDS-Associated Nephropathy/etiology , AIDS-Associated Nephropathy/pathology , AIDS-Associated Nephropathy/physiopathology , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/virology , Cell Adhesion/drug effects , Cell Adhesion/physiology , Cell Communication/drug effects , Cell Line , Coculture Techniques , Epithelial Cells/cytology , HIV-1/genetics , Humans , Kidney Tubules, Proximal/cytology , Polysaccharides/pharmacology , Virus Replication/physiology
17.
PLoS Pathog ; 6(11): e1001170, 2010 Nov 04.
Article in English | MEDLINE | ID: mdl-21079788

ABSTRACT

Human cytomegalovirus (HCMV) infection is associated epidemiologically with poor outcome of renal allografts due to mechanisms which remain largely undefined. Transforming growth factor-ß1 (TGF-ß1), a potent fibrogenic cytokine, is more abundant in rejecting renal allografts that are infected with either HCMV or rat CMV as compared to uninfected, rejecting grafts. TGF-ß1 induces renal fibrosis via epithelial-to-mesenchymal transition (EMT) of renal epithelial cells, a process by which epithelial cells acquire mesenchymal characteristics and a migratory phenotype, and secrete molecules associated with extracellular matrix deposition and remodeling. We report that human renal tubular epithelial cells infected in vitro with HCMV and exposed to TGF-ß1 underwent morphologic and transcriptional changes of EMT, similar to uninfected cells. HCMV infected cells after EMT also activated extracellular latent TGF-ß1 via induction of MMP-2. Renal epithelial cells transiently transfected with only the HCMV IE1 or IE2 open reading frames and stimulated to undergo EMT also induced TGF-ß1 activation associated with MMP-2 production, suggesting a role for these viral gene products in MMP-2 production. Consistent with the function of these immediate early gene products, the antiviral agents ganciclovir and foscarnet did not inhibit TGF-ß1 production after EMT by HCMV infected cells. These results indicate that HCMV infected renal tubular epithelial cells can undergo EMT after exposure to TGF-ß1, similar to uninfected renal epithelial cells, but that HCMV infection by inducing active TGF-ß1 may potentiate renal fibrosis. Our findings provide in vitro evidence for a pathogenic mechanism that could explain the clinical association between HCMV infection, TGF-ß1, and adverse renal allograft outcome.


Subject(s)
Cytomegalovirus Infections/metabolism , Cytomegalovirus/pathogenicity , Kidney Tubules, Proximal/metabolism , Kidney Tubules, Proximal/virology , Transforming Growth Factor beta1/metabolism , Animals , Blotting, Western , Cell Differentiation , Cells, Cultured , Cytomegalovirus Infections/pathology , Cytomegalovirus Infections/virology , Epithelial Cells/metabolism , Epithelial Cells/virology , Epithelial-Mesenchymal Transition , Fluorescent Antibody Technique , Humans , Immunoprecipitation , Kidney Tubules, Proximal/cytology , RNA, Messenger/genetics , Rats , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction
18.
Virology ; 407(2): 368-73, 2010 Nov 25.
Article in English | MEDLINE | ID: mdl-20869740

ABSTRACT

The human polyomavirus BK virus (BKV) is an important opportunistic pathogen whose disease prevalence continues to increase with the growing immunocompromised population. To date, the major determinant of replication in cell culture has not been formally proven. BKV exists as archetype virus and rearranged variants, which are classified based on the DNA sequence of their non-coding control regions (NCCRs). The archetype BKV NCCR is divided into five blocks of sequence and rearranged variants contain deletions and duplications of these blocks. In this study, a genetic system was developed and used to identify the major determinant of replication ability in primary renal proximal tubule epithelial cells, the natural host cell of BKV. This system was also used to analyze NCCR variants isolated from an immunocompromised patient which contain assorted rearrangement patterns and functional differences. This study solidifies the NCCR as the major genetic determinant of BKV replication ability in vitro.


Subject(s)
BK Virus/isolation & purification , Genetic Variation , HIV Infections/complications , Polyomavirus Infections/virology , Regulatory Sequences, Nucleic Acid/genetics , Tumor Virus Infections/virology , AIDS-Related Opportunistic Infections/virology , BK Virus/classification , BK Virus/genetics , Cells, Cultured , DNA, Viral/analysis , DNA, Viral/isolation & purification , Epithelial Cells/virology , HIV Infections/virology , Humans , Immunocompromised Host , Kidney Tubules, Proximal/cytology , Kidney Tubules, Proximal/virology , Sequence Analysis, DNA , Urine/virology , Virology/methods , Virus Replication
19.
Curr Protoc Cell Biol ; Chapter 26: Unit 26.2, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19283732

ABSTRACT

BK virus (BKV) can cause BKV nephritis in renal transplant patients and has become a significant reason for graft loss in this decade. BKV is latent in the urogenital tract and most likely is transported with the donor kidney to recipients. BKV replication occurs in the nucleus of human renal proximal tubular cells (HRPTEC) and daughter viruses are delivered to other cells to spread infection. A few in vitro studies have been reported about the mechanism and kinetics of BKV infection. However, there are still a lot of unknown factors regarding BKV infection. This unit describes the handling of BKV, BKV propagation, determination of titer and ability to infect cells, as well as purification and labeling of BKV in order to analyze BKV cell entry.


Subject(s)
BK Virus , Polyomavirus Infections/virology , Tumor Virus Infections/virology , BK Virus/isolation & purification , BK Virus/pathogenicity , BK Virus/physiology , Cell Culture Techniques , Epithelial Cells/metabolism , Epithelial Cells/pathology , Epithelial Cells/virology , Fluorescent Dyes/metabolism , Humans , Kidney Tubules, Proximal/metabolism , Kidney Tubules, Proximal/pathology , Kidney Tubules, Proximal/virology , Microscopy, Confocal , Polyomavirus Infections/genetics , Polyomavirus Infections/metabolism , Protein Transport , Tumor Virus Infections/genetics , Tumor Virus Infections/metabolism , Tumor Virus Infections/pathology , Viral Proteins/metabolism , Virology/methods , Virus Internalization , Virus Replication
20.
J Virol ; 83(3): 1350-8, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19036822

ABSTRACT

BK virus (BKV) is a nonenveloped, ubiquitous human polyomavirus that establishes a persistent infection in healthy individuals. It can be reactivated, however, in immunosuppressed patients and cause severe diseases, including polyomavirus nephropathy. The entry and disassembly mechanisms of BKV are not well defined. In this report, we characterized several early events during BKV infection in primary human renal proximal tubule epithelial (RPTE) cells, which are natural host cells for BKV. Our results demonstrate that BKV infection in RPTE cells involves an acidic environment relatively early during entry, followed by transport along the microtubule network to reach the endoplasmic reticulum (ER). A distinct disulfide bond isomerization and cleavage pattern of the major capsid protein VP1 was observed, which was also influenced by alterations in pH and disruption of trafficking to the ER. A dominant negative form of Derlin-1, an ER protein required for retro-translocation of certain misfolded proteins, inhibited BKV infection. Consistent with this, we detected an interaction between Derlin-1 and VP1. Finally, we show that proteasome function is also linked to BKV infection and capsid rearrangement. These results indicate that BKV early entry and disassembly are highly regulated processes involving multiple cellular components.


Subject(s)
BK Virus/physiology , Membrane Fusion , Ammonium Chloride/pharmacology , BK Virus/drug effects , BK Virus/isolation & purification , Base Sequence , Blotting, Western , Brefeldin A/pharmacology , Cells, Cultured , DNA Primers , Endoplasmic Reticulum/virology , Humans , Hydrogen-Ion Concentration , Kidney Tubules, Proximal/cytology , Kidney Tubules, Proximal/virology , Microscopy, Fluorescence , Virus Replication
SELECTION OF CITATIONS
SEARCH DETAIL
...