Your browser doesn't support javascript.
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Nat Rev Nephrol ; 19(3): 151, 2023 03.
Artículo en Inglés | MEDLINE | ID: covidwho-2312408
2.
Int J Mol Sci ; 23(22)2022 Nov 17.
Artículo en Inglés | MEDLINE | ID: covidwho-2116209

RESUMEN

Since the outbreak of COVID-19 disease, a bidirectional interaction between kidney disease and the progression of COVID-19 has been demonstrated. Kidney disease is an independent risk factor for mortality of patients with COVID-19 as well as severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection leading to the development of acute kidney injury (AKI) and chronic kidney disease (CKD) in patients with COVID-19. However, the detection of kidney damage in patients with COVID-19 may not occur until an advanced stage based on the current clinical blood and urinary examinations. Some studies have pointed out the development of subclinical acute kidney injury (subAKI) syndrome with COVID-19. This syndrome is characterized by significant tubule interstitial injury without changes in the estimated glomerular filtration rate. Despite the complexity of the mechanism(s) underlying the development of subAKI, the involvement of changes in the protein endocytosis machinery in proximal tubule (PT) epithelial cells (PTECs) has been proposed. This paper focuses on the data relating to subAKI and COVID-19 and the role of PTECs and their protein endocytosis machinery in its pathogenesis.


Asunto(s)
Lesión Renal Aguda , COVID-19 , Insuficiencia Renal Crónica , Humanos , COVID-19/complicaciones , SARS-CoV-2 , Lesión Renal Aguda/metabolismo , Insuficiencia Renal Crónica/metabolismo , Túbulos Renales Proximales/metabolismo
3.
Int J Mol Sci ; 23(13)2022 Jun 29.
Artículo en Inglés | MEDLINE | ID: covidwho-1934128

RESUMEN

Acute kidney injury (AKI) is an increasingly common problem afflicting all ages, occurring in over 20% of non-critically ill hospitalized patients and >30% of children and >50% of adults in critical care units. AKI is associated with serious short-term and long-term consequences, and current therapeutic options are unsatisfactory. Large gaps remain in our understanding of human AKI pathobiology, which have hindered the discovery of novel diagnostics and therapeutics. Although animal models of AKI have been extensively studied, these differ significantly from human AKI in terms of molecular and cellular responses. In addition, animal models suffer from interspecies differences, high costs and ethical considerations. Static two-dimensional cell culture models of AKI also have limited utility since they have focused almost exclusively on hypoxic or cytotoxic injury to proximal tubules alone. An optimal AKI model would encompass several of the diverse specific cell types in the kidney that could be targets of injury. Second, it would resemble the human physiological milieu as closely as possible. Third, it would yield sensitive and measurable readouts that are directly applicable to the human condition. In this regard, the past two decades have seen a dramatic shift towards newer personalized human-based models to study human AKI. In this review, we provide recent developments using human stem cells, organoids, and in silico approaches to advance personalized AKI diagnostics and therapeutics.


Asunto(s)
Lesión Renal Aguda , Organoides , Lesión Renal Aguda/diagnóstico , Lesión Renal Aguda/terapia , Animales , Enfermedad Crítica/terapia , Humanos , Unidades de Cuidados Intensivos , Túbulos Renales Proximales , Células Madre
4.
Life Sci Alliance ; 5(5)2022 05.
Artículo en Inglés | MEDLINE | ID: covidwho-1675573

RESUMEN

Acute kidney injury is associated with mortality in COVID-19 patients. However, host cell changes underlying infection of renal cells with SARS-CoV-2 remain unknown and prevent understanding of the molecular mechanisms that may contribute to renal pathology. Here, we carried out quantitative translatome and whole-cell proteomics analyses of primary renal proximal and distal tubular epithelial cells derived from human donors infected with SARS-CoV-2 or MERS-CoV to disseminate virus and cell type-specific changes over time. Our findings revealed shared pathways modified upon infection with both viruses, as well as SARS-CoV-2-specific host cell modulation driving key changes in innate immune activation and cellular protein quality control. Notably, MERS-CoV infection-induced specific changes in mitochondrial biology that were not observed in response to SARS-CoV-2 infection. Furthermore, we identified extensive modulation in pathways associated with kidney failure that changed in a virus- and cell type-specific manner. In summary, we provide an overview of the effects of SARS-CoV-2 or MERS-CoV infection on primary renal epithelial cells revealing key pathways that may be essential for viral replication.


Asunto(s)
Células Epiteliales/metabolismo , Células Epiteliales/virología , Riñón , Coronavirus del Síndrome Respiratorio de Oriente Medio/fisiología , Proteoma , Proteómica , SARS-CoV-2/fisiología , Biomarcadores , COVID-19/metabolismo , COVID-19/virología , Núcleo Celular/genética , Núcleo Celular/metabolismo , Células Cultivadas , Biología Computacional/métodos , Infecciones por Coronavirus/metabolismo , Infecciones por Coronavirus/virología , Regulación de la Expresión Génica , Interacciones Huésped-Patógeno/genética , Interacciones Huésped-Patógeno/inmunología , Humanos , Túbulos Renales Distales , Túbulos Renales Proximales , Mitocondrias/genética , Mitocondrias/metabolismo , Cultivo Primario de Células , Proteómica/métodos , Replicación Viral
5.
Mayo Clin Proc ; 96(10): 2561-2575, 2021 10.
Artículo en Inglés | MEDLINE | ID: covidwho-1521396

RESUMEN

OBJECTIVE: To compare coronavirus disease 2019 (COVID-19) acute kidney injury (AKI) to sepsis-AKI (S-AKI). The morphology and transcriptomic and proteomic characteristics of autopsy kidneys were analyzed. PATIENTS AND METHODS: Individuals 18 years of age and older who died from COVID-19 and had an autopsy performed at Mayo Clinic between April 2020 to October 2020 were included. Morphological evaluation of the kidneys of 17 individuals with COVID-19 was performed. In a subset of seven COVID-19 cases with postmortem interval of less than or equal to 20 hours, ultrastructural and molecular characteristics (targeted transcriptome and proteomics analyses of tubulointerstitium) were evaluated. Molecular characteristics were compared with archived cases of S-AKI and nonsepsis causes of AKI. RESULTS: The spectrum of COVID-19 renal pathology included macrophage-dominant microvascular inflammation (glomerulitis and peritubular capillaritis), vascular dysfunction (peritubular capillary congestion and endothelial injury), and tubular injury with ultrastructural evidence of mitochondrial damage. Investigation of the spatial architecture using a novel imaging mass cytometry revealed enrichment of CD3+CD4+ T cells in close proximity to antigen-presenting cells, and macrophage-enriched glomerular and interstitial infiltrates, suggesting an innate and adaptive immune tissue response. Coronavirus disease 2019 AKI and S-AKI, as compared to nonseptic AKI, had an enrichment of transcriptional pathways involved in inflammation (apoptosis, autophagy, major histocompatibility complex class I and II, and type 1 T helper cell differentiation). Proteomic pathway analysis showed that COVID-19 AKI and to a lesser extent S-AKI were enriched in necroptosis and sirtuin-signaling pathways, both involved in regulatory response to inflammation. Upregulation of the ceramide-signaling pathway and downregulation of oxidative phosphorylation in COVID-19 AKI were noted. CONCLUSION: This data highlights the similarities between S-AKI and COVID-19 AKI and suggests that mitochondrial dysfunction may play a pivotal role in COVID-19 AKI. This data may allow the development of novel diagnostic and therapeutic targets.


Asunto(s)
Lesión Renal Aguda/patología , COVID-19/patología , Riñón/patología , Sepsis/patología , Lesión Renal Aguda/virología , Adulto , Autopsia , Humanos , Túbulos Renales Proximales/patología , Masculino , Persona de Mediana Edad , Sepsis/virología
6.
JCI Insight ; 6(24)2021 12 22.
Artículo en Inglés | MEDLINE | ID: covidwho-1518199

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
7.
J Am Soc Nephrol ; 32(1): 86-97, 2021 01.
Artículo en Inglés | MEDLINE | ID: covidwho-1496653

RESUMEN

BACKGROUND: Cultured cell lines are widely used for research in the physiology, pathophysiology, toxicology, and pharmacology of the renal proximal tubule. The lines that are most appropriate for a given use depend upon the genes expressed. New tools for transcriptomic profiling using RNA sequencing (RNA-Seq) make it possible to catalog expressed genes in each cell line. METHODS: Fourteen different proximal tubule cell lines, representing six species, were grown on permeable supports under conditions specific for the respective lines. RNA-Seq followed standard procedures. RESULTS: Transcripts expressed in cell lines variably matched transcripts selectively expressed in native proximal tubule. Opossum kidney (OK) cells displayed the highest percentage match (45% of proximal marker genes [TPM threshold =15]), with pig kidney cells (LLC-PK1) close behind (39%). Lower-percentage matches were seen for various human lines, including HK-2 (26%), and lines from rodent kidneys, such as NRK-52E (23%). Nominally, identical OK cells from different sources differed substantially in expression of proximal tubule markers. Mapping cell line transcriptomes to gene sets for various proximal tubule functions (sodium and water transport, protein transport, metabolic functions, endocrine functions) showed that different lines may be optimal for experimentally modeling each function. An online resource (https://esbl.nhlbi.nih.gov/JBrowse/KCT/) has been created to interrogate cell line transcriptome data. Proteomic analysis of NRK-52E cells confirmed low expression of many proximal tubule marker proteins. CONCLUSIONS: No cell line fully matched the transcriptome of native proximal tubule cells. However, some of the lines tested are suitable for the study of particular metabolic and transport processes seen in the proximal tubule.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Túbulos Renales Proximales/metabolismo , Transcriptoma , Animales , Transporte Biológico , Línea Celular , Cromatografía Liquida , Perfilación de la Expresión Génica , Humanos , Internet , Ratones , Zarigüeyas , Proteómica , RNA-Seq , Ratas , Análisis de Secuencia de ARN , Especificidad de la Especie , Porcinos , Espectrometría de Masas en Tándem
8.
J Am Soc Nephrol ; 32(1): 86-97, 2021 01.
Artículo en Inglés | MEDLINE | ID: covidwho-1496652

RESUMEN

BACKGROUND: Cultured cell lines are widely used for research in the physiology, pathophysiology, toxicology, and pharmacology of the renal proximal tubule. The lines that are most appropriate for a given use depend upon the genes expressed. New tools for transcriptomic profiling using RNA sequencing (RNA-Seq) make it possible to catalog expressed genes in each cell line. METHODS: Fourteen different proximal tubule cell lines, representing six species, were grown on permeable supports under conditions specific for the respective lines. RNA-Seq followed standard procedures. RESULTS: Transcripts expressed in cell lines variably matched transcripts selectively expressed in native proximal tubule. Opossum kidney (OK) cells displayed the highest percentage match (45% of proximal marker genes [TPM threshold =15]), with pig kidney cells (LLC-PK1) close behind (39%). Lower-percentage matches were seen for various human lines, including HK-2 (26%), and lines from rodent kidneys, such as NRK-52E (23%). Nominally, identical OK cells from different sources differed substantially in expression of proximal tubule markers. Mapping cell line transcriptomes to gene sets for various proximal tubule functions (sodium and water transport, protein transport, metabolic functions, endocrine functions) showed that different lines may be optimal for experimentally modeling each function. An online resource (https://esbl.nhlbi.nih.gov/JBrowse/KCT/) has been created to interrogate cell line transcriptome data. Proteomic analysis of NRK-52E cells confirmed low expression of many proximal tubule marker proteins. CONCLUSIONS: No cell line fully matched the transcriptome of native proximal tubule cells. However, some of the lines tested are suitable for the study of particular metabolic and transport processes seen in the proximal tubule.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Túbulos Renales Proximales/metabolismo , Transcriptoma , Animales , Transporte Biológico , Línea Celular , Cromatografía Liquida , Perfilación de la Expresión Génica , Humanos , Internet , Ratones , Zarigüeyas , Proteómica , RNA-Seq , Ratas , Análisis de Secuencia de ARN , Especificidad de la Especie , Porcinos , Espectrometría de Masas en Tándem
9.
J Am Soc Nephrol ; 32(1): 86-97, 2021 01.
Artículo en Inglés | MEDLINE | ID: covidwho-1496651

RESUMEN

BACKGROUND: Cultured cell lines are widely used for research in the physiology, pathophysiology, toxicology, and pharmacology of the renal proximal tubule. The lines that are most appropriate for a given use depend upon the genes expressed. New tools for transcriptomic profiling using RNA sequencing (RNA-Seq) make it possible to catalog expressed genes in each cell line. METHODS: Fourteen different proximal tubule cell lines, representing six species, were grown on permeable supports under conditions specific for the respective lines. RNA-Seq followed standard procedures. RESULTS: Transcripts expressed in cell lines variably matched transcripts selectively expressed in native proximal tubule. Opossum kidney (OK) cells displayed the highest percentage match (45% of proximal marker genes [TPM threshold =15]), with pig kidney cells (LLC-PK1) close behind (39%). Lower-percentage matches were seen for various human lines, including HK-2 (26%), and lines from rodent kidneys, such as NRK-52E (23%). Nominally, identical OK cells from different sources differed substantially in expression of proximal tubule markers. Mapping cell line transcriptomes to gene sets for various proximal tubule functions (sodium and water transport, protein transport, metabolic functions, endocrine functions) showed that different lines may be optimal for experimentally modeling each function. An online resource (https://esbl.nhlbi.nih.gov/JBrowse/KCT/) has been created to interrogate cell line transcriptome data. Proteomic analysis of NRK-52E cells confirmed low expression of many proximal tubule marker proteins. CONCLUSIONS: No cell line fully matched the transcriptome of native proximal tubule cells. However, some of the lines tested are suitable for the study of particular metabolic and transport processes seen in the proximal tubule.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Túbulos Renales Proximales/metabolismo , Transcriptoma , Animales , Transporte Biológico , Línea Celular , Cromatografía Liquida , Perfilación de la Expresión Génica , Humanos , Internet , Ratones , Zarigüeyas , Proteómica , RNA-Seq , Ratas , Análisis de Secuencia de ARN , Especificidad de la Especie , Porcinos , Espectrometría de Masas en Tándem
10.
Crit Care ; 25(1): 212, 2021 06 14.
Artículo en Inglés | MEDLINE | ID: covidwho-1269885

RESUMEN

BACKGROUND: The severity of coronavirus disease 2019 (COVID-19) is highly variable between individuals, ranging from asymptomatic infection to critical disease with acute respiratory distress syndrome requiring mechanical ventilation. Such variability stresses the need for novel biomarkers associated with disease outcome. As SARS-CoV-2 infection causes a kidney proximal tubule dysfunction with urinary loss of uric acid, we hypothesized that low serum levels of uric acid (hypouricemia) may be associated with severity and outcome of COVID-19. METHODS: In a retrospective study using two independent cohorts, we investigated and validated the prevalence, kinetics and clinical correlates of hypouricemia among patients hospitalized with COVID-19 to a large academic hospital in Brussels, Belgium. Survival analyses using Cox regression and a competing risk approach assessed the time to mechanical ventilation and/or death. Confocal microscopy assessed the expression of urate transporter URAT1 in kidney proximal tubule cells from patients who died from COVID-19. RESULTS: The discovery and validation cohorts included 192 and 325 patients hospitalized with COVID-19, respectively. Out of the 517 patients, 274 (53%) had severe and 92 (18%) critical COVID-19. In both cohorts, the prevalence of hypouricemia increased from 6% upon admission to 20% within the first days of hospitalization for COVID-19, contrasting with a very rare occurrence (< 1%) before hospitalization for COVID-19. During a median (interquartile range) follow-up of 148 days (50-168), 61 (12%) patients required mechanical ventilation and 93 (18%) died. In both cohorts considered separately and in pooled analyses, low serum levels of uric acid were strongly associated with disease severity (linear trend, P < 0.001) and with progression to death and respiratory failure requiring mechanical ventilation in Cox (adjusted hazard ratio 5.3, 95% confidence interval 3.6-7.8, P < 0.001) or competing risks (adjusted hazard ratio 20.8, 95% confidence interval 10.4-41.4, P < 0.001) models. At the structural level, kidneys from patients with COVID-19 showed a major reduction in urate transporter URAT1 expression in the brush border of proximal tubules. CONCLUSIONS: Among patients with COVID-19 requiring hospitalization, low serum levels of uric acid are common and associate with disease severity and with progression to respiratory failure requiring invasive mechanical ventilation.


Asunto(s)
COVID-19/metabolismo , COVID-19/fisiopatología , Túbulos Renales Proximales/metabolismo , Índice de Severidad de la Enfermedad , Ácido Úrico/sangre , Anciano , Bélgica , COVID-19/complicaciones , Estudios de Cohortes , Enfermedad Crítica/epidemiología , Humanos , Masculino , Persona de Mediana Edad , Transportadores de Anión Orgánico/metabolismo , Proteínas de Transporte de Catión Orgánico/metabolismo , Evaluación de Resultado en la Atención de Salud , Estudios Retrospectivos
11.
J Pharm Pharm Sci ; 24: 227-236, 2021.
Artículo en Inglés | MEDLINE | ID: covidwho-1248472

RESUMEN

PURPOSE: Remdesivir and its active metabolite are predominantly eliminated via renal route; however, information regarding renal uptake transporters is limited. In the present study, the interaction of remdesivir and its nucleoside analog GS-441524 with OATP4C1 was evaluated to provide the detailed information about its renal handling. METHODS: We used HK-2 cells, a proximal tubular cell line derived from normal kidney, to confirm the transport of remdesivir and GS-441524. To assess the involvement of OATP4C1 in handling remdesivir and GS-441524, the uptake study of remdesivir and GS-441524 was performed by using OATP4C1-overexpressing Madin-Darby canine kidney II (MDCKII) cells. Moreover, we also evaluated the IC50 and Ki value of remdesivir. RESULTS: The time-dependent remdesivir uptake in HK-2 cells was observed. The results of inhibition study using OATs and OCT2 inhibitors and OATP4C1 knockdown suggested the involvement of renal drug transporter OATP4C1. Remdesivir was taken up by OATP4C1/MDCKII cells. OATP4C1-mediated uptake of remdesivir increased linearly up to 10 min and reached a steady state at 30 min. Remdesivir inhibited OATP4C1-mediated transport in a concentration-dependent manner with the IC50 and apparent Ki values of 42 ± 7.8 µM and 37 ± 6.9 µM, respectively. CONCLUSIONS: We have provided novel information about renal handling of remdesivir. Furthermore, we evaluated the potential drug interaction via OATP4C1 by calculating the Ki value of remdesivir. OATP4C1 may play a pivotal role in remdesivir therapy for COVID-19, particularly in patients with kidney injury.


Asunto(s)
Adenosina Monofosfato/análogos & derivados , Alanina/análogos & derivados , Antivirales/metabolismo , Tratamiento Farmacológico de COVID-19 , Furanos/metabolismo , Túbulos Renales Proximales/metabolismo , Transportadores de Anión Orgánico/metabolismo , Pirroles/metabolismo , Triazinas/metabolismo , Adenosina/análogos & derivados , Adenosina Monofosfato/metabolismo , Adenosina Monofosfato/uso terapéutico , Alanina/metabolismo , Alanina/uso terapéutico , Animales , Antivirales/uso terapéutico , COVID-19/metabolismo , Línea Celular , Perros , Relación Dosis-Respuesta a Droga , Aprobación de Drogas , Furanos/uso terapéutico , Humanos , Riñón/efectos de los fármacos , Riñón/metabolismo , Túbulos Renales Proximales/efectos de los fármacos , Células de Riñón Canino Madin Darby , Transportadores de Anión Orgánico/antagonistas & inhibidores , Pirroles/uso terapéutico , Triazinas/uso terapéutico
12.
Eur J Pharmacol ; 888: 173487, 2020 Dec 05.
Artículo en Inglés | MEDLINE | ID: covidwho-1049775

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
13.
Kidney Int ; 98(6): 1502-1518, 2020 12.
Artículo en Inglés | MEDLINE | ID: covidwho-1023697

RESUMEN

COVID-19 morbidity and mortality are increased via unknown mechanisms in patients with diabetes and kidney disease. SARS-CoV-2 uses angiotensin-converting enzyme 2 (ACE2) for entry into host cells. Because ACE2 is a susceptibility factor for infection, we investigated how diabetic kidney disease and medications alter ACE2 receptor expression in kidneys. Single cell RNA profiling of kidney biopsies from healthy living donors and patients with diabetic kidney disease revealed ACE2 expression primarily in proximal tubular epithelial cells. This cell-specific localization was confirmed by in situ hybridization. ACE2 expression levels were unaltered by exposures to renin-angiotensin-aldosterone system inhibitors in diabetic kidney disease. Bayesian integrative analysis of a large compendium of public -omics datasets identified molecular network modules induced in ACE2-expressing proximal tubular epithelial cells in diabetic kidney disease (searchable at hb.flatironinstitute.org/covid-kidney) that were linked to viral entry, immune activation, endomembrane reorganization, and RNA processing. The diabetic kidney disease ACE2-positive proximal tubular epithelial cell module overlapped with expression patterns seen in SARS-CoV-2-infected cells. Similar cellular programs were seen in ACE2-positive proximal tubular epithelial cells obtained from urine samples of 13 hospitalized patients with COVID-19, suggesting a consistent ACE2-coregulated proximal tubular epithelial cell expression program that may interact with the SARS-CoV-2 infection processes. Thus SARS-CoV-2 receptor networks can seed further research into risk stratification and therapeutic strategies for COVID-19-related kidney damage.


Asunto(s)
Enzima Convertidora de Angiotensina 2/metabolismo , COVID-19/metabolismo , Nefropatías Diabéticas/metabolismo , Túbulos Renales Proximales/metabolismo , SARS-CoV-2/metabolismo , Adulto , Anciano , Antagonistas de Receptores de Angiotensina/farmacología , Antagonistas de Receptores de Angiotensina/uso terapéutico , Inhibidores de la Enzima Convertidora de Angiotensina/farmacología , Inhibidores de la Enzima Convertidora de Angiotensina/uso terapéutico , COVID-19/complicaciones , COVID-19/virología , Estudios de Casos y Controles , Nefropatías Diabéticas/tratamiento farmacológico , Femenino , Perfilación de la Expresión Génica , Redes Reguladoras de Genes , Interacciones Huésped-Patógeno , Humanos , Túbulos Renales Proximales/efectos de los fármacos , Masculino , Persona de Mediana Edad
14.
Adv Chronic Kidney Dis ; 27(5): 365-376, 2020 09.
Artículo en Inglés | MEDLINE | ID: covidwho-975047

RESUMEN

Acute kidney injury (AKI) is common among hospitalized patients with Coronavirus Infectious Disease 2019 (COVID-19), with the occurrence of AKI ranging from 0.5% to 80%. The variability in the occurrence of AKI has been attributed to the difference in geographic locations, race/ethnicity, and severity of illness. AKI among hospitalized patients is associated with increased length of stay and in-hospital deaths. Even patients with AKI who survive to hospital discharge are at risk of developing chronic kidney disease or end-stage kidney disease. An improved knowledge of the pathophysiology of AKI in COVID-19 is crucial to mitigate and manage AKI and to improve the survival of patients who developed AKI during COVID-19. The goal of this article is to provide our current understanding of the etiology and the pathophysiology of AKI in the setting of COVID-19.


Asunto(s)
Lesión Renal Aguda/metabolismo , COVID-19/metabolismo , Citocinas/metabolismo , Glomerulonefritis/metabolismo , Microangiopatías Trombóticas/metabolismo , Lesión Renal Aguda/etiología , Lesión Renal Aguda/patología , Lesión Renal Aguda/fisiopatología , Antibacterianos/efectos adversos , Antivirales/efectos adversos , Apolipoproteína L1/genética , Ácido Ascórbico/efectos adversos , Azotemia/metabolismo , Azotemia/patología , Azotemia/fisiopatología , COVID-19/patología , COVID-19/fisiopatología , Progresión de la Enfermedad , Glomerulonefritis/patología , Glomerulonefritis/fisiopatología , Glomerulonefritis Membranosa/metabolismo , Glomerulonefritis Membranosa/patología , Glomerulonefritis Membranosa/fisiopatología , Mortalidad Hospitalaria , Humanos , Túbulos Renales Proximales/lesiones , Tiempo de Internación , Mioglobina/metabolismo , Nefritis Intersticial/metabolismo , Nefritis Intersticial/patología , Nefritis Intersticial/fisiopatología , Nefrosis Lipoidea/metabolismo , Nefrosis Lipoidea/patología , Nefrosis Lipoidea/fisiopatología , Insuficiencia Renal Crónica , Rabdomiólisis/metabolismo , SARS-CoV-2 , Índice de Severidad de la Enfermedad , Microangiopatías Trombóticas/patología , Microangiopatías Trombóticas/fisiopatología , Vitaminas/efectos adversos , Tratamiento Farmacológico de COVID-19
15.
Rev Med Virol ; 31(3): e2176, 2021 05.
Artículo en Inglés | MEDLINE | ID: covidwho-815924

RESUMEN

The novel coronavirus (SARS-CoV-2) has turned into a life-threatening pandemic disease (Covid-19). About 5% of patients with Covid-19 have severe symptoms including septic shock, acute respiratory distress syndrome, and the failure of several organs, while most of them have mild symptoms. Frequently, the kidneys are involved through direct or indirect mechanisms. Kidney involvement mainly manifests itself as proteinuria and acute kidney injury (AKI). The SARS-CoV-2-induced kidney damage is expected to be multifactorial; directly it can infect the kidney podocytes and proximal tubular cells and based on an angiotensin-converting enzyme 2 (ACE2) pathway it can lead to acute tubular necrosis, protein leakage in Bowman's capsule, collapsing glomerulopathy and mitochondrial impairment. The SARS-CoV-2-driven dysregulation of the immune responses including cytokine storm, macrophage activation syndrome, and lymphopenia can be other causes of the AKI. Organ interactions, endothelial dysfunction, hypercoagulability, rhabdomyolysis, and sepsis are other potential mechanisms of AKI. Moreover, lower oxygen delivery to kidney may cause an ischaemic injury. Understanding the fundamental molecular pathways and pathophysiology of kidney injury and AKI in Covid-19 is necessary to develop management strategies and design effective therapies.


Asunto(s)
Lesión Renal Aguda/patología , COVID-19/fisiopatología , Síndrome de Liberación de Citoquinas/patología , Coagulación Intravascular Diseminada/patología , Linfopenia/patología , Necrosis/patología , Proteinuria/patología , Sepsis/patología , Lesión Renal Aguda/inmunología , Lesión Renal Aguda/virología , Enzima Convertidora de Angiotensina 2/genética , Enzima Convertidora de Angiotensina 2/inmunología , COVID-19/inmunología , COVID-19/virología , Síndrome de Liberación de Citoquinas/inmunología , Síndrome de Liberación de Citoquinas/virología , Citocinas/genética , Citocinas/inmunología , Coagulación Intravascular Diseminada/inmunología , Coagulación Intravascular Diseminada/virología , Interacciones Huésped-Patógeno/genética , Interacciones Huésped-Patógeno/inmunología , Humanos , Túbulos Renales Proximales/inmunología , Túbulos Renales Proximales/fisiopatología , Linfopenia/inmunología , Linfopenia/virología , Necrosis/inmunología , Necrosis/virología , Podocitos/inmunología , Podocitos/patología , Proteinuria/inmunología , Proteinuria/virología , SARS-CoV-2/inmunología , SARS-CoV-2/patogenicidad , Sepsis/inmunología , Sepsis/virología , Serina Endopeptidasas/genética , Serina Endopeptidasas/inmunología , Glicoproteína de la Espiga del Coronavirus/genética , Glicoproteína de la Espiga del Coronavirus/inmunología
16.
Cell Rep ; 32(12): 108175, 2020 09 22.
Artículo en Inglés | MEDLINE | ID: covidwho-747293

RESUMEN

To predict the tropism of human coronaviruses, we profile 28 SARS-CoV-2 and coronavirus-associated receptors and factors (SCARFs) using single-cell transcriptomics across various healthy human tissues. SCARFs include cellular factors both facilitating and restricting viral entry. Intestinal goblet cells, enterocytes, and kidney proximal tubule cells appear highly permissive to SARS-CoV-2, consistent with clinical data. Our analysis also predicts non-canonical entry paths for lung and brain infections. Spermatogonial cells and prostate endocrine cells also appear to be permissive to SARS-CoV-2 infection, suggesting male-specific vulnerabilities. Both pro- and anti-viral factors are highly expressed within the nasal epithelium, with potential age-dependent variation, predicting an important battleground for coronavirus infection. Our analysis also suggests that early embryonic and placental development are at moderate risk of infection. Lastly, SCARF expression appears broadly conserved across a subset of primate organs examined. Our study establishes a resource for investigations of coronavirus biology and pathology.


Asunto(s)
Infecciones por Coronavirus/patología , Mucosa Nasal/metabolismo , Neumonía Viral/patología , Receptores Virales/genética , Tropismo Viral/genética , Internalización del Virus , Células A549 , Enzima Convertidora de Angiotensina 2 , Animales , Betacoronavirus/crecimiento & desarrollo , COVID-19 , Línea Celular , Chlorocebus aethiops , Enterocitos/metabolismo , Perfilación de la Expresión Génica , Células Caliciformes/metabolismo , Células HEK293 , Humanos , Túbulos Renales Proximales/citología , Túbulos Renales Proximales/metabolismo , Mucosa Nasal/virología , Pandemias , Peptidil-Dipeptidasa A/genética , Peptidil-Dipeptidasa A/metabolismo , SARS-CoV-2 , Serina Endopeptidasas/genética , Serina Endopeptidasas/metabolismo , Análisis de la Célula Individual , Células Vero
18.
Kidney Int ; 98(5): 1296-1307, 2020 11.
Artículo en Inglés | MEDLINE | ID: covidwho-704001

RESUMEN

Coronavirus disease 2019 (COVID-19) is commonly associated with kidney damage, and the angiotensin converting enzyme 2 (ACE2) receptor for SARS-CoV-2 is highly expressed in the proximal tubule cells. Whether patients with COVID-19 present specific manifestations of proximal tubule dysfunction remains unknown. To test this, we examined a cohort of 49 patients requiring hospitalization in a large academic hospital in Brussels, Belgium. There was evidence of proximal tubule dysfunction in a subset of patients with COVID-19, as attested by low-molecular-weight proteinuria (70-80%), neutral aminoaciduria (46%), and defective handling of uric acid (46%) or phosphate (19%). None of the patients had normoglycemic glucosuria. Proximal tubule dysfunction was independent of pre-existing comorbidities, glomerular proteinuria, nephrotoxic medications or viral load. At the structural level, kidneys from patients with COVID-19 showed prominent tubular injury, including in the initial part of the proximal tubule, with brush border loss, acute tubular necrosis, intraluminal debris, and a marked decrease in the expression of megalin in the brush border. Transmission electron microscopy identified particles resembling coronaviruses in vacuoles or cisternae of the endoplasmic reticulum in proximal tubule cells. Among features of proximal tubule dysfunction, hypouricemia with inappropriate uricosuria was independently associated with disease severity and with a significant increase in the risk of respiratory failure requiring invasive mechanical ventilation using Cox (adjusted hazard ratio 6.2, 95% CI 1.9-20.1) or competing risks (adjusted sub-distribution hazard ratio 12.1, 95% CI 2.7-55.4) survival models. Thus, our data establish that SARS-CoV-2 causes specific manifestations of proximal tubule dysfunction and provide novel insights into COVID-19 severity and outcome.


Asunto(s)
Infecciones por Coronavirus/fisiopatología , Túbulos Renales Proximales/fisiopatología , Neumonía Viral/fisiopatología , Anciano , Anciano de 80 o más Años , Bélgica/epidemiología , Betacoronavirus , COVID-19 , Estudios de Casos y Controles , Infecciones por Coronavirus/mortalidad , Infecciones por Coronavirus/patología , Infecciones por Coronavirus/terapia , Humanos , Túbulos Renales Proximales/ultraestructura , Masculino , Persona de Mediana Edad , Pandemias , Neumonía Viral/mortalidad , Neumonía Viral/patología , Neumonía Viral/terapia , SARS-CoV-2
19.
Mol Genet Genomic Med ; 8(10): e1442, 2020 10.
Artículo en Inglés | MEDLINE | ID: covidwho-692077

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
20.
Mol Syst Biol ; 16(7): e9610, 2020 07.
Artículo en Inglés | MEDLINE | ID: covidwho-680519

RESUMEN

The novel SARS-coronavirus 2 (SARS-CoV-2) poses a global challenge on healthcare and society. For understanding the susceptibility for SARS-CoV-2 infection, the cell type-specific expression of the host cell surface receptor is necessary. The key protein suggested to be involved in host cell entry is angiotensin I converting enzyme 2 (ACE2). Here, we report the expression pattern of ACE2 across > 150 different cell types corresponding to all major human tissues and organs based on stringent immunohistochemical analysis. The results were compared with several datasets both on the mRNA and protein level. ACE2 expression was mainly observed in enterocytes, renal tubules, gallbladder, cardiomyocytes, male reproductive cells, placental trophoblasts, ductal cells, eye, and vasculature. In the respiratory system, the expression was limited, with no or only low expression in a subset of cells in a few individuals, observed by one antibody only. Our data constitute an important resource for further studies on SARS-CoV-2 host cell entry, in order to understand the biology of the disease and to aid in the development of effective treatments to the viral infection.


Asunto(s)
Peptidil-Dipeptidasa A/metabolismo , Sistema Respiratorio/metabolismo , Enzima Convertidora de Angiotensina 2 , Betacoronavirus , Vasos Sanguíneos/metabolismo , Conjuntiva/metabolismo , Enterocitos/metabolismo , Femenino , Vesícula Biliar/metabolismo , Interacciones Microbiota-Huesped , Humanos , Inmunohistoquímica , Túbulos Renales Proximales/metabolismo , Masculino , Espectrometría de Masas , Miocitos Cardíacos/metabolismo , Especificidad de Órganos , Peptidil-Dipeptidasa A/genética , Placenta/metabolismo , Embarazo , RNA-Seq , SARS-CoV-2 , Análisis de la Célula Individual , Testículo/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA