Your browser doesn't support javascript.
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
Saudi J Kidney Dis Transpl ; 32(6): 1523-1544, 2021.
Article in English | MEDLINE | ID: covidwho-1975051

ABSTRACT

The major targets of coronavirus disease 2019 (COVID-19) are the respiratory and immune systems. However, a significant proportion of hospitalized patients had kidney dysfunction. The histopathological surveys have principally focused on respiratory, hematopoietic, and immune systems, whereas histopathologic data of kidney injury are lacking. Our study aimed to summarize the renal histopathological findings in COVID-19 from the published case report and case series. We conducted a systematic searching of databases such as MEDLINE, EMBASE, and Cochrane Library for published reports of COVID-19 patients with renal histopathological changes from autopsy studies and from "for cause" indication biopsies. Included in our study are case reports and case series with extractable quantitative data on patient demographics such as age, sex, ethnicity, as well as data on renal function tests, their comorbidities, and biopsy to study the histopathological changes. Data were analyzed with Microsoft Excel. To evaluate the methodological quality, we chose the framework for appraisal, synthesis, and application of evidence suggested by Murad et al. Systematic searches of literature found 31 studies that fulfilled the eligibility criteria. These studies included a total of 139 cases, where individual case details including clinical and histopathological findings were available. The median age of the cases was 62 years with a male:female ratio of 2.5:1. Associated comorbidities were noted in 78.4% of cases. The majority of the cases had renal dysfunction with proteinuria which was documented in more than two-thirds of the cases. The histopathological findings observed the frequent tubular involvement manifested by acute tubular injury. Regarding glomerular pathology, collapsing glomerulopathy emerged as a distinct lesion in these patients and was noted among 46.8% of cases with glomerular lesions. A small subset of cases (4.3%) had thrombotic microangiopathy. Collapsing glomerulopathy emerged as a hallmark of glomerular changes among COVID-19 patients. Tubular damage is common and is linked to multiple factors including ischemia, sepsis among others. In the form of thrombotic microangiopathy seen in a subset of patients, vascular damage hints toward the hyper-coagulable state associated with the infection. The demonstration of viral particles in renal tissue remains debatable and requires further study.


Subject(s)
COVID-19 , Kidney Diseases , Thrombotic Microangiopathies , Female , Humans , Kidney/pathology , Kidney/physiology , Kidney Diseases/pathology , Kidney Glomerulus/pathology , Male , Middle Aged , Proteinuria/etiology , Thrombotic Microangiopathies/complications
2.
J Investig Med High Impact Case Rep ; 10: 23247096221093888, 2022.
Article in English | MEDLINE | ID: covidwho-1808264

ABSTRACT

We report a case of dialysis dependence in a patient with COVID-19-associated nephropathy (COVAN) who had minimal respiratory manifestations. A 25-year-old man with a history of multiple sclerosis in remission presented with mild dyspnea due to COVID-19 pneumonia and was found to have rapidly worsening kidney function. He only required nasal cannula and was able to be weaned off within a few days. Despite having only mild respiratory disease, his kidney function worsened and urgent hemodialysis was started for hyperkalemia and uremic encephalopathy. Kidney biopsy demonstrated collapsing glomerulopathy due to COVID-19 with moderate interstitial fibrosis and tubular atrophy. His kidney function did not recover, and he unfortunately now has been dependent on hemodialysis for over 3 months. Multiple case reports have described COVAN causing dialysis dependence, but to our knowledge this is the first reported case of COVAN causing dialysis dependence in a patient with such mild respiratory disease. Currently the indications for intensive COVID-19 therapies are based on oxygen requirements. This case demonstrates that the oxygen requirement may not fully reflect the severity of COVID-19 and raises the question of whether these therapies should be considered in patients with COVAN.


Subject(s)
COVID-19 , Kidney Diseases , Adult , COVID-19/complications , Female , Humans , Kidney Diseases/pathology , Male , Oxygen , Renal Dialysis
3.
Transplant Proc ; 54(6): 1465-1470, 2022.
Article in English | MEDLINE | ID: covidwho-1713002

ABSTRACT

BACKGROUND: Collapsing glomerulopathy (CGN) secondary to HIV or COVID-19 infection mainly occurs in patients of African American descent due to APOL-1 gene mutations, but CGN is occasionally reported in white patients. CGNs are rarely reported in renal transplant biopsies and their association with idiopathic focal segmental glomerulosclerosis (FSGS) is unclear. METHODS AND RESULTS: Patient #1 was a 48-year-old Caucasian white man who had a renal transplant 8 years ago and was recently diagnosed with COVID-19 infection. Two weeks post infection, his serum creatinine (SCr) increased to 2.01 mg/dL from a baseline of 1.40 mg/dL, and he developed concomitant nephrotic range proteinuria. The first renal transplant biopsy showed FSGS. Four weeks later, his sCr level increased to 2.65 mg/dL with worsening proteinuria, and a second renal transplant biopsy revealed CGN. Patient #2 was a 32-year-old African American man whose native renal biopsy revealed primary FSGS. He received a renal transplant with initial post-transplant sCr level at 1.17 mg/dL. Four months later, his sCr and protein-to-creatinine ratio began to rise. Sequential biopsies revealed that the patient had developed recurrent FSGS, which progressed to show features of CGN. The CGN was further confirmed in his transplant kidney graft at autopsy later. CONCLUSIONS: This is the first case report of CGN in a white renal recipient with COVID-19 infection. The pathologic presentations of FSGS progressing to collapsing FSGS in our 2 renal transplant recipients suggest that FSGS and GGN may share a common pathophysiologic mechanism of podocytopathy.


Subject(s)
COVID-19 , Glomerulosclerosis, Focal Segmental , Kidney Diseases , Kidney Transplantation , Adult , Creatinine , Glomerulosclerosis, Focal Segmental/complications , Glomerulosclerosis, Focal Segmental/pathology , Humans , Kidney/pathology , Kidney Diseases/pathology , Kidney Transplantation/adverse effects , Male , Middle Aged , Proteinuria/complications
4.
Kidney Int ; 101(5): 1017-1026, 2022 05.
Article in English | MEDLINE | ID: covidwho-1700922

ABSTRACT

Collapsing glomerulopathy is a histologically distinct variant of focal and segmental glomerulosclerosis that presents with heavy proteinuria and portends a poor prognosis. Collapsing glomerulopathy can be triggered by viral infections such as HIV or SARS-CoV-2. Transcriptional profiling of collapsing glomerulopathy lesions is difficult since only a few glomeruli may exhibit this histology within a kidney biopsy and the mechanisms driving this heterogeneity are unknown. Therefore, we used recently developed digital spatial profiling (DSP) technology which permits quantification of mRNA at the level of individual glomeruli. Using DSP, we profiled 1,852 transcripts in glomeruli isolated from formalin fixed paraffin embedded sections from HIV or SARS-CoV-2-infected patients with biopsy-confirmed collapsing glomerulopathy and used normal biopsy sections as controls. Even though glomeruli with collapsing features appeared histologically similar across both groups of patients by light microscopy, the increased resolution of DSP uncovered intra- and inter-patient heterogeneity in glomerular transcriptional profiles that were missed in early laser capture microdissection studies of pooled glomeruli. Focused validation using immunohistochemistry and RNA in situ hybridization showed good concordance with DSP results. Thus, DSP represents a powerful method to dissect transcriptional programs of pathologically discernible kidney lesions.


Subject(s)
COVID-19 , Glomerulosclerosis, Focal Segmental , HIV Infections , Kidney Diseases , Female , Glomerulosclerosis, Focal Segmental/pathology , Humans , Kidney Diseases/genetics , Kidney Diseases/pathology , Kidney Glomerulus/pathology , Male , SARS-CoV-2
5.
Biomolecules ; 12(2)2022 02 12.
Article in English | MEDLINE | ID: covidwho-1686604

ABSTRACT

The onset of coronavirus disease (COVID-19) as a pandemic infection, has led to increasing insights on its pathophysiology and clinical features being revealed, such as a noticeable kidney involvement. In this study, we describe the histopathological, immunofluorescence, and ultrastructural features of biopsy-proven kidney injury observed in a series of SARS-CoV-2 positive cases in our institution from April 2020 to November 2021. We retrieved and retrospectively reviewed nine cases (two pediatric and seven adults) that experienced nephrotic syndrome (six cases), acute kidney injury (two cases), and a clinically silent microhematuria and leukocyturia. Kidney biopsies were investigated by means of light microscopy, direct immunofluorescence, and electron microscopy. The primary diagnoses were minimal change disease (four cases), acute tubular necrosis (two cases), collapsing glomerulopathy (two cases), and C3 glomerulopathy (one case). None of the cases showed viral or viral-like particles on ultrastructural analysis. Novel and specific histologic features on kidney biopsy related to SARS-CoV-2 infection have been gradually disclosed and reported, harboring relevant clinical and therapeutic implications. Recognizing and properly diagnosing renal involvement in patients experiencing COVID-19 could be challenging (due to the lack of direct proof of viral infection, e.g., viral particles) and requires a proper integration of clinical and pathological data.


Subject(s)
COVID-19/complications , Kidney Diseases/complications , Kidney Diseases/virology , Kidney/injuries , Kidney/virology , Adolescent , Aged , Aged, 80 and over , Biopsy , COVID-19/pathology , COVID-19/virology , Female , Humans , Italy , Kidney/pathology , Kidney/ultrastructure , Kidney Diseases/pathology , Male , Middle Aged , Retrospective Studies
6.
Kidney Int ; 101(6): 1216-1231, 2022 06.
Article in English | MEDLINE | ID: covidwho-1665244

ABSTRACT

Risk variants of the apolipoprotein-L1 (APOL1) gene are associated with severe kidney disease, putting homozygous carriers at risk. Since APOL1 lacks orthologs in all major model organisms, a wide range of mechanisms frequently in conflict have been described for APOL1-associated nephropathies. The genetic toolkit in Drosophila allows unique in vivo insights into disrupted cellular homeostasis. To perform a mechanistic analysis, we expressed human APOL1 control and gain-of-function kidney risk variants in the podocyte-like garland cells of Drosophila nephrocytes and a wing precursor tissue. Expression of APOL1 risk variants was found to elevate endocytic function of garland cell nephrocytes that simultaneously showed early signs of cell death. Wild-type APOL1 had a significantly milder effect, while a control transgene with deletion of the short BH3 domain showed no overt phenotype. Nephrocyte endo-lysosomal function and slit diaphragm architecture remained unaffected by APOL1 risk variants, but endoplasmic reticulum (ER) swelling, chaperone induction, and expression of the reporter Xbp1-EGFP suggested an ER stress response. Pharmacological inhibition of ER stress diminished APOL1-mediated cell death and direct ER stress induction enhanced nephrocyte endocytic function similar to expression of APOL1 risk variants. We confirmed APOL1-dependent ER stress in the Drosophila wing precursor where silencing the IRE1-dependent branch of ER stress signaling by inhibition with Xbp1-RNAi abrogated cell death, representing the first rescue of APOL1-associated cytotoxicity in vivo. Thus, we uncovered ER stress as an essential consequence of APOL1 risk variant expression in vivo in Drosophila, suggesting a central role of this pathway in the pathogenesis of APOL1-associated nephropathies.


Subject(s)
Kidney Diseases , Podocytes , Animals , Apolipoprotein L1/genetics , Drosophila/genetics , Endoplasmic Reticulum Stress/genetics , Humans , Kidney Diseases/pathology , Podocytes/pathology
7.
Int J Mol Sci ; 22(12)2021 Jun 20.
Article in English | MEDLINE | ID: covidwho-1472414

ABSTRACT

Acute kidney injury (AKI) and chronic kidney disease (CKD) are rising in global prevalence and cause significant morbidity for patients. Current treatments are limited to slowing instead of stabilising or reversing disease progression. In this review, we describe mesenchymal stem cells (MSCs) and their constituents, extracellular vesicles (EVs) as being a novel therapeutic for CKD. MSC-derived EVs (MSC-EVs) are membrane-enclosed particles, including exosomes, which carry genetic information that mimics the phenotype of their cell of origin. MSC-EVs deliver their cargo of mRNA, miRNA, cytokines, and growth factors to target cells as a form of paracrine communication. This genetically reprograms pathophysiological pathways, which are upregulated in renal failure. Since the method of exosome preparation significantly affects the quality and function of MSC-exosomes, this review compares the methodologies for isolating exosomes from MSCs and their role in tissue regeneration. More specifically, it summarises the therapeutic efficacy of MSC-EVs in 60 preclinical animal models of AKI and CKD and the cargo of biomolecules they deliver. MSC-EVs promote tubular proliferation and angiogenesis, and inhibit apoptosis, oxidative stress, inflammation, the epithelial-to-mesenchymal transition, and fibrosis, to alleviate AKI and CKD. By reprogramming these pathophysiological pathways, MSC-EVs can slow or even reverse the progression of AKI to CKD, and therefore offer potential to transform clinical practice.


Subject(s)
Biological Therapy , Extracellular Vesicles/metabolism , Extracellular Vesicles/transplantation , Kidney Diseases/therapy , Mesenchymal Stem Cells/metabolism , Acute Kidney Injury/diagnosis , Acute Kidney Injury/etiology , Acute Kidney Injury/metabolism , Acute Kidney Injury/therapy , Animals , Apoptosis/drug effects , Biological Therapy/methods , Cell Differentiation , Cell Proliferation/drug effects , Cell Self Renewal , Chemical Fractionation , Disease Management , Disease Susceptibility , Exosomes/metabolism , Humans , Kidney Diseases/etiology , Kidney Diseases/pathology , Mesenchymal Stem Cells/cytology , Protective Agents , Renal Insufficiency/diagnosis , Renal Insufficiency/etiology , Renal Insufficiency, Chronic/diagnosis , Renal Insufficiency, Chronic/etiology , Renal Insufficiency, Chronic/metabolism , Renal Insufficiency, Chronic/therapy
8.
Aust J Gen Pract ; 50(7): 441-444, 2021 07.
Article in English | MEDLINE | ID: covidwho-1289398

ABSTRACT

BACKGROUND: COVID-19 has been at the forefront of public and scientific attention since the initial report in December 2019. The kidney is one of the target organs of the causative SARS-CoV-2 virus. OBJECTIVE: The aim of this article is to discuss the current understanding of COVID-19 renal disease from a primary care perspective, with the caveat that our knowledge of the pathogenesis, clinical course and outcome of the disease is still rapidly evolving. DISCUSSION: The kidney is one of the target organs of the causative SARS-CoV-2 virus, affecting the endothelium, podocytes and renal tubular epithelial cells. Clinical presentation ranges from isolated proteinuria, haematuria to severe acute kidney injury (AKI) requiring renal replacement therapy. Renal dysfunction associated with COVID-19 has a worse prognosis whether it be in the form of AKI or worsening of pre-existing chronic kidney disease, or in patients undergoing renal replacement therapy.


Subject(s)
COVID-19/complications , COVID-19/therapy , Kidney Diseases/therapy , Kidney Diseases/virology , COVID-19/pathology , Humans , Kidney Diseases/pathology
9.
BMC Nephrol ; 22(1): 91, 2021 03 15.
Article in English | MEDLINE | ID: covidwho-1136213

ABSTRACT

BACKGROUND: COVID-19 infection in kidney transplant recipients often lead to allograft dysfunction. The allograft injury has various histopathological manifestations. Our case illustrates the unusual combination of allograft rejection, acute kidney injury secondary to oxalate nephropathy and SARS CoV-2 nephropathy as the cause of irreversible allograft failure. CASE PRESENTATION: A 56 year old renal allograft recipient presented with a history of fever and diarrhoea for the preceding 4 weeks, tested positive for Sars-CoV2 on nasal swab and was found to have severe allograft dysfunction, necessitating haemodialysis. He subsequently underwent an allograft biopsy, which demonstrated antibody mediated rejection along with the presence of extensive oxalate deposition in the tubules. Ultrastructural examination demonstrated spherical spiked particles in the glomerular capillary endothelium and the presence of tubulo-reticular inclusions suggestive of an active COVID-19 infection within the kidney. The intra-tubular oxalate deposition was considered to be the result of high dose, supplemental Vitamin C used as an immune booster in many patients with COVID - 19 infection in India. CONCLUSIONS: This case highlights the complex pathology that may be seen in following COVID-19 disease and the need for kidney biopsies in these patients to better understand the aetiology of disease.


Subject(s)
Ascorbic Acid/adverse effects , COVID-19/complications , Graft Rejection/etiology , Hyperoxaluria/complications , Kidney Transplantation , Primary Graft Dysfunction/etiology , Acute Kidney Injury/etiology , Ascorbic Acid/administration & dosage , COVID-19/diagnosis , Fatal Outcome , Humans , Kidney Diseases/complications , Kidney Diseases/pathology , Male , Middle Aged , Primary Graft Dysfunction/pathology , Primary Graft Dysfunction/virology
10.
Nephron ; 145(3): 275-279, 2021.
Article in English | MEDLINE | ID: covidwho-1127626

ABSTRACT

CONTEXT: Determining whether SARS-CoV-2 causes direct infection of the kidneys is challenging due to limitations in imaging and molecular tools. Subject of Review: A growing number of conflicting kidney biopsy and autopsy reports highlight this controversial issue. Second Opinion: Based on the collective evidence, therapies that improve hemodynamic stability and oxygenation, or dampen complement activation, are likely to ameliorate acute kidney injury in COVID-19. At this time, whether inhibition of viral infection and replication directly modulates kidney damage is inconclusive.


Subject(s)
COVID-19/complications , Kidney Diseases/etiology , Acute Kidney Injury/etiology , Autopsy , Biopsy , COVID-19/therapy , COVID-19/virology , Humans , Kidney/pathology , Kidney/virology , Kidney Diseases/pathology , Kidney Diseases/therapy , Kidney Diseases/virology , Nephritis, Interstitial/etiology
11.
Front Immunol ; 11: 599417, 2020.
Article in English | MEDLINE | ID: covidwho-1000095

ABSTRACT

The complement system comprises the frontline of the innate immune system. Triggered by pathogenic surface patterns in different pathways, the cascade concludes with the formation of a membrane attack complex (MAC; complement components C5b to C9) and C5a, a potent anaphylatoxin that elicits various inflammatory signals through binding to C5a receptor 1 (C5aR1). Despite its important role in pathogen elimination, priming and recruitment of myeloid cells from the immune system, as well as crosstalk with other physiological systems, inadvertent activation of the complement system can result in self-attack and overreaction in autoinflammatory diseases. Consequently, it constitutes an interesting target for specialized therapies. The paradigm of safe and efficacious terminal complement pathway inhibition has been demonstrated by the approval of eculizumab in paroxysmal nocturnal hematuria. In addition, complement contribution in rare kidney diseases, such as lupus nephritis, IgA nephropathy, atypical hemolytic uremic syndrome, C3 glomerulopathy, or antineutrophil cytoplasmic antibody-associated vasculitis has been demonstrated. This review summarizes the involvement of the terminal effector agents of the complement system in these diseases and provides an overview of inhibitors for complement components C5, C5a, C5aR1, and MAC that are currently in clinical development. Furthermore, a link between increased complement activity and lung damage in severe COVID-19 patients is discussed and the potential for use of complement inhibitors in COVID-19 is presented.


Subject(s)
Complement C5a/antagonists & inhibitors , Complement Inactivating Agents/therapeutic use , Complement Membrane Attack Complex/antagonists & inhibitors , Kidney Diseases/drug therapy , Receptor, Anaphylatoxin C5a/antagonists & inhibitors , Anti-Neutrophil Cytoplasmic Antibody-Associated Vasculitis/drug therapy , Anti-Neutrophil Cytoplasmic Antibody-Associated Vasculitis/pathology , Antibodies, Monoclonal, Humanized/therapeutic use , Complement Activation/immunology , Complement System Proteins/metabolism , Glomerulonephritis, IGA/drug therapy , Glomerulonephritis, IGA/pathology , Humans , Kidney/pathology , Kidney Diseases/immunology , Kidney Diseases/pathology , Lupus Nephritis/drug therapy , Lupus Nephritis/pathology
12.
J Mol Histol ; 51(6): 613-628, 2020 Dec.
Article in English | MEDLINE | ID: covidwho-813346

ABSTRACT

The outbreak of severe acute respiratory syndrome coronavirus 2 (SARS-CoV2) in December 2019 form Wuhan, China leads to coronavirus disease 2019 (COVID-19) pandemic. While the common cold symptoms are observed in mild cases, COVID-19 is accompanied by multiorgan failure in severe patients. The involvement of different organs in severe patients results in lengthening the hospitalization duration and increasing the mortality rate. In this review, we aimed to investigate the involvement of different organs in COVID-19 patients, particularly in severe cases. Also, we tried to define the potential underlying mechanisms of SARS-CoV2 induced multiorgan failure. The multi-organ dysfunction is characterized by acute lung failure, acute liver failure, acute kidney injury, cardiovascular disease, and as well as a wide spectrum of hematological abnormalities and neurological disorders. The most important mechanisms are related to the direct and indirect pathogenic features of SARS-CoV2. Although the presence of angiotensin-converting enzyme 2, a receptor of SARS-CoV2 in the lung, heart, kidney, testis, liver, lymphocytes, and nervous system was confirmed, there are controversial findings to about the observation of SARS-CoV2 RNA in these organs. Moreover, the organ failure may be induced by the cytokine storm, a result of increased levels of inflammatory mediators, endothelial dysfunction, coagulation abnormalities, and infiltration of inflammatory cells into the organs. Therefore, further investigations are needed to detect the exact mechanisms of pathogenesis. Since the involvement of several organs in COVID-19 patients is important for clinicians, increasing their knowledge may help to improve the outcomes and decrease the rate of mortality and morbidity.


Subject(s)
Coronavirus Infections/pathology , Heart Diseases/pathology , Kidney Diseases/pathology , Liver Diseases/pathology , Multiple Organ Failure/pathology , Pneumonia, Viral/pathology , Angiotensin-Converting Enzyme 2 , Betacoronavirus , COVID-19 , Cytokine Release Syndrome/pathology , Heart Diseases/virology , Humans , Kidney/pathology , Kidney Diseases/virology , Liver/pathology , Liver Diseases/virology , Lung/pathology , Multiple Organ Failure/virology , Myocardium/pathology , Pandemics , Peptidyl-Dipeptidase A/metabolism , SARS-CoV-2
14.
J Am Soc Nephrol ; 31(9): 1959-1968, 2020 Sep.
Article in English | MEDLINE | ID: covidwho-652873

ABSTRACT

BACKGROUND: Coronavirus disease 2019 (COVID-19) is thought to cause kidney injury by a variety of mechanisms. To date, pathologic analyses have been limited to patient reports and autopsy series. METHODS: We evaluated biopsy samples of native and allograft kidneys from patients with COVID-19 at a single center in New York City between March and June of 2020. We also used immunohistochemistry, in situ hybridization, and electron microscopy to examine this tissue for presence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). RESULTS: The study group included 17 patients with COVID-19 (12 men, 12 black; median age of 54 years). Sixteen patients had comorbidities, including hypertension, obesity, diabetes, malignancy, or a kidney or heart allograft. Nine patients developed COVID-19 pneumonia. Fifteen patients (88%) presented with AKI; nine had nephrotic-range proteinuria. Among 14 patients with a native kidney biopsy, 5 were diagnosed with collapsing glomerulopathy, 1 was diagnosed with minimal change disease, 2 were diagnosed with membranous glomerulopathy, 1 was diagnosed with crescentic transformation of lupus nephritis, 1 was diagnosed with anti-GBM nephritis, and 4 were diagnosed with isolated acute tubular injury. The three allograft specimens showed grade 2A acute T cell-mediated rejection, cortical infarction, or acute tubular injury. Genotyping of three patients with collapsing glomerulopathy and the patient with minimal change disease revealed that all four patients had APOL1 high-risk gene variants. We found no definitive evidence of SARS-CoV-2 in kidney cells. Biopsy diagnosis informed treatment and prognosis in all patients. CONCLUSIONS: Patients with COVID-19 develop a wide spectrum of glomerular and tubular diseases. Our findings provide evidence against direct viral infection of the kidneys as the major pathomechanism for COVID-19-related kidney injury and implicate cytokine-mediated effects and heightened adaptive immune responses.


Subject(s)
Betacoronavirus , Coronavirus Infections/pathology , Kidney/pathology , Pneumonia, Viral/pathology , Adult , Aged , Betacoronavirus/isolation & purification , Biopsy , COVID-19 , Coronavirus Infections/complications , Coronavirus Infections/immunology , Female , Humans , Kidney/ultrastructure , Kidney/virology , Kidney Diseases/pathology , Male , Middle Aged , Pandemics , Pneumonia, Viral/complications , Pneumonia, Viral/immunology , SARS-CoV-2
15.
Vet Q ; 40(1): 190-197, 2020 Dec.
Article in English | MEDLINE | ID: covidwho-598972

ABSTRACT

Background: The natural MERS-CoV infection in dromedary camels is understudied. Recent experimental studies showed no obvious clinical signs in the infected dromedary camels.Aim: To study the pathological changes associated with natural MERS-CoV infection in dromedary camels.Methods: Tissues from three MERS-CoV positive animals as well as two negative animals were collected and examined for the presence of pathological changes. The screening of the animals was carried out first by the rapid agglutination test and then confirmed by the RT-PCR. The selected animals ranged from six to twelve months in age. The sensitivity of the latter technique was much higher in the detection of MERS-CoV than the Rapid test (14 out of 75 animals positive or 18% versus 31 out of 75 positive or 41%).Results: MERS-CoV induced marked desquamation of the respiratory epithelium accompanied by lamina propria and submucosal mononuclear cells infiltration, epithelial hyperplasia in the respiratory tract, and interstitial pneumonia. Ciliary cell loss was seen in the trachea and turbinate. In addition, degeneration of glomerular capillaries with the complete destruction of glomerular tufts that were replaced with fibrinous exudate in renal corpuscles in the renal cortex were noticed. Expression of the MERS-CoV-S1 and MERS-CoV-N proteins was revealed in respiratory tract, and kidneys.Conclusion: To our knowledge, this is the first study describing the pathological changes of MERS-CoV infection in dromedary camels under natural conditions. In contrast to experimental infection in case of spontaneous infection interstitial pneumonea is evident at least in some affected animals.


Subject(s)
Camelus/virology , Coronavirus Infections/veterinary , Lung Diseases, Interstitial/veterinary , Middle East Respiratory Syndrome Coronavirus/isolation & purification , Animals , Coronavirus Infections/pathology , Coronavirus Infections/virology , Female , Kidney Diseases/pathology , Kidney Diseases/veterinary , Kidney Diseases/virology , Lung Diseases, Interstitial/pathology , Lung Diseases, Interstitial/virology , Male , Saudi Arabia , Viral Proteins/analysis
SELECTION OF CITATIONS
SEARCH DETAIL