Your browser doesn't support javascript.
Montrer: 20 | 50 | 100
Résultats 1 - 18 de 18
Filtre
2.
Front Immunol ; 14: 961642, 2023.
Article Dans Anglais | MEDLINE | ID: covidwho-2306453

Résumé

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the main cause of COVID-19, causing hundreds of millions of confirmed cases and more than 18.2 million deaths worldwide. Acute kidney injury (AKI) is a common complication of COVID-19 that leads to an increase in mortality, especially in intensive care unit (ICU) settings, and chronic kidney disease (CKD) is a high risk factor for COVID-19 and its related mortality. However, the underlying molecular mechanisms among AKI, CKD, and COVID-19 are unclear. Therefore, transcriptome analysis was performed to examine common pathways and molecular biomarkers for AKI, CKD, and COVID-19 in an attempt to understand the association of SARS-CoV-2 infection with AKI and CKD. Three RNA-seq datasets (GSE147507, GSE1563, and GSE66494) from the GEO database were used to detect differentially expressed genes (DEGs) for COVID-19 with AKI and CKD to search for shared pathways and candidate targets. A total of 17 common DEGs were confirmed, and their biological functions and signaling pathways were characterized by enrichment analysis. MAPK signaling, the structural pathway of interleukin 1 (IL-1), and the Toll-like receptor pathway appear to be involved in the occurrence of these diseases. Hub genes identified from the protein-protein interaction (PPI) network, including DUSP6, BHLHE40, RASGRP1, and TAB2, are potential therapeutic targets in COVID-19 with AKI and CKD. Common genes and pathways may play pathogenic roles in these three diseases mainly through the activation of immune inflammation. Networks of transcription factor (TF)-gene, miRNA-gene, and gene-disease interactions from the datasets were also constructed, and key gene regulators influencing the progression of these three diseases were further identified among the DEGs. Moreover, new drug targets were predicted based on these common DEGs, and molecular docking and molecular dynamics (MD) simulations were performed. Finally, a diagnostic model of COVID-19 was established based on these common DEGs. Taken together, the molecular and signaling pathways identified in this study may be related to the mechanisms by which SARS-CoV-2 infection affects renal function. These findings are significant for the effective treatment of COVID-19 in patients with kidney diseases.


Sujets)
Atteinte rénale aigüe , COVID-19 , Insuffisance rénale chronique , Humains , COVID-19/complications , COVID-19/génétique , SARS-CoV-2 , Simulation de docking moléculaire , Atteinte rénale aigüe/génétique , Insuffisance rénale chronique/génétique , Protéines adaptatrices de la transduction du signal
4.
J Med Case Rep ; 17(1): 153, 2023 Apr 07.
Article Dans Anglais | MEDLINE | ID: covidwho-2257072

Résumé

BACKGROUND: Acute kidney injury is now recognized as a common complication of coronavirus disease 2019, affecting up to 46% of patients, with acute tubular injury as the most common etiology. Recently, we have seen an increase in cases of collapsing glomerulonephritis in patients with coronavirus disease 2019, also known as coronavirus disease 2019-associated nephropathy. It has been noted to be seen with a higher incidence in African American patients who are carriers of the APOL1 variant allele. CASE PRESENTATION: A 47-year-old African American male with a past medical history of asthma presented to the emergency department with complaints of intermittent chest pain, shortness of breath, and worsening confusion. On admission, he was found to be hemodynamically stable, but labs were significant for elevated creatinine and blood urea nitrogen, signifying acute kidney injury. He was admitted and taken for emergent dialysis. During his hospitalization, he was found to be positive for coronavirus disease 2019. Renal biopsy was done, which showed collapsing glomerulopathy, and the patient continues to require outpatient dialysis after discharge. CONCLUSION: Collapsing glomerulonephritis has emerged as a complication in patients with coronavirus disease 2019. This condition should be particularly suspected in African American patients who present with acute kidney injury, nephrotic-range proteinuria, and who are positive for coronavirus disease 2019. Current treatment options are limited to supportive treatment and renal replacement therapy. More clinical cases and trials are needed to better understand and improve therapeutic outcomes in these patients.


Sujets)
Atteinte rénale aigüe , Apolipoprotéine L1 , , COVID-19 , Glomérulonéphrite , Humains , Mâle , Adulte d'âge moyen , Atteinte rénale aigüe/étiologie , Atteinte rénale aigüe/génétique , Atteinte rénale aigüe/anatomopathologie , Atteinte rénale aigüe/thérapie , Apolipoprotéine L1/génétique , Biopsie , COVID-19/complications , Glomérulonéphrite/étiologie , Glomérulonéphrite/génétique , Glomérulonéphrite/anatomopathologie , Glomérulonéphrite/thérapie , Rein/anatomopathologie , Dialyse rénale
5.
Clin J Am Soc Nephrol ; 16(12): 1790-1796, 2021 12.
Article Dans Anglais | MEDLINE | ID: covidwho-2232275

Résumé

BACKGROUND AND OBJECTIVES: Black Americans have a higher incidence of kidney disease compared with populations that do not have recent African ancestry. Two risk variants in the APOL1 are responsible for a portion of this higher risk. We sought to assess the odds of AKI conferred by APOL1 risk alleles in patients with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. DESIGN, SETTING, PARTICIPANTS, & MEASUREMENTS: Black Americans who tested positive for coronavirus disease 2019 (COVID-19) were genotyped to determine APOL1 risk allele status. We assessed the incidence of AKI, persistent AKI, and AKI requiring KRT within 21 days of the PCR-based diagnosis. Outcomes were adjusted for age, sex, body mass index, hypertension, eGFR, and use of angiotensin-converting enzyme inhibitor/angiotensin receptor blocker. RESULTS: In total, 126 cases of SARS-CoV-2 infection were included within a 5-month period, with 16 (13%) and 110 (87%) cases with two and zero/one APOL1 high-risk alleles, respectively. AKI occurred in 11 (69%) patients with two APOL1 high-risk alleles and 39 (35%) patients with zero/one high-risk alleles (adjusted odds ratio, 4.41; 95% confidence interval, 1.11 to 17.52; P=0.04). Persistent AKI occurred in eight (50%) patients with two APOL1 high-risk alleles and 21 (19%) of those with zero/one high-risk alleles (adjusted odds ratio, 3.53; 95% confidence interval, 1.8 to 11.57; P=0.04). AKI KRT occurred in four (25%) of those with two APOL1 high-risk alleles and eight (7%) of those with zero/one high-risk alleles (adjusted odds ratio, 4.99; 95% confidence interval, 1.02 to 24.4, P=0.05). CONCLUSIONS: APOL1 high-risk alleles are associated with greater odds of AKI in Black American patients with COVID-19.


Sujets)
Atteinte rénale aigüe , COVID-19 , Humains , /génétique , Apolipoprotéine L1/génétique , COVID-19/génétique , SARS-CoV-2 , Génotype , Atteinte rénale aigüe/génétique , Facteurs de risque , Apolipoprotéines/génétique
6.
Front Immunol ; 13: 931210, 2022.
Article Dans Anglais | MEDLINE | ID: covidwho-2065505

Résumé

Atypical hemolytic uremic syndrome (aHUS) an important form of a thrombotic microangiopathy (TMA) that can frequently lead to acute kidney injury (AKI). An important subset of aHUS is the anti-factor H associated aHUS. This variant of aHUS can occur due to deletion of the complement factor H genes, CFHR1 and CFHR3, along with the presence of anti-factor H antibodies. However, it is a point of interest to note that not all patients with anti-factor H associated aHUS have a CFHR1/R3 deletion. Factor-H has a vital role in the regulation of the complement system, specifically the alternate pathway. Therefore, dysregulation of the complement system can lead to inflammatory or autoimmune diseases. Patients with this disease respond well to treatment with plasma exchange therapy along with Eculizumab and immunosuppressant therapy. Anti-factor H antibody associated aHUS has a certain genetic predilection therefore there is focus on further advancements in the diagnosis and management of this disease. In this article we discuss the baseline characteristics of patients with anti-factor H associated aHUS, their triggers, various treatment modalities and future perspectives.


Sujets)
Atteinte rénale aigüe , Syndrome hémolytique et urémique atypique , Protéines du système du complément , Atteinte rénale aigüe/génétique , Atteinte rénale aigüe/immunologie , Atteinte rénale aigüe/thérapie , Anticorps/génétique , Anticorps/immunologie , Syndrome hémolytique et urémique atypique/complications , Syndrome hémolytique et urémique atypique/génétique , Syndrome hémolytique et urémique atypique/immunologie , Syndrome hémolytique et urémique atypique/thérapie , Protéines du sang/génétique , Protéines inhibitrices de la fraction C3b du complément/génétique , Facteur H du complément/antagonistes et inhibiteurs , Facteur H du complément/génétique , Facteur H du complément/immunologie , Protéines du système du complément/génétique , Protéines du système du complément/immunologie , Humains , Échange plasmatique
7.
Genome Med ; 14(1): 103, 2022 09 09.
Article Dans Anglais | MEDLINE | ID: covidwho-2021328

Résumé

BACKGROUND: Acute kidney injury (AKI) occurs frequently in critically ill patients and is associated with adverse outcomes. Cellular mechanisms underlying AKI and kidney cell responses to injury remain incompletely understood. METHODS: We performed single-nuclei transcriptomics, bulk transcriptomics, molecular imaging studies, and conventional histology on kidney tissues from 8 individuals with severe AKI (stage 2 or 3 according to Kidney Disease: Improving Global Outcomes (KDIGO) criteria). Specimens were obtained within 1-2 h after individuals had succumbed to critical illness associated with respiratory infections, with 4 of 8 individuals diagnosed with COVID-19. Control kidney tissues were obtained post-mortem or after nephrectomy from individuals without AKI. RESULTS: High-depth single cell-resolved gene expression data of human kidneys affected by AKI revealed enrichment of novel injury-associated cell states within the major cell types of the tubular epithelium, in particular in proximal tubules, thick ascending limbs, and distal convoluted tubules. Four distinct, hierarchically interconnected injured cell states were distinguishable and characterized by transcriptome patterns associated with oxidative stress, hypoxia, interferon response, and epithelial-to-mesenchymal transition, respectively. Transcriptome differences between individuals with AKI were driven primarily by the cell type-specific abundance of these four injury subtypes rather than by private molecular responses. AKI-associated changes in gene expression between individuals with and without COVID-19 were similar. CONCLUSIONS: The study provides an extensive resource of the cell type-specific transcriptomic responses associated with critical illness-associated AKI in humans, highlighting recurrent disease-associated signatures and inter-individual heterogeneity. Personalized molecular disease assessment in human AKI may foster the development of tailored therapies.


Sujets)
Atteinte rénale aigüe , COVID-19 , Atteinte rénale aigüe/génétique , COVID-19/génétique , Maladie grave , Humains , Rein , Transcriptome
8.
Kidney360 ; 2(7): 1095-1106, 2021 07 29.
Article Dans Anglais | MEDLINE | ID: covidwho-1776832

Résumé

Background: The actions of angiotensin-converting enzyme 2 (ACE2) oppose those of the renin-angiotensin-aldosterone system. ACE2 may be a cytoprotectant in some tissues. This study examined ACE2 expression in models of AKI. Methods: ACE2 mRNA and protein expression and ACE2 activity were assessed in murine ischemic AKI. Renal ACE2 mRNA expression was evaluated in LPS-induced AKI in wild-type (C57BL/6J) mice, in heme oxygenase-1+/+ and heme oxygenase-1-/- mice, and after unilateral ureteral obstruction (UUO) in wild-type mice. The effect of sex and age on renal ACE2 protein expression was also assessed. Results: In ischemic AKI, ACE2 mRNA and protein expression and ACE2 activity were reduced as compared with such indices in the intact kidney. In ischemic AKI, ACE2, which, in health, is prominently expressed in the tubular epithelium, especially proximal tubules, is decreased in expression in these segments. Decreased ACE2 expression in AKI did not reflect reduced GFR, because ACE2 mRNA expression was unaltered after UUO. LPS induced renal ACE2 mRNA expression in wild-type mice, but this effect did not occur in heme oxygenase-1-deficient mice. In ischemic and LPS-induced AKI, renal expression of the Mas receptor was increased. In the intact kidney, renal ACE2 protein expression decreased in female mice as compared with male mice, but was unaltered with age. Conclusion: We conclude that renal ACE2 expression is decreased in ischemic AKI, characterized by decreased GFR and abundant cell death, but is upregulated in LPS-induced AKI, an effect requiring heme oxygenase-1. Determining the significance of ACE2 expression in experimental AKI merits further study. We suggest that understanding the mechanism underlying ACE2 downregulation in AKI may offer insights relevant to COVID-19: ACE2 expression is downregulated after ACE2 mediates SARS-CoV-2 cellular entry; such downregulation is proinflammatory; and AKI commonly occurs and determines outcomes in COVID-19.


Sujets)
Atteinte rénale aigüe , Angiotensin-converting enzyme 2 , Atteinte rénale aigüe/génétique , Angiotensin-converting enzyme 2/génétique , Animaux , Femelle , Rein , Mâle , Souris , Souris de lignée C57BL , Souris knockout
9.
JAMA Intern Med ; 182(4): 386-395, 2022 04 01.
Article Dans Anglais | MEDLINE | ID: covidwho-1653126

Résumé

IMPORTANCE: Coronavirus disease 2019 (COVID-19) confers significant risk of acute kidney injury (AKI). Patients with COVID-19 with AKI have high mortality rates. OBJECTIVE: Individuals with African ancestry with 2 copies of apolipoprotein L1 (APOL1) variants G1 or G2 (high-risk group) have significantly increased rates of kidney disease. We tested the hypothesis that the APOL1 high-risk group is associated with a higher-risk of COVID-19-associated AKI and death. DESIGN, SETTING, AND PARTICIPANTS: This retrospective cohort study included 990 participants with African ancestry enrolled in the Million Veteran Program who were hospitalized with COVID-19 between March 2020 and January 2021 with available genetic information. EXPOSURES: The primary exposure was having 2 APOL1 risk variants (RV) (APOL1 high-risk group), compared with having 1 or 0 risk variants (APOL1 low-risk group). MAIN OUTCOMES AND MEASURES: The primary outcome was AKI. The secondary outcomes were stages of AKI severity and death. Multivariable logistic regression analyses adjusted for preexisting comorbidities, medications, and inpatient AKI risk factors; 10 principal components of ancestry were performed to study these associations. We performed a subgroup analysis in individuals with normal kidney function prior to hospitalization (estimated glomerular filtration rate ≥60 mL/min/1.73 m2). RESULTS: Of the 990 participants with African ancestry, 905 (91.4%) were male with a median (IQR) age of 68 (60-73) years. Overall, 392 (39.6%) patients developed AKI, 141 (14%) developed stages 2 or 3 AKI, 28 (3%) required dialysis, and 122 (12.3%) died. One hundred twenty-five (12.6%) of the participants were in the APOL1 high-risk group. Patients categorized as APOL1 high-risk group had significantly higher odds of AKI (adjusted odds ratio [OR], 1.95; 95% CI, 1.27-3.02; P = .002), higher AKI severity stages (OR, 2.03; 95% CI, 1.37-2.99; P < .001), and death (OR, 2.15; 95% CI, 1.22-3.72; P = .007). The association with AKI persisted in the subgroup with normal kidney function (OR, 1.93; 95% CI, 1.15-3.26; P = .01). Data analysis was conducted between February 2021 and April 2021. CONCLUSIONS AND RELEVANCE: In this cohort study of veterans with African ancestry hospitalized with COVID-19 infection, APOL1 kidney risk variants were associated with higher odds of AKI, AKI severity, and death, even among individuals with prior normal kidney function.


Sujets)
Atteinte rénale aigüe , COVID-19 , Anciens combattants , Atteinte rénale aigüe/génétique , /génétique , Sujet âgé , Apolipoprotéine L1/génétique , Études de cohortes , Femelle , Hospitalisation , Humains , Mâle , Adulte d'âge moyen , Études rétrospectives , Facteurs de risque
10.
Adv Sci (Weinh) ; 9(3): e2103248, 2022 01.
Article Dans Anglais | MEDLINE | ID: covidwho-1527412

Résumé

COVID-19 is infected by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and can cause severe multiple organ injury and death. Kidney is one of major target organs of COVID-19 and acute kidney injury (AKI) is common in critically ill COVID-19 patients. However, mechanisms through which COVID-19 causes AKI remain largely unknown and treatment remains unspecific and ineffective. Here, the authors report that normal kidney-specifically overexpressing SARS-CoV-2 N develops AKI, which worsens in mice under ischemic condition. Mechanistically, it is uncovered that SARS-CoV-2 N-induced AKI is Smad3-dependent as SARS-CoV-2 N protein can interact with Smad3 and enhance TGF-ß/Smad3 signaling to cause tubular epithelial cell death and AKI via the G1 cell cycle arrest mechanism. This is further confirmed in Smad3 knockout mice and cells in which deletion of Smad3 protects against SARS-CoV-2 N protein-induced cell death and AKI in vivo and in vitro. Most significantly, it is also found that targeting Smad3 with a Smad3 pharmacological inhibitor is able to inhibit SARS-CoV-2 N-induced AKI. In conclusion, the authors identify that SARS-CoV-2 N protein is a key mediator for AKI and induces AKI via the Smad3-dependent G1 cell cycle arrest mechanism. Targeting Smad3 may represent as a novel therapy for COVID-19-asscoaited AKI.


Sujets)
Atteinte rénale aigüe , COVID-19 , Protéines de la nucléocapside des coronavirus , Points de contrôle de la phase G1 du cycle cellulaire , SARS-CoV-2 , Protéine Smad-3 , Atteinte rénale aigüe/génétique , Atteinte rénale aigüe/métabolisme , Atteinte rénale aigüe/virologie , Animaux , COVID-19/génétique , COVID-19/métabolisme , Lignée cellulaire , Protéines de la nucléocapside des coronavirus/génétique , Protéines de la nucléocapside des coronavirus/métabolisme , Modèles animaux de maladie humaine , Cellules HEK293 , Humains , Souris , Souris knockout , Phosphoprotéines/génétique , Phosphoprotéines/métabolisme , SARS-CoV-2/génétique , SARS-CoV-2/métabolisme , Protéine Smad-3/génétique , Protéine Smad-3/métabolisme
11.
Cell Commun Signal ; 19(1): 76, 2021 07 13.
Article Dans Anglais | MEDLINE | ID: covidwho-1318284

Résumé

Hypoxia is a pathological condition common to many diseases, although multiple organ injuries induced by hypoxia are often overlooked. There is increasing evidence to suggest that the hypoxic environment may activate innate immune cells and suppress adaptive immunity, further stimulating inflammation and inhibiting immunosurveillance. We found that dysfunctional immune regulation may aggravate hypoxia-induced tissue damage and contribute to secondary injury. Among the diverse mechanisms of hypoxia-induced immune dysfunction identified to date, the role of programmed death-ligand 1 (PD-L1) has recently attracted much attention. Besides leading to tumour immune evasion, PD-L1 has also been found to participate in the progression of the immune dysfunction which mediates hypoxia-induced multiple organ injury. In this review, we aimed to summarise the role of immune dysfunction in hypoxia-induced multiple organ injury, the effects of hypoxia on the cellular expression of PD-L1, and the effects of upregulated PD-L1 expression on immune regulation. Furthermore, we summarise the latest information pertaining to the involvement, diagnostic value, and therapeutic potential of immunosuppression induced by PD-L1 in various types of hypoxia-related diseases, including cancers, ischemic stroke, acute kidney injury, and obstructive sleep apnoea. Video Abstract.


Sujets)
Immunité acquise/génétique , Antigène CD274/immunologie , Inflammation/immunologie , Hypoxie tumorale/génétique , Atteinte rénale aigüe/génétique , Atteinte rénale aigüe/immunologie , Immunité acquise/immunologie , Antigène CD274/génétique , Humains , Immunité innée/génétique , Inflammation/génétique , Accident vasculaire cérébral ischémique/génétique , Accident vasculaire cérébral ischémique/immunologie , Monitorage immunologique , Tumeurs/génétique , Tumeurs/immunologie , Syndrome d'apnées obstructives du sommeil/génétique , Syndrome d'apnées obstructives du sommeil/immunologie , Hypoxie tumorale/immunologie
12.
Curr Opin Nephrol Hypertens ; 30(4): 444-449, 2021 07 01.
Article Dans Anglais | MEDLINE | ID: covidwho-1240971

Résumé

PURPOSE OF REVIEW: In this paper, we seek to review coronavirus disease 2019 (COVID-19) associated kidney injury with a focus on what is known about pathophysiology. RECENT FINDINGS: Kidney injury is a common complication of SARS-CoV-2 infection and is associated with increased morbidity and mortality. Acute tubular necrosis and glomerular injury are two common findings. Direct viral effect, endothelial dysfunction, and podocyte and tubular epithelial injury have been described. COVID-19-related glomerular injury may also be associated with high-risk APOL1 genotype. SUMMARY: Data on COVID-19 renal involvement have suggested novel mechanisms of kidney injury that need to be further elucidated. More data are needed on renal involvement in milder disease, renal-specific therapeutic interventions, and long-term sequelae.


Sujets)
Atteinte rénale aigüe/étiologie , Atteinte rénale aigüe/physiopathologie , COVID-19/complications , COVID-19/physiopathologie , Atteinte rénale aigüe/génétique , Atteinte rénale aigüe/thérapie , COVID-19/thérapie , Génotype , Humains , Maladies du rein/étiologie , Maladies du rein/génétique , Maladies du rein/physiopathologie , Maladies du rein/thérapie
13.
J Int Med Res ; 49(5): 3000605211015555, 2021 May.
Article Dans Anglais | MEDLINE | ID: covidwho-1228969

Résumé

Coronavirus disease 2019 (COVID-19) is an emerging infectious disease caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), which began as an outbreak in Wuhan, China and has spread rapidly across the globe. Although most infections are mild, patients with severe and critical COVID-19 infections face deterioration of respiratory function and may also have extrapulmonary manifestations, mostly affecting the kidney, digestive tract, heart, and nervous system. Here, we prospectively evaluated the presence of SARS-CoV-2 genetic material using reverse-transcription polymerase chain reaction in urine samples obtained from patients with COVID-19 receiving critical care. Among 51 included patients, we found higher serum creatinine levels, a longer hospital stay, and more frequent need for dialysis in urine-positive patients. These findings could suggest that, in predisposed patients, a direct viral cytopathic effect may contribute to a more severe disease phenotype.


Sujets)
Atteinte rénale aigüe , COVID-19 , Atteinte rénale aigüe/diagnostic , Atteinte rénale aigüe/génétique , Chine/épidémiologie , Humains , RT-PCR , SARS-CoV-2 , Indice de gravité de la maladie
14.
J Am Soc Nephrol ; 31(4): 716-730, 2020 04.
Article Dans Anglais | MEDLINE | ID: covidwho-992927

Résumé

BACKGROUND: Although AKI lacks effective therapeutic approaches, preventive strategies using preconditioning protocols, including caloric restriction and hypoxic preconditioning, have been shown to prevent injury in animal models. A better understanding of the molecular mechanisms that underlie the enhanced resistance to AKI conferred by such approaches is needed to facilitate clinical use. We hypothesized that these preconditioning strategies use similar pathways to augment cellular stress resistance. METHODS: To identify genes and pathways shared by caloric restriction and hypoxic preconditioning, we used RNA-sequencing transcriptome profiling to compare the transcriptional response with both modes of preconditioning in mice before and after renal ischemia-reperfusion injury. RESULTS: The gene expression signatures induced by both preconditioning strategies involve distinct common genes and pathways that overlap significantly with the transcriptional changes observed after ischemia-reperfusion injury. These changes primarily affect oxidation-reduction processes and have a major effect on mitochondrial processes. We found that 16 of the genes differentially regulated by both modes of preconditioning were strongly correlated with clinical outcome; most of these genes had not previously been directly linked to AKI. CONCLUSIONS: This comparative analysis of the gene expression signatures in preconditioning strategies shows overlapping patterns in caloric restriction and hypoxic preconditioning, pointing toward common molecular mechanisms. Our analysis identified a limited set of target genes not previously known to be associated with AKI; further study of their potential to provide the basis for novel preventive strategies is warranted. To allow for optimal interactive usability of the data by the kidney research community, we provide an online interface for user-defined interrogation of the gene expression datasets (http://shiny.cecad.uni-koeln.de:3838/IRaP/).


Sujets)
Atteinte rénale aigüe/génétique , Atteinte rénale aigüe/prévention et contrôle , Restriction calorique , Hypoxie , Préconditionnement ischémique/méthodes , ARN messager/métabolisme , Lésion d'ischémie-reperfusion/génétique , Lésion d'ischémie-reperfusion/prévention et contrôle , Animaux , Analyse de profil d'expression de gènes , Mâle , Souris , Souris de lignée C57BL , ARN messager/génétique
15.
Aging (Albany NY) ; 13(8): 10821-10832, 2020 11 21.
Article Dans Anglais | MEDLINE | ID: covidwho-946449

Résumé

BACKGROUND: Novel Coronavirus disease 2019 (COVID-19) was first detected in pneumonia patients in Wuhan, China in December 2019. Based on the current understanding, COVID-19 has become a global issue. Presumably, numerous studies have found that SARS-CoV-2 also transpires in kidney tissue with permanent viral loads. However, it is elusive as to whether SARS-CoV-2 can directly damage the kidney or induce acute renal failure. Hence, to comprehensively understand the impact of COVID-19 on kidney damage, we conducted a retrospective series of case studies to assess kidney functions. Additionally, ACE2 distribution in kidney tissue was analyzed through RNAseq data in open-access databases. RESULTS: According to the findings from transcriptome analysis, we revealed higher ACE2 expression levels in females than males. Similar results were more noticeable in the elderly than in young adults. Furthermore, single-cell RNA sequencing data analysis showed high ACE2 expression in kidney tubule and collecting duct principal cells as well as glomerular parietal epithelial cells. On their admission, the patient's serum creatinine and blood urea nitrogen (BUN) were elevated to between 36.13% and 16.80%, respectively. The estimated glomerular filtration rate (EGFR) of < 60 ml/min per 1.73 m2 was reported in 10.92 % of the patients. Notably, at admission, increased BUN time varied linearly following the generalized additive mixed model. Thus, the hourly-increase of BUN in patients was 0.495 (95%CI: 0.263, 0.726). CONCLUSION: Based on clinical findings, it was ascertained that COVID-19 can damage renal function, but it seldom causes acute renal failure. Coronavirus may directly bind to ACE2-positive cells and damage kidney tissue in the analysis of scRNA-seq data in kidney tissue. Therefore, this evidence suggests that kidney tissue act as the SARS-CoV-2 infection site and the findings could provide insight into the pathophysiology of kidney damage. METHODS: We systematically analyzed ACE2 expression profiles in organs based on open-access datasets for healthy individuals. Meanwhile, single-cell sequencing data for kidney samples were collected and analyzed. Assessments on kidney functions were conducted on 119 selected COVID-19 positive patients admitted from 10th February - 18th March 2020, in hospital in Wuhan City, Hubei Province. Consequently, their clinical records and laboratory findings, such as the estimated glomerular filtration rate (eGFR), Blood Urea Nitrogen (BUN), Creatinine, and Comorbidities, were collected.


Sujets)
Atteinte rénale aigüe , Angiotensin-converting enzyme 2 , COVID-19 , Transcriptome/génétique , Atteinte rénale aigüe/génétique , Atteinte rénale aigüe/métabolisme , Atteinte rénale aigüe/virologie , Adulte , Sujet âgé , Sujet âgé de 80 ans ou plus , Angiotensin-converting enzyme 2/analyse , Angiotensin-converting enzyme 2/génétique , Angiotensin-converting enzyme 2/métabolisme , COVID-19/complications , COVID-19/génétique , COVID-19/métabolisme , Bases de données génétiques , Femelle , Humains , Rein/métabolisme , Mâle , Adulte d'âge moyen , SARS-CoV-2 , Jeune adulte
16.
J Am Soc Nephrol ; 32(1): 33-40, 2021 01.
Article Dans Anglais | MEDLINE | ID: covidwho-937294

Résumé

BACKGROUND: Studies have documented AKI with high-grade proteinuria in patients with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. In some patients, biopsies have revealed collapsing glomerulopathy, a distinct form of glomerular injury that has been associated with other viruses, including HIV. Previous patient reports have described patients of African ancestry who developed nephrotic-range proteinuria and AKI early in the course of disease. METHODS: In this patient series, we identified six patients with coronavirus disease 2019 (COVID-19), AKI, and nephrotic-range proteinuria. COVID-19 was diagnosed by a positive nasopharyngeal swab RT-PCR for SARS-CoV-2 infection. We examined biopsy specimens from one transplanted kidney and five native kidneys. Three of the six patients underwent genetic analysis of APOL1, the gene encoding the APOL1 protein, from DNA extracted from peripheral blood. In addition, we purified genomic DNA from paraffin-embedded tissue and performed APOL1 genotype analysis of one of the native biopsies and the donor kidney graft. RESULTS: All six patients were of recent African ancestry. They developed COVID-19-associated AKI with podocytopathy, collapsing glomerulopathy, or both. Patients exhibited generally mild respiratory symptoms, and no patient required ventilator support. Genetic testing performed in three patients confirmed high-risk APOL1 genotypes. One APOL1 high-risk patient developed collapsing glomerulopathy in the engrafted kidney, which was transplanted from a donor who carried a low-risk APOL1 genotype; this contradicts current models of APOL1-mediated kidney injury, and suggests that intrinsic renal expression of APOL1 may not be the driver of nephrotoxicity and specifically, of podocyte injury. CONCLUSIONS: Glomerular disease presenting as proteinuria with or without AKI is an important presentation of COVID-19 infection and may be associated with a high-risk APOL1 genotype.


Sujets)
Atteinte rénale aigüe/étiologie , Apolipoprotéine L1/génétique , , COVID-19/complications , Glomérule rénal/physiopathologie , SARS-CoV-2 , Atteinte rénale aigüe/ethnologie , Atteinte rénale aigüe/génétique , Atteinte rénale aigüe/physiopathologie , /génétique , Apolipoprotéine L1/physiologie , Biopsie , Néphropathies diabétiques/complications , Femelle , Prédisposition génétique à une maladie , Génotype , Hématurie/étiologie , Humains , Hypertension artérielle/complications , Glomérule rénal/anatomopathologie , Transplantation rénale , Mâle , Adulte d'âge moyen , Modèles biologiques , Podocytes/anatomopathologie , Podocytes/virologie , Protéinurie/étiologie , Risque , SARS-CoV-2/pathogénicité , Tropisme viral
17.
J Am Soc Nephrol ; 31(8): 1688-1695, 2020 Aug.
Article Dans Anglais | MEDLINE | ID: covidwho-633952

Résumé

BACKGROUND: Kidney involvement is a feature of COVID-19 and it can be severe in Black patients. Previous research linked increased susceptibility to collapsing glomerulopathy, including in patients with HIV-associated nephropathy, to apo L1 (APOL1) variants that are more common in those of African descent. METHODS: To investigate genetic, histopathologic, and molecular features in six Black patients with COVID-19 presenting with AKI and de novo nephrotic-range proteinuria, we obtained biopsied kidney tissue, which was examined by in situ hybridization for viral detection and by NanoString for COVID-19 and acute tubular injury-associated genes. We also collected peripheral blood for APOL1 genotyping. RESULTS: This case series included six Black patients with COVID-19 (four men, two women), mean age 55 years. At biopsy day, mean serum creatinine was 6.5 mg/dl and mean urine protein-creatinine ratio was 11.5 g. Kidney biopsy specimens showed collapsing glomerulopathy, extensive foot process effacement, and focal/diffuse acute tubular injury. Three patients had endothelial reticular aggregates. We found no evidence of viral particles or SARS-CoV-2 RNA. NanoString showed elevated chemokine gene expression and changes in expression of genes associated with acute tubular injury compared with controls. All six patients had an APOL1 high-risk genotype. Five patients needed dialysis (two of whom died); one partially recovered without dialysis. CONCLUSIONS: Collapsing glomerulopathy in Black patients with COVID-19 was associated with high-risk APOL1 variants. We found no direct viral infection in the kidneys, suggesting a possible alternative mechanism: a "two-hit" combination of genetic predisposition and cytokine-mediated host response to SARS-CoV-2 infection. Given this entity's resemblance with HIV-associated nephropathy, we propose the term COVID-19-associated nephropathy to describe it.


Sujets)
Atteinte rénale aigüe/génétique , Apolipoprotéine L1/génétique , Infections à coronavirus/génétique , Glomérule rénal/virologie , Pneumopathie virale/génétique , Atteinte rénale aigüe/complications , Adulte , Sujet âgé , Allèles , Biopsie , , COVID-19 , Infections à coronavirus/complications , Créatinine/sang , Femelle , Génotype , Humains , Rein/anatomopathologie , Glomérule rénal/physiopathologie , Tubules rénaux/anatomopathologie , Mâle , Adulte d'âge moyen , Pandémies , Pneumopathie virale/complications , Risque
SÉLECTION CITATIONS
Détails de la recherche