Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
Biochim Biophys Acta Mol Basis Dis ; 1870(5): 167155, 2024 06.
Artículo en Inglés | MEDLINE | ID: mdl-38579939

RESUMEN

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.


Asunto(s)
Endocitosis , Túbulos Renales Proximales , SARS-CoV-2 , Glicoproteína de la Espiga del Coronavirus , Receptor Toll-Like 4 , Receptor Toll-Like 4/metabolismo , Endocitosis/efectos de los fármacos , Humanos , Túbulos Renales Proximales/metabolismo , Túbulos Renales Proximales/virología , Animales , Glicoproteína de la Espiga del Coronavirus/metabolismo , SARS-CoV-2/metabolismo , Células HEK293 , Porcinos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Fosforilación , COVID-19/metabolismo , COVID-19/virología , COVID-19/patología , Albúminas/metabolismo , Células LLC-PK1 , Células Epiteliales/metabolismo , Células Epiteliales/virología
2.
J Virol ; 98(3): e0180223, 2024 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-38334329

RESUMEN

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.


Asunto(s)
Enzima Convertidora de Angiotensina 2 , Riñón , Organoides , SARS-CoV-2 , Internalización del Virus , Humanos , Enzima Convertidora de Angiotensina 2/metabolismo , COVID-19/complicaciones , COVID-19/virología , Riñón/citología , Riñón/efectos de los fármacos , Riñón/metabolismo , Riñón/virología , Lisinopril/farmacología , Lisinopril/metabolismo , Organoides/citología , Organoides/efectos de los fármacos , Organoides/metabolismo , Organoides/virología , Pandemias , SARS-CoV-2/metabolismo , SARS-CoV-2/patogenicidad , Glicoproteína de la Espiga del Coronavirus/metabolismo , Internalización del Virus/efectos de los fármacos , Peptidil-Dipeptidasa A/metabolismo , Inhibidores de la Enzima Convertidora de Angiotensina/farmacología , Lesión Renal Aguda/etiología , Lesión Renal Aguda/metabolismo , Lesión Renal Aguda/virología , Túbulos Renales Proximales/citología , Túbulos Renales Proximales/efectos de los fármacos , Túbulos Renales Proximales/metabolismo , Túbulos Renales Proximales/virología , Receptores de Coronavirus/metabolismo , Modelos Biológicos , Serina Endopeptidasas/metabolismo , Endosomas/efectos de los fármacos , Endosomas/metabolismo , Endosomas/virología , Regulación de la Expresión Génica/efectos de los fármacos , Células Madre/citología
3.
JCI Insight ; 6(24)2021 12 22.
Artículo en Inglés | MEDLINE | ID: mdl-34767537

RESUMEN

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.


Asunto(s)
Lesión Renal Aguda/orina , COVID-19/orina , Túbulos Renales Proximales/virología , Riñón/virología , Organoides/virología , SARS-CoV-2/patogenicidad , Lesión Renal Aguda/etiología , Adulto , Anciano , Enzima Convertidora de Angiotensina 2/genética , Animales , Apoptosis , Cápsula Glomerular/citología , Cápsula Glomerular/virología , COVID-19/complicaciones , Chlorocebus aethiops , Femenino , Técnicas de Inactivación de Genes , Mortalidad Hospitalaria , Hospitalización , Humanos , Riñón/metabolismo , Riñón/patología , Túbulos Renales Proximales/metabolismo , Túbulos Renales Proximales/patología , Masculino , Persona de Mediana Edad , Organoides/metabolismo , Podocitos/virología , Enfermedades Renales Poliquísticas , Proteína Quinasa D2/genética , Proteoma , Receptores de Coronavirus/genética , Reproducibilidad de los Resultados , Transcriptoma , Células Vero , Tropismo Viral , Replicación Viral
4.
Eur J Pharmacol ; 888: 173487, 2020 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-32805256

RESUMEN

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.


Asunto(s)
Acetazolamida/uso terapéutico , Lesión Renal Aguda/etiología , Lesión Renal Aguda/prevención & control , Inhibidores de Anhidrasa Carbónica/uso terapéutico , Infecciones por Coronavirus/complicaciones , Neumonía Viral/complicaciones , Enzima Convertidora de Angiotensina 2 , COVID-19 , Humanos , Túbulos Renales Proximales/citología , Túbulos Renales Proximales/virología , Pandemias , Peptidil-Dipeptidasa A/metabolismo , Resultado del Tratamiento
5.
Mol Genet Genomic Med ; 8(10): e1442, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32744436

RESUMEN

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.


Asunto(s)
Lesión Renal Aguda/virología , Enzima Convertidora de Angiotensina 2/metabolismo , COVID-19/patología , Glomérulos Renales/metabolismo , Túbulos Renales Proximales/metabolismo , Receptores Virales/genética , Lesión Renal Aguda/patología , Enzima Convertidora de Angiotensina 2/genética , Secuencia de Bases , Humanos , Glomérulos Renales/patología , Glomérulos Renales/virología , Túbulos Renales Proximales/patología , Túbulos Renales Proximales/virología , Análisis de Componente Principal , SARS-CoV-2/metabolismo , Análisis de Secuencia de ARN , Serina Endopeptidasas/metabolismo , Análisis de la Célula Individual
6.
Int J Mol Sci ; 21(9)2020 May 05.
Artículo en Inglés | MEDLINE | ID: mdl-32380787

RESUMEN

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 [...].


Asunto(s)
Lesión Renal Aguda/mortalidad , Lesión Renal Aguda/virología , Betacoronavirus/patogenicidad , Infecciones por Coronavirus/virología , Túbulos Renales Proximales/metabolismo , Túbulos Renales Proximales/virología , Neumonía Viral/virología , Lesión Renal Aguda/fisiopatología , Lesión Renal Aguda/prevención & control , Angiotensina II/farmacología , Enzima Convertidora de Angiotensina 2 , COVID-19 , Catepsinas/metabolismo , Muerte Celular/efectos de los fármacos , Infecciones por Coronavirus/mortalidad , Infecciones por Coronavirus/patología , Infecciones por Coronavirus/fisiopatología , Creatinina/sangre , Enfermedad Crítica/mortalidad , Endosomas/efectos de los fármacos , Endosomas/enzimología , Endosomas/metabolismo , Humanos , Concentración de Iones de Hidrógeno , Incidencia , Túbulos Renales Proximales/fisiopatología , Lisosomas/efectos de los fármacos , Lisosomas/enzimología , Lisosomas/metabolismo , Pandemias , Peptidil-Dipeptidasa A/metabolismo , Neumonía Viral/mortalidad , Neumonía Viral/patología , Neumonía Viral/fisiopatología , Receptores Virales/metabolismo , SARS-CoV-2 , Síndrome Respiratorio Agudo Grave/sangre , Síndrome Respiratorio Agudo Grave/mortalidad , Síndrome Respiratorio Agudo Grave/fisiopatología , Internalización del Virus , Replicación Viral
7.
Biomaterials ; 219: 119367, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31344514

RESUMEN

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.


Asunto(s)
Herpesvirus Suido 1/fisiología , Enfermedades Renales/virología , Riñón/fisiopatología , Riñón/virología , Dispositivos Laboratorio en un Chip , Absorción Fisiológica/efectos de los fármacos , Angiotensina II/farmacología , Animales , Polaridad Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Perros , Herpesvirus Suido 1/efectos de los fármacos , Riñón/efectos de los fármacos , Riñón/patología , Túbulos Renales Proximales/efectos de los fármacos , Túbulos Renales Proximales/patología , Túbulos Renales Proximales/fisiopatología , Túbulos Renales Proximales/virología , Células de Riñón Canino Madin Darby , Microfluídica , Sodio/metabolismo
8.
Kidney Int ; 93(2): 355-364, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29061332

RESUMEN

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.


Asunto(s)
Células Dendríticas/metabolismo , Células Epiteliales/metabolismo , Rechazo de Injerto/metabolismo , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Trasplante de Riñón/efectos adversos , Túbulos Renales Proximales/metabolismo , Comunicación Paracrina , Fagocitosis , Receptor Toll-Like 9/metabolismo , Presentación de Antígeno , Apoptosis , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Línea Celular , Técnicas de Cocultivo , Citomegalovirus/inmunología , Citomegalovirus/patogenicidad , Células Dendríticas/inmunología , Células Epiteliales/inmunología , Células Epiteliales/patología , Células Epiteliales/virología , Rechazo de Injerto/inmunología , Rechazo de Injerto/patología , Rechazo de Injerto/virología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/inmunología , Interacciones Huésped-Patógeno , Humanos , Interferón-alfa/metabolismo , Isoantígenos/inmunología , Isoantígenos/metabolismo , Túbulos Renales Proximales/inmunología , Túbulos Renales Proximales/patología , Túbulos Renales Proximales/virología , Activación de Linfocitos , Fenotipo , Transducción de Señal , Receptor Toll-Like 9/inmunología
9.
J Innate Immun ; 9(6): 574-586, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28877527

RESUMEN

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.


Asunto(s)
Infecciones por Virus de Epstein-Barr/inmunología , Herpesvirus Humano 4/genética , Túbulos Renales Proximales/virología , MicroARNs/genética , ARN Viral/genética , Línea Celular , Interacciones Huésped-Patógeno , Humanos , Evasión Inmune , Inmunidad Innata , Túbulos Renales Proximales/inmunología , Moléculas de Patrón Molecular Asociado a Patógenos/inmunología , Receptores de Reconocimiento de Patrones/metabolismo , Receptores de Ácido Retinoico/metabolismo , Transducción de Señal
10.
Emerg Microbes Infect ; 6(8): e77, 2017 Aug 23.
Artículo en Inglés | MEDLINE | ID: mdl-28831192

RESUMEN

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.


Asunto(s)
Túbulos Renales Proximales/patología , Túbulos Renales Proximales/virología , Urotelio/virología , Infección por el Virus Zika/virología , Virus Zika/fisiología , Animales , Apoptosis , Línea Celular Tumoral , Efecto Citopatogénico Viral , Modelos Animales de Enfermedad , Reservorios de Enfermedades/virología , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Interferón Tipo I/genética , Interferón Tipo I/metabolismo , Riñón/patología , Riñón/virología , Túbulos Renales Proximales/citología , Ratones , Ratones Endogámicos C57BL , Orina/virología , Urotelio/citología , Carga Viral , Replicación Viral , Virus Zika/aislamiento & purificación , Infección por el Virus Zika/patología
11.
Virology ; 492: 66-72, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26901486

RESUMEN

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.


Asunto(s)
Virus BK/efectos de los fármacos , Caveolina 1/genética , Caveolina 2/genética , Cadenas Pesadas de Clatrina/genética , Células Epiteliales/efectos de los fármacos , Interacciones Huésped-Patógeno , Virus BK/genética , Virus BK/metabolismo , Caveolina 1/antagonistas & inhibidores , Caveolina 1/metabolismo , Caveolina 2/antagonistas & inhibidores , Caveolina 2/metabolismo , Línea Celular , Cadenas Pesadas de Clatrina/antagonistas & inhibidores , Cadenas Pesadas de Clatrina/metabolismo , Células Epiteliales/virología , Gangliósido G(M1)/farmacología , Gangliósidos/farmacología , Regulación de la Expresión Génica , Humanos , Túbulos Renales Proximales/efectos de los fármacos , Túbulos Renales Proximales/virología , MicroARNs/genética , MicroARNs/metabolismo , Proteínas de Transporte de Monosacáridos/antagonistas & inhibidores , Proteínas de Transporte de Monosacáridos/genética , Proteínas de Transporte de Monosacáridos/metabolismo , Cultivo Primario de Células , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Internalización del Virus/efectos de los fármacos
12.
J Proteome Res ; 14(10): 4413-24, 2015 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-26354146

RESUMEN

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.


Asunto(s)
Virus BK/fisiología , Núcleo Celular/metabolismo , Reparación del ADN , Células Epiteliales/metabolismo , Proteoma/metabolismo , Núcleo Celular/química , Núcleo Celular/patología , Núcleo Celular/virología , Cromatografía Liquida , Daño del ADN , Células Epiteliales/patología , Células Epiteliales/virología , Regulación de la Expresión Génica , Redes Reguladoras de Genes , Interacciones Huésped-Patógeno , Humanos , Marcaje Isotópico , Túbulos Renales Proximales/metabolismo , Túbulos Renales Proximales/patología , Túbulos Renales Proximales/virología , Anotación de Secuencia Molecular , Cultivo Primario de Células , Proteoma/genética , Proteoma/aislamiento & purificación , Transducción de Señal , Espectrometría de Masas en Tándem , Transcripción Genética
13.
Virology ; 474: 110-6, 2015 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-25463609

RESUMEN

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.


Asunto(s)
Virus BK/fisiología , Proteínas de la Cápside/fisiología , Señales de Localización Nuclear/fisiología , Transporte Activo de Núcleo Celular/efectos de los fármacos , Transporte Activo de Núcleo Celular/fisiología , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Virus BK/genética , Virus BK/patogenicidad , Proteínas de la Cápside/química , Proteínas de la Cápside/genética , Células Cultivadas , Técnicas de Silenciamiento del Gen , Interacciones Huésped-Patógeno , Humanos , Ivermectina/farmacología , Túbulos Renales Proximales/virología , Lisina/química , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Señales de Localización Nuclear/química , Señales de Localización Nuclear/genética , Homología de Secuencia de Aminoácido , Internalización del Virus , beta Carioferinas/antagonistas & inhibidores , beta Carioferinas/genética , beta Carioferinas/fisiología
14.
Int J Mol Med ; 31(5): 1017-29, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23483208

RESUMEN

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.


Asunto(s)
Apoptosis , Janus Quinasa 2/metabolismo , Túbulos Renales Proximales/metabolismo , Túbulos Renales Proximales/virología , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Transactivadores/metabolismo , Adolescente , Adulto , Western Blotting , Núcleo Celular/metabolismo , Núcleo Celular/ultraestructura , Proliferación Celular , Supervivencia Celular , Técnica del Anticuerpo Fluorescente , Glomerulonefritis/metabolismo , Glomerulonefritis/patología , Glomerulonefritis/virología , Humanos , Túbulos Renales Proximales/patología , Túbulos Renales Proximales/ultraestructura , Persona de Mediana Edad , Fosforilación , Transfección , Proteínas Reguladoras y Accesorias Virales , Adulto Joven , Proteína X Asociada a bcl-2/metabolismo
15.
Antiviral Res ; 92(1): 115-23, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21798289

RESUMEN

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.


Asunto(s)
Antivirales/farmacología , Virus BK/efectos de los fármacos , Virus BK/fisiología , Regulación hacia Abajo/efectos de los fármacos , Fluoroquinolonas/farmacología , Infecciones por Polyomavirus/virología , Replicación Viral/efectos de los fármacos , Virus BK/genética , Línea Celular , Células Cultivadas , Células Epiteliales/virología , Regulación Viral de la Expresión Génica/efectos de los fármacos , Humanos , Túbulos Renales Proximales/citología , Túbulos Renales Proximales/virología
16.
J Am Soc Nephrol ; 22(3): 496-507, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21335514

RESUMEN

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.


Asunto(s)
Comunicación Celular/fisiología , Células Epiteliales/virología , Regulación Viral de la Expresión Génica/fisiología , VIH-1/fisiología , Túbulos Renales Proximales/virología , Nefropatía Asociada a SIDA/etiología , Nefropatía Asociada a SIDA/patología , Nefropatía Asociada a SIDA/fisiopatología , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/virología , Adhesión Celular/efectos de los fármacos , Adhesión Celular/fisiología , Comunicación Celular/efectos de los fármacos , Línea Celular , Técnicas de Cocultivo , Células Epiteliales/citología , VIH-1/genética , Humanos , Túbulos Renales Proximales/citología , Polisacáridos/farmacología , Replicación Viral/fisiología
17.
PLoS Pathog ; 6(11): e1001170, 2010 Nov 04.
Artículo en Inglés | MEDLINE | ID: mdl-21079788

RESUMEN

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.


Asunto(s)
Infecciones por Citomegalovirus/metabolismo , Citomegalovirus/patogenicidad , Túbulos Renales Proximales/metabolismo , Túbulos Renales Proximales/virología , Factor de Crecimiento Transformador beta1/metabolismo , Animales , Western Blotting , Diferenciación Celular , Células Cultivadas , Infecciones por Citomegalovirus/patología , Infecciones por Citomegalovirus/virología , Células Epiteliales/metabolismo , Células Epiteliales/virología , Transición Epitelial-Mesenquimal , Técnica del Anticuerpo Fluorescente , Humanos , Inmunoprecipitación , Túbulos Renales Proximales/citología , ARN Mensajero/genética , Ratas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal
18.
Virology ; 407(2): 368-73, 2010 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-20869740

RESUMEN

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.


Asunto(s)
Virus BK/aislamiento & purificación , Variación Genética , Infecciones por VIH/complicaciones , Infecciones por Polyomavirus/virología , Secuencias Reguladoras de Ácidos Nucleicos/genética , Infecciones Tumorales por Virus/virología , Infecciones Oportunistas Relacionadas con el SIDA/virología , Virus BK/clasificación , Virus BK/genética , Células Cultivadas , ADN Viral/análisis , ADN Viral/aislamiento & purificación , Células Epiteliales/virología , Infecciones por VIH/virología , Humanos , Huésped Inmunocomprometido , Túbulos Renales Proximales/citología , Túbulos Renales Proximales/virología , Análisis de Secuencia de ADN , Orina/virología , Virología/métodos , Replicación Viral
19.
Curr Protoc Cell Biol ; Chapter 26: Unit 26.2, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19283732

RESUMEN

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.


Asunto(s)
Virus BK , Infecciones por Polyomavirus/virología , Infecciones Tumorales por Virus/virología , Virus BK/aislamiento & purificación , Virus BK/patogenicidad , Virus BK/fisiología , Técnicas de Cultivo de Célula , Células Epiteliales/metabolismo , Células Epiteliales/patología , Células Epiteliales/virología , Colorantes Fluorescentes/metabolismo , Humanos , Túbulos Renales Proximales/metabolismo , Túbulos Renales Proximales/patología , Túbulos Renales Proximales/virología , Microscopía Confocal , Infecciones por Polyomavirus/genética , Infecciones por Polyomavirus/metabolismo , Transporte de Proteínas , Infecciones Tumorales por Virus/genética , Infecciones Tumorales por Virus/metabolismo , Infecciones Tumorales por Virus/patología , Proteínas Virales/metabolismo , Virología/métodos , Internalización del Virus , Replicación Viral
20.
J Virol ; 83(3): 1350-8, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19036822

RESUMEN

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.


Asunto(s)
Virus BK/fisiología , Fusión de Membrana , Cloruro de Amonio/farmacología , Virus BK/efectos de los fármacos , Virus BK/aislamiento & purificación , Secuencia de Bases , Western Blotting , Brefeldino A/farmacología , Células Cultivadas , Cartilla de ADN , Retículo Endoplásmico/virología , Humanos , Concentración de Iones de Hidrógeno , Túbulos Renales Proximales/citología , Túbulos Renales Proximales/virología , Microscopía Fluorescente , Replicación Viral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA