1
|
Shaffer A, Meyerowitz EA. Clinical Manifestations of SARS-CoV-2 Infection in Immunocompetent Adults in the Era of Widespread Population Immunity and Omicron Sublineage Viruses. Infect Dis Clin North Am 2025; 39:233-251. [PMID: 40068975 DOI: 10.1016/j.idc.2025.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/10/2025]
Abstract
While most SARS-CoV-2 infections and reinfections in the era of widespread population immunity with omicron subsub-lineage variants are mild for immunocompetent individuals, any manifestation previously seen during the pandemic phase is still possible. COVID-19 may affect any organ system. Previous infections and prior vaccines protect against symptomatic future SARS-CoV-2 infections, though this protection wanes over time.
Collapse
Affiliation(s)
- Alexander Shaffer
- Division of Infectious Diseases, Department of Medicine, Montefiore Medical Center, 111 East 210th Street, Bronx, NY 10467, USA; Albert Einstein College of Medicine, Bronx, NY, USA
| | - Eric A Meyerowitz
- Division of Infectious Diseases, Department of Medicine, Montefiore Medical Center, 111 East 210th Street, Bronx, NY 10467, USA; Albert Einstein College of Medicine, Bronx, NY, USA.
| |
Collapse
|
2
|
Zhang DD, Liu Y, Wang W, Wu W, Chen J, Wan L, Wu L, Huang XR, Lan HY, Yu X. SARS-CoV-2 N protein induces hypokalemia in acute kidney injury mice via ENaC-dependent mechanism. Mol Ther 2025:S1525-0016(25)00363-6. [PMID: 40336195 DOI: 10.1016/j.ymthe.2025.04.043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Revised: 04/03/2025] [Accepted: 04/30/2025] [Indexed: 05/09/2025] Open
Abstract
Hypokalemia is a prevalent complication of COVID-19 patients with acute kidney injury (AKI); however, mechanisms have yet to be fully understood. By single-nucleus RNA sequencing, we found that COVID-19 patients with AKI were associated with a marked upregulation of the epithelial sodium channel (ENaC) in the renal tubular epithelial cells (TECs). By using a mouse model of AKI induced by kidney-specific overexpressing SARS-CoV-2 N protein, we detected that overexpression of renal SARS-CoV-2 N protein could induce hypokalemia and AKI, which was associated with the upregulation of ENaC, ROMK, and BK proteins. Functionally, a patch-clamp study revealed that the overexpression of SARS-CoV-2 N protein largely increased the ENaC current in the TECs. Mechanically, we uncovered that kidney-specific overexpressing SARS-CoV-2 N protein could activate ENaC to cause hypokalemia and AKI directly by binding to the ENaCα and ENaCγ subunits and indirectly by activating the p38 MAPK pathway. Importantly, treatment with an ENaC specific inhibitor could protect against SARS-CoV-2 N-induced hypokalemia and AKI, revealing a regulatory role and therapeutic target of ENaC in SARS-CoV-2 N-induced hypokalemia and AKI. In conclusion, hypokalemia in COVID-19 AKI is induced by SARS-CoV-2 N protein via the ENaC-dependent mechanism. Targeting ENaC may offer a novel therapy for COVID-19 patients with AKI.
Collapse
Affiliation(s)
- Dan-Dan Zhang
- Departments of Nephrology and Pathology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Science, Guangzhou, China; Department of Physiology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Yang Liu
- Cancer Institute, The First Hospital of Jilin University, Changchun, Jilin 130000, China
| | - Wenbiao Wang
- Departments of Nephrology and Pathology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Science, Guangzhou, China
| | - Wenjing Wu
- Departments of Nephrology and Pathology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Science, Guangzhou, China; Department of Nephrology, Hubei Provincial Hospital of Traditional Chinese Medicine, Affiliated Hospital of Hubei University of Chinese Medicine, Wuhan, China
| | - Junzhe Chen
- Department of Nephrology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
| | - Lin Wan
- Departments of Nephrology and Pathology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Science, Guangzhou, China
| | - Liumei Wu
- Departments of Nephrology and Pathology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Science, Guangzhou, China
| | - Xiao-Ru Huang
- Departments of Nephrology and Pathology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Science, Guangzhou, China; Departments of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
| | - Hui-Yao Lan
- Departments of Nephrology and Pathology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Science, Guangzhou, China; Departments of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China.
| | - Xueqing Yu
- Departments of Nephrology and Pathology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Science, Guangzhou, China.
| |
Collapse
|
3
|
Hachimi A, El-Mansoury B, Merzouki M. Incidence, pathophysiology, risk factors, histopathology, and outcomes of COVID-19-induced acute kidney injury: A narrative review. Microb Pathog 2025; 202:107360. [PMID: 39894232 DOI: 10.1016/j.micpath.2025.107360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 01/28/2025] [Accepted: 01/30/2025] [Indexed: 02/04/2025]
Abstract
The COVID-19 pandemic, caused by the SARS-CoV-2 virus, has led to a significant burden on global healthcare systems. COVID-19-induced acute kidney injury (AKI) is among one of the complications, that has emerged as a critical and frequent condition in COVID-19 patients. This AKI among COVID-19 patients is associated with poor outcomes, and high mortality rates, especially in those with severe AKI or requiring renal replacement therapy. COVID-19-induced AKI represents a significant complication with complex pathophysiology and multifactorial risk factors. Indeed, several pathophysiological mechanisms, including direct viral invasion of renal cells, systemic inflammation, endothelial and thrombotic abnormalities as well as nephrotoxic drugs and rhabdomyolysis are believed to underlie this condition. Moreover, histopathological and immunohistopathological findings commonly observed in postmortem studies include acute tubular necrosis, glomerular injury, and the presence of viral particles within renal tissue and urine. Identified risk factors for developing AKI vary among studies, depending on regions, underlying conditions, and the severity of the disease. Moreover, histopathological and immunohistopathological findings commonly observed in postmortem studies include show acute tubular necrosis, glomerular injury, and viral particles within renal tissue and urine. While, identified risk factors for developing AKI vary among studies, according to regions, underlying conditions, and the gravity of the disease. This narrative review aims to synthesize current knowledge on the incidence, pathophysiology, risk factors, histopathology, and outcomes of AKI induced by COVID-19.
Collapse
Affiliation(s)
- Abdelhamid Hachimi
- Medical ICU, Mohammed VI(th) University Hospital of Marrakech, Marrakech, Morocco; Morpho-Science Research Laboratory, Faculty of Medicine and Pharmacy, Cadi Ayyad University, Marrakech, Morocco; Life Sciences Department, Bioengineering Laboratory, Faculty of Sciences and Technics, Sultan Moulay Slimane University, Beni Mellal, Morocco
| | - Bilal El-Mansoury
- Nutritional Physiopathologies, Neuroscience and Toxicology Team, Laboratory of Anthropogenic, Biotechnology and Health, Faculty of Sciences, Chouaib Doukkali University, El Jadida, Morocco
| | - Mohamed Merzouki
- Life Sciences Department, Bioengineering Laboratory, Faculty of Sciences and Technics, Sultan Moulay Slimane University, Beni Mellal, Morocco.
| |
Collapse
|
4
|
Kissling VM, Eitner S, Bottone D, Cereghetti G, Wick P. Systematic Comparison of Commercial Uranyl-Alternative Stains for Negative- and Positive-Staining Transmission Electron Microscopy of Organic Specimens. Adv Healthc Mater 2025:e2404870. [PMID: 40302369 DOI: 10.1002/adhm.202404870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 03/31/2025] [Indexed: 05/02/2025]
Abstract
Negative- and positive-staining transmission electron microscopy (ns/psTEM) is a cornerstone of research and diagnostics, enabling nanometer-resolution analysis of organic specimens from nanoparticles to cells without requiring costly cryo-equipment. For nearly 70 years, uranyl salts like uranyl acetate (UA) have been the gold-standard ns/psTEM-stains. However, mounting safety concerns due to their high toxicity and radioactivity have led to stricter regulations and expensive licensing requirements. Consequently, there is an urgent global demand for safer, more sustainable stains that deliver uranyl-comparable, high-quality ns/psTEM. Here, the commercially available stain-alternatives UranyLess, UAR, UA-Zero, PTA, STAIN 77, Nano-W, NanoVan, and lead citrate are systematically assessed against UA. The stains are evaluated regarding their contrast, resolution, stain-distribution, and ease-of-use in ns/psTEM across a diverse sample set, including polymethylmethacrylate-nanoplastics, phosphatidylcholine-liposomes, Influenza-A viruses, globular ferritin, fibrillar pyruvate kinase amyloids, and human lung-carcinoma cell-sections. It is shown that for this variety of samples, a ready-to-use uranyl-alternative is commercially available with comparable or even superior ns/psTEM-performance to UA using an efficient staining-protocol. Furthermore, the GUIDE4U tool is developed for the fast identification of the appropriate uranyl-replacements for each sample of interest, saving ns/psTEM-users time and costs while ensuring excellent staining results for ultrastructural analysis, thereby further catalyzing the use of safer stains.
Collapse
Affiliation(s)
- Vera M Kissling
- Nanomaterials in Health Laboratory, Department of Materials Meet Life, Swiss Federal Laboratories for Materials Science and Technology (Empa), St. Gallen, 9014, Switzerland
| | - Stephanie Eitner
- Nanomaterials in Health Laboratory, Department of Materials Meet Life, Swiss Federal Laboratories for Materials Science and Technology (Empa), St. Gallen, 9014, Switzerland
| | - Davide Bottone
- Nanomaterials in Health Laboratory, Department of Materials Meet Life, Swiss Federal Laboratories for Materials Science and Technology (Empa), St. Gallen, 9014, Switzerland
| | - Gea Cereghetti
- Centre for Misfolding Diseases, Department of Chemistry, University of Cambridge, Cambridge, CB2 1EW, UK
| | - Peter Wick
- Nanomaterials in Health Laboratory, Department of Materials Meet Life, Swiss Federal Laboratories for Materials Science and Technology (Empa), St. Gallen, 9014, Switzerland
| |
Collapse
|
5
|
Marcellino A, Bloise S, Pirone C, Brandino G, Barberi A, Del Giudice E, Martucci V, Sanseviero M, Ventriglia F, Lubrano R. Increased Measured GFR and Proteinuria in Children with Previous Infection by SARS-CoV-2: Should We Be Concerned? Microorganisms 2025; 13:1008. [PMID: 40431181 DOI: 10.3390/microorganisms13051008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2025] [Revised: 04/12/2025] [Accepted: 04/19/2025] [Indexed: 05/29/2025] Open
Abstract
Over the past 3 years, several kidney complications in children with severe involvement by SARS-CoV-2 have been described. However, literature data are still lacking regarding possible kidney injury in children with paucisymptomatic SARS-CoV-2 infection. Thus, we retrospectively evaluated renal function in those patients. Children between 3 and 18 years, without any renal disease, with previous paucisymptomatic SARS-CoV-2 infection from May 2020 to March 2022, were recruited at our post-COVID-19 outpatient clinic. We retrospectively collected: Glomerular filtration rate, Fractional-excretion-of-sodium (FENa), tubular-reabsorption-of-phosphate (TRP), calcium-creatinine-urine ratio (CaU/CrU); proteinuria/m2/day and microhematuria by urine cytofluorometry. A total of 148 children were enrolled after a median period of 3 (IQR 6) months after infection. Twenty-six patients (17.6%) had reduced GFR, fifty (33.9%) had hyperfiltration, eleven (7.4%) had abnormal FENa and/or TRP, twenty-two (14.9%) had hypercalciuria, seventy-eight (52.7%) had pathological daily proteinuria. Microhematuria was found in sixteen (10.9%) subjects. Hyperfiltration was more prevalent among males (38.9% vs. 22.4%, p = 0.027); CaU/CrU [median 0.08 (IQR 0.09) vs. 0.13 (IQR 0.13) p = 0.003] was significantly higher in females. Our data suggest that SARS-CoV-2 could determine, in a significant proportion of children, kidney damage characterized by hyperfiltration, proteinuria, and hematuria, warranting strict follow-up in these patients.
Collapse
Affiliation(s)
- Alessia Marcellino
- Pediatrics and Neonatology Unit, Maternal-Child Department, Santa Maria Goretti Hospital, Sapienza University of Rome, 04100 Latina, Italy
| | - Silvia Bloise
- Pediatrics and Neonatology Unit, Maternal-Child Department, Santa Maria Goretti Hospital, Sapienza University of Rome, 04100 Latina, Italy
| | - Carmelo Pirone
- Dermatopathic Institute of the Immaculate (IDI-IRCCS), 00167 Rome, Italy
| | - Giulia Brandino
- Pediatrics and Neonatology Unit, Maternal-Child Department, Santa Maria Goretti Hospital, Sapienza University of Rome, 04100 Latina, Italy
| | - Antonio Barberi
- Radiology Unit, Santa Maria Goretti Hospital, Sapienza University of Rome, 04100 Latina, Italy
| | - Emanuela Del Giudice
- Pediatrics and Neonatology Unit, Maternal-Child Department, Santa Maria Goretti Hospital, Sapienza University of Rome, 04100 Latina, Italy
| | - Vanessa Martucci
- Pediatrics and Neonatology Unit, Maternal-Child Department, Santa Maria Goretti Hospital, Sapienza University of Rome, 04100 Latina, Italy
| | - Mariateresa Sanseviero
- Pediatrics and Neonatology Unit, Maternal-Child Department, Santa Maria Goretti Hospital, Sapienza University of Rome, 04100 Latina, Italy
| | - Flavia Ventriglia
- Pediatrics and Neonatology Unit, Maternal-Child Department, Santa Maria Goretti Hospital, Sapienza University of Rome, 04100 Latina, Italy
| | - Riccardo Lubrano
- Pediatrics and Neonatology Unit, Maternal-Child Department, Santa Maria Goretti Hospital, Sapienza University of Rome, 04100 Latina, Italy
| |
Collapse
|
6
|
Qiu S, Hadidchi R, Vichare A, Lu JY, Hou W, Henry S, Akalin E, Duong TQ. SARS-CoV-2 Infection Is Associated with an Accelerated eGFR Decline in Kidney Transplant Recipients up to Four Years Post Infection. Diagnostics (Basel) 2025; 15:1091. [PMID: 40361909 PMCID: PMC12072077 DOI: 10.3390/diagnostics15091091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2025] [Revised: 04/15/2025] [Accepted: 04/22/2025] [Indexed: 05/15/2025] Open
Abstract
Background/Objectives: Although kidney transplant recipients (KTRs) who are immune-compromised have been shown to be at high risk of adverse acute COVID-19 outcomes (i.e., mortality and critical illness), the long-term outcomes of KTRs with a history of SARS-CoV-2 infection are unknown. We aimed to compare long-term outcomes of KTRs with and without exposure to SARS-CoV-2. Methods: This study retrospectively evaluated 1815 KTRs in the Montefiore Health System from 4 January 2001 to 31 January 2024. The final cohorts consisted of KTRs who survived COVID-19 (n = 510) and matched KTRs without COVID-19 (n = 510, controls). Outcomes were defined as all-cause mortality and changes in estimated glomerular filtration rate (eGFR) and urine protein to creatinine ratio (UPCR) from 30 days up to four years post index date. Kaplan-Meier survival analysis and Cox proportional modeling were performed for mortality. Generalized estimating equations were used to analyze changes in eGFR and UPCR across time. Results: There was no significant group difference in long-term all-cause mortality (adjusted hazard ratio = 0.66, [0.43, 1.01] p = 0.057). eGFR in controls and COVID-19 patients before infection similarly decreased -0.98 units/year [-1.50, -0.46]. By contrast, eGFR declined at a significantly greater rate (-1.80 units/year [-2.45, -1.15]) in KTRs after COVID-19 compared to KTRs without COVID-19. This association was only seen among male and not female KTRs. COVID-19 status was not significantly associated with rate of change in UPCR or acute kidney rejection rate. Conclusions: SARS-CoV-2 infection was associated with an accelerated decline in eGFR up to four years post infection, suggesting potential long-term implications for graft health. These findings underscore the importance of vigilant monitoring and management of kidney function post SARS-CoV-2 infection in this vulnerable population.
Collapse
Affiliation(s)
- Shawn Qiu
- Department of Radiology, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, NY 10461, USA; (S.Q.); (R.H.); (A.V.); (J.Y.L.); (W.H.); (S.H.)
| | - Roham Hadidchi
- Department of Radiology, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, NY 10461, USA; (S.Q.); (R.H.); (A.V.); (J.Y.L.); (W.H.); (S.H.)
| | - Aditi Vichare
- Department of Radiology, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, NY 10461, USA; (S.Q.); (R.H.); (A.V.); (J.Y.L.); (W.H.); (S.H.)
| | - Justin Y. Lu
- Department of Radiology, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, NY 10461, USA; (S.Q.); (R.H.); (A.V.); (J.Y.L.); (W.H.); (S.H.)
| | - Wei Hou
- Department of Radiology, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, NY 10461, USA; (S.Q.); (R.H.); (A.V.); (J.Y.L.); (W.H.); (S.H.)
| | - Sonya Henry
- Department of Radiology, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, NY 10461, USA; (S.Q.); (R.H.); (A.V.); (J.Y.L.); (W.H.); (S.H.)
| | - Enver Akalin
- Department of Medicine (Nephrology), Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, NY 10461, USA;
| | - Tim Q. Duong
- Department of Radiology, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, NY 10461, USA; (S.Q.); (R.H.); (A.V.); (J.Y.L.); (W.H.); (S.H.)
| |
Collapse
|
7
|
Lloreta-Trull J, Marin-Corral J, Juanpere N, Pascual-Guardia S, Gimeno J, Naranjo D, Segalés L, Hernández S, Simón M, Serrano L, Casado B, Lloveras B, Gea J. Muscle disease in severe COVID-19 patients: a microangiopathic myopathy. Ultrastruct Pathol 2025; 49:296-305. [PMID: 40257175 DOI: 10.1080/01913123.2025.2488809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 03/07/2025] [Accepted: 04/01/2025] [Indexed: 04/22/2025]
Abstract
Patients surviving coronavirus disease of 2019 (COVID-19) often complain of skeletal muscle weakness that may be very limiting and long-lasting. There are almost no studies on the skeletal muscle of these patients, and electron microscopic data are scarce. We assessed the ultrastructural changes in the quadriceps of eight patients with COVID-19 and found a combination of features different from those reported in corticosteroid myopathy and acute relaxant-steroid myopathy. The most remarkable and constant changes involved the endothelial cells and consisted of massive amounts of pinocytotic vesicles, degenerative changes, platelet aggregates and, most characteristic of all, an increase in the external lamina thickness that seems to stem from reduplication due to successive bouts of endothelial cell damage and subsequent regeneration. Viral particles were not found in any of the cases. This distinct and quite common set of alterations defines the myopathy associated with infection by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). This association seems to be the result of an inflammatory process that would arise in infected cells but could damage non-infected endomysial blood vessels, thus resulting in persistent changes of the microvasculature that would be related to long-standing myopathic clinical features.
Collapse
Affiliation(s)
- Josep Lloreta-Trull
- Department of Pathology, Hospital del Mar-Parc de Salut Mar, Institut Hospital del Mar d'Investigacions Mèdiques (IMIM), Barcelona, Spain
- Department of Experimental and Health Sciences (CEXS), Universitat Pompeu Fabra, Barcelona, Spain
| | - Judith Marin-Corral
- Intensive Care Department, Hospital del Mar., Critical Pathology Research Group (GREPAC), IMIM, Barcelona, Spain
| | - Nuria Juanpere
- Department of Pathology, Hospital del Mar-Parc de Salut Mar, Institut Hospital del Mar d'Investigacions Mèdiques (IMIM), Barcelona, Spain
- Department of Experimental and Health Sciences (CEXS), Universitat Pompeu Fabra, Barcelona, Spain
| | - Sergi Pascual-Guardia
- Department of Experimental and Health Sciences (CEXS), Universitat Pompeu Fabra, Barcelona, Spain
- Department of Respiratory Medicine, Hospital del Mar-IMIM, Barcelona, Spain
| | - Javier Gimeno
- Department of Pathology, Hospital del Mar-Parc de Salut Mar, Institut Hospital del Mar d'Investigacions Mèdiques (IMIM), Barcelona, Spain
- Department of Experimental and Health Sciences (CEXS), Universitat Pompeu Fabra, Barcelona, Spain
| | - Dolores Naranjo
- Department of Pathology, Hospital del Mar-Parc de Salut Mar, Institut Hospital del Mar d'Investigacions Mèdiques (IMIM), Barcelona, Spain
| | - Laura Segalés
- Department of Pathology, Hospital del Mar-Parc de Salut Mar, Institut Hospital del Mar d'Investigacions Mèdiques (IMIM), Barcelona, Spain
- Department of Experimental and Health Sciences (CEXS), Universitat Pompeu Fabra, Barcelona, Spain
| | - Silvia Hernández
- Department of Pathology, Hospital del Mar-Parc de Salut Mar, Institut Hospital del Mar d'Investigacions Mèdiques (IMIM), Barcelona, Spain
- Department of Experimental and Health Sciences (CEXS), Universitat Pompeu Fabra, Barcelona, Spain
| | - Mercedes Simón
- Department of Pathology, Hospital del Mar-Parc de Salut Mar, Institut Hospital del Mar d'Investigacions Mèdiques (IMIM), Barcelona, Spain
| | - Laia Serrano
- Department of Pathology, Hospital del Mar-Parc de Salut Mar, Institut Hospital del Mar d'Investigacions Mèdiques (IMIM), Barcelona, Spain
| | - Beatriz Casado
- Department of Pathology, Hospital del Mar-Parc de Salut Mar, Institut Hospital del Mar d'Investigacions Mèdiques (IMIM), Barcelona, Spain
| | - Belén Lloveras
- Department of Pathology, Hospital del Mar-Parc de Salut Mar, Institut Hospital del Mar d'Investigacions Mèdiques (IMIM), Barcelona, Spain
- Department of Experimental and Health Sciences (CEXS), Universitat Pompeu Fabra, Barcelona, Spain
| | - Joaquim Gea
- Department of Experimental and Health Sciences (CEXS), Universitat Pompeu Fabra, Barcelona, Spain
- Department of Respiratory Medicine, Hospital del Mar-IMIM, Barcelona, Spain
| |
Collapse
|
8
|
Yu B, Yang M, Yu J, Wang D, Luo H, Cheng M, Zhang S, Li G, Wang L, Qian G, Zhang D, Li S, Ren Z, Kan Q. The effectiveness and safety of azvudine treatment in COVID‐19 patients with kidney disease based on a multicenter retrospective cohort study. VIEW 2025. [DOI: 10.1002/viw.20240075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 03/10/2025] [Indexed: 04/25/2025] Open
Abstract
AbstractKidney disease has been the main risk factor of poor prognosis for COVID‐19 patients. The effectiveness and safety of azvudine treatment in COVID‐19 patients with kidney disease have not been reported. Herein, we conducted a nine‐center and retrospective cohort study in China (ClinicalTrials: NCT06349655) that enrolled 32,864 hospitalized COVID‐19 patients, in which 4192 patients were pre‐existed with kidney disease. After exclusions and propensity score matching, a total of 831 kidney disease patients treated with azvudine and 831 kidney disease patients treated without any anti‐viral treatment (normal group) were selected. Based on Kaplan–Meier and Cox regression analysis, we found that azvudine administration had significantly decreased risks of all‐cause death (p < 0.0001) and composite disease progression (p = 0.012) as compared to the normal group. Multivariate Cox proportional hazards regression analysis demonstrated that the hazard ratio of all‐cause death was 0.64 (95% CI: 0.503–0.826, p < 0.001) and the hazard ratio of composite disease progression was 0.81 (95% CI: 0.658–1.004, p = 0.05). The subgroup analysis of different characteristics indicated no significant influence of single factor in both all‐cause mortality and composite disease progression. Five sensitivity analyses were employed to verify the robustness of our results. Safety analysis based on adverse event rate demonstrated an increased rate of hypertriglyceridemia after azvudine administration. In conclusion, we are the first to report the effectiveness and safety of azvudine treatment in COVID‐19 patients with kidney disease and demonstrate that azvudine could reduce the risk of all‐cause death without significant adverse events based on a large‐scale, multicenter, retrospective cohort study.
Collapse
Affiliation(s)
- Bo Yu
- Department of Pharmacy The First Affiliated Hospital of Zhengzhou University Zhengzhou China
| | - Mengzhao Yang
- Department of Infectious Diseases State Key Laboratory of Antiviral Drugs Pingyuan Laboratory The First Affiliated Hospital of Zhengzhou University Zhengzhou China
| | - Jia Yu
- Department of Infectious Diseases State Key Laboratory of Antiviral Drugs Pingyuan Laboratory The First Affiliated Hospital of Zhengzhou University Zhengzhou China
| | - Daming Wang
- Department of Infectious Diseases State Key Laboratory of Antiviral Drugs Pingyuan Laboratory The First Affiliated Hospital of Zhengzhou University Zhengzhou China
| | - Hong Luo
- Guangshan County People's Hospital, Guangshan County Xinyang China
| | - Ming Cheng
- Department of Medical Information The First Affiliated Hospital of Zhengzhou University Zhengzhou China
| | - Shixi Zhang
- Department of Infectious Diseases Shangqiu Municipal Hospital Shangqiu China
| | - Guotao Li
- Department of Infectious Diseases Luoyang Central Hospital Affiliated of Zhengzhou University Luoyang China
| | - Ling Wang
- Department of Clinical Laboratory Henan Provincial Chest Hospital Affiliated of Zhengzhou University Zhengzhou China
| | - Guowu Qian
- Department of Gastrointestinal Surgery Nanyang Central Hospital Nanyang China
| | - Donghua Zhang
- Department of Infectious Diseases Anyang City Fifth People's Hospital Anyang China
| | - Silin Li
- Department of Respiratory and Critical Care Medicine Fengqiu County People's Hospital Xinxiang China
| | - Zhigang Ren
- Department of Infectious Diseases State Key Laboratory of Antiviral Drugs Pingyuan Laboratory The First Affiliated Hospital of Zhengzhou University Zhengzhou China
| | - Quancheng Kan
- Henan Key Laboratory of Precision Clinical Pharmacy Zhengzhou University Zhengzhou China
| |
Collapse
|
9
|
Dey P, Mal N, Sinha R, Kumar A, Kumar P, Saroj U, Chaudhuri PK, Sinha MBK, Guria R, Mishra B, Prasad ML, Kumar D, Kumar S, Prasad MK. Neutrophil gelatinase-associated lipocalin as a predictive biomarker of acute kidney injury in COVID-19 infection: A systematic review and meta-analysis. J Family Med Prim Care 2025; 14:1194-1206. [PMID: 40396107 PMCID: PMC12088576 DOI: 10.4103/jfmpc.jfmpc_1513_24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 10/08/2024] [Accepted: 10/13/2024] [Indexed: 05/22/2025] Open
Abstract
Background Coronavirus 2019 (COVID-19) is an infectious disease caused by a novel coronavirus, SARS-CoV-2. Acute kidney injury (AKI) affects approximately 20-40% of patients with COVID-19 admitted to the intensive care unit (ICU), and it is a complication that has been linked to increased morbidity and mortality. Neutrophil gelatinase-associated lipocalin (NGAL) is a biomarker of acute kidney injury. Methods Articles were searched from databases such as PubMed, Google Scholar, and Cochrane Library till June 2023. Pooled sensitivity, specificity, area under the curve (AUC), diagnostic odds ratio (DOR), and summery receiver operating curve (SROC) were calculated with 95% confidence interval. I2 statistics and Chi-square test were used to look for the heterogeneity in between studies. Meta-regression was conducted to look for the source of heterogeneity and GRADE analysis was performed to look for the certainty of evidence. Results Altogether, eight studies were selected (4 serum/5 urine), out of which one study had both serum and urine NGAL data. The total sample size was 1,067 (349 serum/718 urine). For serum and urine NGAL, the pooled sensitivity was 0.79 (95% CI: 0.72-0.84) and 0.75 (95% CI: 0.68-0.80), pooled specificity was 0.87 (95% CI: 0.81-0.91) and 0.85 (95% CI: 0.77-0.91), DOR was 24 (95% CI: 14-43), and 17 (95% CI: 9-32) and AUC was 0.90 (95% CI: 0.87-0.92) and 0.80 (95% CI: 0.76-0.83), respectively. Conclusion Both serum and urine NGAL have favourable pooled sensitivity, specificity, DOR and AUC for the diagnosis of AKI in COVID-19 infection, however, with low certainty of evidence.
Collapse
Affiliation(s)
- Puja Dey
- Department of Medicine, Rajendra Institute of Medical Sciences, Ranchi, Jharkhand, India
| | - Nilanjan Mal
- Department of Medicine, Rajendra Institute of Medical Sciences, Ranchi, Jharkhand, India
| | - Rashmi Sinha
- Department of Medicine, Rajendra Institute of Medical Sciences, Ranchi, Jharkhand, India
| | - Amit Kumar
- Department of Laboratory Medicine, Rajendra Institute of Medical Sciences, Ranchi, Jharkhand, India
| | - Pramod Kumar
- Department of Biochemistry, Hi-Tech Medical College and Hospital, Rourkela, Odisha, India
| | - Usha Saroj
- Department of Blood Centre and Transfusion Medicine, Rajendra Institute of Medical Sciences, Ranchi, Jharkhand, India
| | - Partha Kumar Chaudhuri
- Department of Paediatrics, Rajendra Institute of Medical Sciences, Ranchi, Jharkhand, India
| | | | - Rishi Guria
- Department of Medicine, Rajendra Institute of Medical Sciences, Ranchi, Jharkhand, India
| | - Brajesh Mishra
- Department of TB and Chest, Rajendra Institute of Medical Sciences, Ranchi, Jharkhand, India
| | - Manohar Lal Prasad
- Department of Medicine, Rajendra Institute of Medical Sciences, Ranchi, Jharkhand, India
| | - Divakar Kumar
- Department of Medicine, Rajendra Institute of Medical Sciences, Ranchi, Jharkhand, India
| | - Satish Kumar
- Department of Medicine, Rajendra Institute of Medical Sciences, Ranchi, Jharkhand, India
| | - Manoj Kumar Prasad
- Department of Medicine, Rajendra Institute of Medical Sciences, Ranchi, Jharkhand, India
| |
Collapse
|
10
|
Yang Q, Zhang M, Dong Z, Deng F. A predictive model to explore risk factors for Henoch-Schönlein purpura nephritis in children: a retrospective cross-sectional study. Front Public Health 2025; 13:1507408. [PMID: 40177080 PMCID: PMC11961411 DOI: 10.3389/fpubh.2025.1507408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 02/28/2025] [Indexed: 04/05/2025] Open
Abstract
Objective The risk factors for Henoch-Schönlein purpura nephritis (HSPN) remain largely unclear, particularly in family environment and vaccination. This study aimed to develop a predictive framework to quantify the risk of HSPN by examining family environmental factors and COVID-19 vaccination outcomes in children with Henoch-Schönlein purpura (HSP) in Anhui, China. Methods This study retrospectively analyzed 362 children diagnosed with HSP at Anhui Children's Hospital between January 2020 and February 2024. A questionnaire was designed to collect information from enrolled children. For patients with incomplete medical records, parents were contacted via phone or the questionnaire was sent to them to complete the survey. After data collection, the patients were split randomly into a training group and a validation group at a 7:3 ratio, univariate and multivariate logistic regression analyses were performed to identify risk factors for nephritis, and a nomogram was constructed from these factors to provide a visual prediction of the likelihood of nephritis in HSP. The nomogram's performance was evaluated in both the training and validation groups using the area under the receiver operating characteristic (AUC) curve, calibration plots, and decision curve analysis (DCA). Results The study identified family income/month, age of onset, BMI, number of recurrences, and COVID-19 vaccination status as independent risk factors for HSPN. A nomogram was subsequently developed afterward using these factors. In the training group, the nomogram achieved an area under the curve (AUC) of 0.83 (95% CI: 0.78-0.88), while in the validation group, the AUC was 0.90 (95% CI: 0.84-0.96), demonstrating strong predictive performance. The calibration curve showed that the nomogram's predictions were well-aligned with the actual outcomes. Additionally, DCA indicated that the nomogram provided considerable clinical net benefit. Conclusion The nomogram offers accurate risk prediction for nephritis in children with HSP, helping healthcare professionals identify high-risk patients early and make informed clinical decisions.
Collapse
Affiliation(s)
- Qianwen Yang
- Department of Nephrology, Children’s Hospital of Anhui Medical University, Children’s Medical Center of Anhui Medical University, Hefei, Anhui, China
- Department of Pediatrics, Hefei Maternal and Child Health Hospital, Hefei, Anhui, China
| | - Maoyang Zhang
- Department of Nephrology, Children’s Hospital of Anhui Medical University, Children’s Medical Center of Anhui Medical University, Hefei, Anhui, China
| | - Zilong Dong
- Department of Nephrology, Children’s Hospital of Anhui Medical University, Children’s Medical Center of Anhui Medical University, Hefei, Anhui, China
| | - Fang Deng
- Department of Nephrology, Children’s Hospital of Anhui Medical University, Children’s Medical Center of Anhui Medical University, Hefei, Anhui, China
| |
Collapse
|
11
|
Shyong O, Alfakhri N, Bates SV, Carroll RW, Gallagher K, Huang L, Madhavan V, Murphy SA, Okrzesik SA, Yager PH, Yonker LM, Lok J. Multisystem Inflammatory Syndrome in Children: A Comprehensive Review Over the Past Five Years. J Intensive Care Med 2025:8850666251320558. [PMID: 40096057 DOI: 10.1177/08850666251320558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
Multisystem Inflammatory Syndrome in Children: A Comprehensive Review over the Past Five Years This review explores many facets of Multisystem Inflammatory Syndrome in Children (MIS-C) over the previous 5 years. In the time since the COVID 19 pandemic gripped our medical systems, we can now explore the data that has been collected from the previous years. The literature has allowed us to better understand the impact of COVID 19 and the post illness occurrence of a severe systemic inflammatory disease on our youngest patient populations. This paper will outline the pathophysiology of MIS-C, the treatments utilized, short and long-term patient outcomes including epidemiological factors.
Collapse
Affiliation(s)
- Olivia Shyong
- Department of Pediatrics, Pediatric Critical Care Medicine, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Nora Alfakhri
- Department of Pediatrics, Pediatric Critical Care Medicine, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Sara V Bates
- Harvard Medical School, Boston, MA, USA
- Department of Pediatrics, Newborn Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Ryan W Carroll
- Department of Pediatrics, Pediatric Critical Care Medicine, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Krista Gallagher
- Department of Pediatrics, Pediatric Critical Care Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Lena Huang
- Touro University Nevada, College of Osteopathic Medicine, Henderson, NV, USA
| | - Vandana Madhavan
- Harvard Medical School, Boston, MA, USA
- Department of Pediatrics, Pediatric Infectious Disease, Massachusetts General Hospital, Boston, MA, USA
| | - Sarah A Murphy
- Department of Pediatrics, Pediatric Critical Care Medicine, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Sylvia A Okrzesik
- Department of Pharmacy, Massachusetts General Hospital, Boston, MA, USA
| | - Phoebe H Yager
- Department of Pediatrics, Pediatric Critical Care Medicine, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Lael M Yonker
- Harvard Medical School, Boston, MA, USA
- Department of Pediatrics, Pediatric Pulmonary Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Josephine Lok
- Department of Pediatrics, Pediatric Critical Care Medicine, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| |
Collapse
|
12
|
Kolhe NV, Fluck R, Taal M. Effect of COVID-19 with or without acute kidney injury on inpatient mortality in England: a national observational study using administrative data. BMJ Open 2025; 15:e095020. [PMID: 40090681 PMCID: PMC11911683 DOI: 10.1136/bmjopen-2024-095020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Accepted: 02/06/2025] [Indexed: 03/18/2025] Open
Abstract
OBJECTIVES To evaluate hospital outcomes and their predictors during the pandemic for patients with and without COVID-19, stratified by the presence of acute kidney injury (AKI). DESIGN Retrospective observation study using the Hospital Episodes Statistics database for England. PARTICIPANTS 2 385 337 unique hospital admissions of adult patients from March 2020 to March 2021 in England. MAIN OUTCOME MEASURES COVID-19 cases were identified by the International Classification of Diseases, Tenth Revision, Clinical Modification (ICD-10-CM) code of U07.1. Patients with suspected COVID-19 (U07.2 code) and patients with end-stage kidney disease on chronic dialysis (N18.6 and Z99.2) were excluded. AKI cases were identified by the ICD10 code. Patients were categorised into four groups based on COVID-19 and AKI diagnoses: Group 1-neither; Group 2-COVID-19 only; Group 3-AKI only; Group 4-both. A multivariable logistic regression model was created with in-hospital mortality as the outcome, including diagnostic groups, demographics, admission methods, comorbidity severity, deprivation index and intensive therapy unit (ITU) admission. RESULTS Among 2 385 337 admissions involving 663 628 patients, 856 544 had AKI (N17 codes) and 1 528 793 did not. Among patients without AKI, there were 1,008,774 admissions among 133,988 individuals without COVID-19 (Group 1) and 520,019 admissions among 256,037 individuals with COVID-19 (Group 2). Among patients with AKI, there were 630,342 admissions among 218,270 individuals without COVID-19 (Group 3) and 226,202 admissions among 55,333 individuals with COVID-19 (Group 4). Patients in group 4 were older (75.4 ± 13.8 years) and had greater length of stay (17.1 ± 17 days) than all other groups. They also had and had a greater proportion of males, ethnic minorities and comorbidities than other groups. Mortality was highest in Group 4 (28.7%) and lowest in Group 1 (1.1%). The increased risk of death persisted after controlling for multiple baseline factors (OR for death vs Group 1: Group 4-22.28, Group 2-9.67, Group 3-6.44). ITU admission was least required in Group 1 (1.2%) and most in Group 4 (10.9%), with mortality at 4.8% versus 47.8%, respectively. CONCLUSIONS Patients with COVID-19 and AKI have a high risk of mortality and should be recognised early and provided with optimal support. Planning for future pandemics should ensure adequate critical care and acute dialysis capacity. TRIAL REGISTRATION NUMBER NCT04579562.
Collapse
Affiliation(s)
- Nitin V Kolhe
- Department of Renal Medicine, University Hospitals of Derby and Burton NHS Foundation Trust, Derby, UK
- Division of Medical Sciences and Graduate Entry Medicine, University of Nottingham, Derby, UK
| | - Richard Fluck
- Department of Renal Medicine, University Hospitals of Derby and Burton NHS Foundation Trust, Derby, UK
| | - Maarten Taal
- Department of Renal Medicine, University Hospitals of Derby and Burton NHS Foundation Trust, Derby, UK
- Division of Medical Sciences and Graduate Entry Medicine, University of Nottingham, Derby, UK
| |
Collapse
|
13
|
Adilović M. COVID-19 related complications. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2025; 213:259-314. [PMID: 40246346 DOI: 10.1016/bs.pmbts.2025.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/19/2025]
Abstract
The COVID-19 pandemic has significantly impacted global healthcare systems, revealed vulnerabilities and prompted a re-evaluation of medical practices. Acute complications from the virus, including cardiovascular and neurological issues, have underscored the necessity for timely medical interventions. Advances in diagnostic methods and personalized therapies have been pivotal in mitigating severe outcomes. Additionally, Long COVID has emerged as a complex challenge, affecting various body systems and leading to respiratory, cardiovascular, neurological, psychological, and musculoskeletal problems. This broad spectrum of complications highlights the importance of multidisciplinary management approaches that prioritize therapy, rehabilitation, and patient-centered care. Vulnerable populations such as paediatric patients, pregnant women, and immunocompromised individuals face unique risks and complications, necessitating continuous monitoring and tailored management strategies to reduce morbidity and mortality associated with COVID-19.
Collapse
Affiliation(s)
- Muhamed Adilović
- Department of Genetics and Bioengineering, Faculty of Engineering and Natural Sciences, International University of Sarajevo, Hrasnička cesta, Sarajevo, Bosnia and Herzegovina.
| |
Collapse
|
14
|
Edyko P, Zdunek M, Nowicka M, Kurnatowska I. Prevalence and Risk Factors for Acute Kidney Injury in COVID-19-Hospitalized Patients in Poland Across Three Pandemic Periods. J Clin Med 2025; 14:1384. [PMID: 40004913 PMCID: PMC11856720 DOI: 10.3390/jcm14041384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2024] [Revised: 12/28/2024] [Accepted: 02/17/2025] [Indexed: 02/27/2025] Open
Abstract
Background/Objectives: Acute kidney injury (AKI) is a serious and prevalent complication of COVID-19. This study examines the prevalence, risk factors, and outcomes of AKI in hospitalized COVID-19 patients. Methods: We analyzed the data of 1223 adult COVID-19 hospitalized patients from a single district hospital during three pandemic periods: 3 November 2020-31 December 2020, 17 March 2021-8 May 2021, and 4 November 2021-21 February 2022. The analysis included demographic data, comorbidities, laboratory results, chest radiographs (CT lung scans), and outcomes. Results: We found an overall AKI incidence of 29.02%. AKI patients versus non-AKI ones were significantly older (median age 76.0 vs. 71.0, p < 0.001) and had more comorbidities, especially previous renal diseases, heart failure, coronary artery disease, and hypertension; they also significantly more often used diuretics, angiotensin receptor blockers (ARBs), and angiotensin-converting enzyme inhibitors (ACE-Is). AKI patients more frequently presented with abnormal CT lung scans and had higher white blood cell counts, lower lymphocytes percentages, higher C-reactive protein (CRP) levels, and lower platelet counts. They more often required oxygen therapy, more days of hospitalization, and had higher mortality rates. Conclusions: Older age, comorbidities, the use of diuretics, and renin-angiotensin system inhibitors (RASI) are key risk factors for AKI, which is consequently linked to a more severe disease course and poorer prognosis.
Collapse
Affiliation(s)
- Paweł Edyko
- Student Scientific Society Affiliated with the Department of Internal Medicine and Transplant Nephrology, Medical University of Łódź, 90-419 Łódź, Poland; (P.E.); (M.Z.)
| | - Marta Zdunek
- Student Scientific Society Affiliated with the Department of Internal Medicine and Transplant Nephrology, Medical University of Łódź, 90-419 Łódź, Poland; (P.E.); (M.Z.)
| | - Maja Nowicka
- Department of Internal Medicine and Transplant Nephrology, Medical University of Łódź, Kopcińskiego 22, 90-419 Łódź, Poland;
| | - Ilona Kurnatowska
- Department of Internal Medicine and Transplant Nephrology, Medical University of Łódź, Kopcińskiego 22, 90-419 Łódź, Poland;
| |
Collapse
|
15
|
Pathak A, Agrawal DK. Role of Gut Microbiota in Long COVID: Impact on Immune Function and Organ System Health. ARCHIVES OF MICROBIOLOGY & IMMUNOLOGY 2025; 9:38-53. [PMID: 40051430 PMCID: PMC11883900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/09/2025]
Abstract
SARS-CoV-2 infection has led to a range of long-lasting symptoms, collectively referred to as long COVID. Current research highlights the critical role of angiotensin-converting enzyme 2 (ACE2) in regulating gut microbiota diversity, vascular function, and homeostasis within the renin-angiotensin system (RAS). ACE2 is utilized by the SARS-CoV-2 virus to enter host cells, but its downregulation following infection contributes to gut microbiota dysbiosis and RAS disruption. These imbalances have been linked to a range of long COVID symptoms, including joint pain, chest pain, chronic cough, fatigue, brain fog, anxiety, depression, myalgia, peripheral neuropathy, memory difficulties, and impaired attention. This review investigates the dysregulation caused by SARS-CoV-2 infection and the long-term effects it has on various organ systems, including the musculoskeletal, neurological, renal, respiratory, and cardiovascular systems. We explored the bidirectional interactions between the gut microbiota, immune function, and these organ systems, focusing on how microbiota dysregulation contributes to the chronic inflammation and dysfunction observed in long COVID symptoms. Understanding these interactions is key for identifying effective therapeutic strategies and interventional targets aimed at mitigating the impact of long COVID on organ health and improving patient outcomes.
Collapse
Affiliation(s)
- Angelie Pathak
- Department of Translational Research, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, California 91766 USA
| | - Devendra K Agrawal
- Department of Translational Research, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, California 91766 USA
| |
Collapse
|
16
|
Nankivell BJ, P'ng C, Tran T, Draper J, Ko D, Luu I, Basile K, Kable K, Sciberras F, Wong G, Kok J. The Effects of COVID-19 in Kidney Transplantation: Evidence From Tissue Pathology. Transplantation 2025; 109:352-361. [PMID: 39020461 DOI: 10.1097/tp.0000000000005121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/19/2024]
Abstract
BACKGROUND The biological effects of SARS-CoV-2 infection in transplanted kidneys are uncertain with little pathological information. METHODS This single-center, prospective observational study evaluated kidney transplant biopsies from recipients of deceased donors with COVID-19, current recipients contracting SARS-CoV-2 Omicron variant in 2022, against prior BK virus (BKV) infection and uninfected (without SARS-CoV-2 or BKV) samples, as respective positive and negative comparators (n = 503 samples). RESULTS We demonstrated nonvirus tubular injury in implanted tissue from infected donors and prevalent recipients with mild acute COVID-19 and acute kidney injury, excluding direct viral infection as a cause of kidney damage. COVID particles were absent in 4116 ultrastructural images of 295 renal tubules from 4 patients with acute COVID-19. No viral cytopathic effect, viral allograft nephropathy, or SARS-CoV-2 RNA was detected in acute tissues, nor in 128 sequential samples from infected donors or recipients with COVID-19. Following recipient COVID-19 (mean 16.8 ± 12.0 wk post-infection), the biopsy-prevalence of rejection was 33.0% (n = 100 biopsies) versus 13.4% for contemporaneous uninfected controls (n = 337; P < 0.001). Prior COVID-19 was an independent risk factor for incident rejection using multivariable generalized estimating equation adjusted for competing risks (odds ratio, 2.195; 95% confidence interval, 1.189-4.052; P = 0.012). Landmark and matched-pair analyses confirmed an association of SARS-CoV-2 with subsequent transplant rejection, with a similar pattern following BKV infection. CONCLUSIONS Transplantation from COVID-19+ deceased donors yielded good recipient outcomes without evidence of viral tissue transmission. Acute kidney injury during COVID-19 was mediated by archetypical tubular injury and infection correlated with an increased risk of subsequent rejection.
Collapse
Affiliation(s)
- Brian J Nankivell
- Department of Renal Medicine, Westmead Hospital, Westmead, NSW, Australia
| | - Chow P'ng
- Tissue Pathology and Diagnostic Oncology and Electron Microscopy Units, Westmead Hospital, Westmead, NSW, Australia
| | - Thomas Tran
- Tissue Pathology and Diagnostic Oncology and Electron Microscopy Units, Westmead Hospital, Westmead, NSW, Australia
| | - Jenny Draper
- Centre for Infectious Diseases & Microbiology Laboratory Services, NSW Health Pathology-Institute of Clinical Pathology and Medical Research, Westmead Hospital, Westmead, NSW, Australia
| | - Danny Ko
- Centre for Infectious Diseases & Microbiology Laboratory Services, NSW Health Pathology-Institute of Clinical Pathology and Medical Research, Westmead Hospital, Westmead, NSW, Australia
| | - Ivan Luu
- Centre for Infectious Diseases & Microbiology Laboratory Services, NSW Health Pathology-Institute of Clinical Pathology and Medical Research, Westmead Hospital, Westmead, NSW, Australia
| | - Kerri Basile
- Centre for Infectious Diseases & Microbiology Laboratory Services, NSW Health Pathology-Institute of Clinical Pathology and Medical Research, Westmead Hospital, Westmead, NSW, Australia
| | - Kathy Kable
- Department of Renal Medicine, Westmead Hospital, Westmead, NSW, Australia
| | | | - Germaine Wong
- Department of Renal Medicine, Westmead Hospital, Westmead, NSW, Australia
| | - Jen Kok
- Centre for Infectious Diseases & Microbiology Laboratory Services, NSW Health Pathology-Institute of Clinical Pathology and Medical Research, Westmead Hospital, Westmead, NSW, Australia
| |
Collapse
|
17
|
Wang S, Sun J, Hu Y, Zhang W, Qin B, Li M, Zhang N, Wang S, Zhou T, Liu M, Ma C, Deng X, Bai Y, Qu G, Liu L, Shi H, Zhou B, Li K, Yang B, Li S, Wang F, Ma J, Zhang L, Wang Y, An L, Liu W, Chang Q, Zhang R, Yin X, Yang Y, Ao Q, Ma Q, Yan S, Huang H, Song P, Zhao S, Gao L, Lu W, Xu L, Lei L, Wang K, Song Q, Zhang Z, Fang X, He Y, Zhang Q, Jia J, Zhu P. Clinical and Multiorgan Proteomics Characteristics of the Diverse Fatal Phase in Super Elderly Patients With SARS-CoV-2 Infection: A Descriptive Study. J Med Virol 2025; 97:e70207. [PMID: 39921383 DOI: 10.1002/jmv.70207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Revised: 01/14/2025] [Accepted: 01/25/2025] [Indexed: 02/10/2025]
Abstract
This study aims to identify the risk factors associated with clinical outcomes and the proteomic changes in organs related to fatal SARS-CoV-2 infection within the super-elderly population. This retrospective analysis included all elderly individuals with COVID-19 admitted to the Second Medical Center of PLA General Hospital from December 2022 to January 2023. The follow-up period ended on March 30, 2023. During this time, epidemiological, demographic, laboratory, and outcome data were analyzed descriptively. Proteomic sequencing was performed on super-elderly patients who died from COVID-19 at different stages of the disease. A total of 352 elderly COVID-19 patients, with a mean age of 89.84 ± 8.54 years, were included in this study. During a median follow-up period of 98 days, 79 patients died. Deceased patients were older and more likely to have cardiovascular and cerebrovascular diseases, with a lower prevalence of lipid-lowering therapy. The number of deaths in the acute and post-acute phases were 34 and 45, respectively. Proteomics data suggest that the immune systems of patients who died in the acute phase underwent a more rapid and severe onslaught. Patients in the post-acute phase showed higher levels of viral genome replication and a more robust immune response. However, the over-activation of the immune system led to systemic organ dysfunction. Effective management of comorbidities may improve the prognosis of COVID-19 in super-elderly patients. The continuous replication of the SARS-CoV-2 virus and its subsequent impact on the immune system are critical determinants of survival time in this demographic.
Collapse
Affiliation(s)
- Shuxia Wang
- Department of Geriatrics, The Second Medical Center & National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Beijing, China
| | - Jin Sun
- Department of Geriatrics, The Second Medical Center & National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Beijing, China
- Medical School of Chinese PLA, Beijing, China
| | - Yazhuo Hu
- Institute of Geriatrics, The Second Medical Center, Beijing Key Laboratory of Aging and Geriatrics, National Clinical Research Center for Geriatrics Diseases, Chinese PLA General Hospital, Beijing, China
| | - Wei Zhang
- Department of Integrative Therapy, The Second Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Bangguo Qin
- Medical School of Chinese PLA, Beijing, China
| | - Man Li
- Department of Geriatrics, The Second Medical Center & National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Beijing, China
| | - Nan Zhang
- Department of Geriatrics, The Second Medical Center & National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Beijing, China
- Medical School of Chinese PLA, Beijing, China
| | - Shengshu Wang
- Institute of Geriatrics, The Second Medical Center, Beijing Key Laboratory of Aging and Geriatrics, National Clinical Research Center for Geriatrics Diseases, Chinese PLA General Hospital, Beijing, China
| | - Tingyu Zhou
- National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Beijing, China
| | - Miao Liu
- Department of Anti-NBC Medicine, Graduate School of Chinese PLA General Hospital, Beijing, China
| | - Cong Ma
- Department of Health Medicine, The Second Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Xinli Deng
- Department of Clinical Laboratory, The Second Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Yongyi Bai
- Department of Cardiology, The Second Medical Center & National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Beijing, China
| | - Geping Qu
- Department of Respiratory and Critical Care Medicine, The Second Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Lin Liu
- Department of Respiratory and Critical Care Medicine, The Second Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Hui Shi
- Department of Gastroenterology, The Second Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Bo Zhou
- Department of Neurology, The Second Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Ke Li
- Department of Neurology, The Second Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Bo Yang
- Department of Hematology, The Second Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Suxia Li
- Department of Hematology, The Second Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Fan Wang
- Department of Cardiology, The Second Medical Center & National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Beijing, China
| | - Jinling Ma
- Department of Cardiology, The Second Medical Center & National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Beijing, China
| | - Lu Zhang
- Department of Cardiology, The Second Medical Center & National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Beijing, China
| | - Yajuan Wang
- Department of Respiratory and Critical Care Medicine, The Second Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Li An
- Department of Respiratory and Critical Care Medicine, The Second Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Wenhui Liu
- Department of Gastroenterology, The Second Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Qing Chang
- Department of Gastroenterology, The Second Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Ru Zhang
- Department of Gastroenterology, The Second Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Xi Yin
- Department of Neurology, The Second Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Yang Yang
- Department of Neurology, The Second Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Qiangguo Ao
- Department of Nephrology, The Second Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Qiang Ma
- Department of Nephrology, The Second Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Shuangtong Yan
- Department of Endocrinology, The Second Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Haili Huang
- Department of MedicaI Oncology, The Second Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Peng Song
- Department of MedicaI Oncology, The Second Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Shu Zhao
- Department of MedicaI Oncology, The Second Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Linggen Gao
- Department of General Surgery, The Second Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Wenning Lu
- Department of General Surgery, The Second Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Lining Xu
- Department of General Surgery, The Second Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Li Lei
- Department of Health Medicine, The Second Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Keyu Wang
- Department of Clinical Laboratory, The Second Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Qing Song
- Department of Critical Care Medicine, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Zhijian Zhang
- Department of Respiratory and Critical Care Medicine, The Second Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Xiangqun Fang
- Department of Respiratory and Critical Care Medicine, The Second Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Yao He
- Institute of Geriatrics, The Second Medical Center, Beijing Key Laboratory of Aging and Geriatrics, National Clinical Research Center for Geriatrics Diseases, Chinese PLA General Hospital, Beijing, China
| | - Qi Zhang
- The Second Medical Center, PLA General Hospital, Beijing, China
| | - Jianjun Jia
- Institute of Geriatrics, The Second Medical Center, Beijing Key Laboratory of Aging and Geriatrics, National Clinical Research Center for Geriatrics Diseases, Chinese PLA General Hospital, Beijing, China
| | - Ping Zhu
- Department of Geriatrics, The Second Medical Center & National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
18
|
Perschinka F, Mayerhöfer T, Engelbrecht T, Graf A, Zajic P, Metnitz P, Joannidis M. Impact of mechanical ventilation on severe acute kidney injury in critically ill patients with and without COVID-19 - a multicentre propensity matched analysis. Ann Intensive Care 2025; 15:17. [PMID: 39862353 PMCID: PMC11762028 DOI: 10.1186/s13613-025-01424-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 12/10/2024] [Indexed: 01/27/2025] Open
Abstract
BACKGROUND Acute kidney injury (AKI) is common in critically ill patients and is associated with increased morbidity and mortality. Its complications often require renal replacement therapy (RRT). Invasive mechanical ventilation (IMV) and infections are considered risk factors for the occurrence of AKI. The use of IMV and non-invasive ventilation (NIV) has changed over the course of the pandemic. Concomitant with this change in treatment a reduction in the incidences of AKI and RRT was observed. We aimed to investigate the impact of IMV on RRT initiation by comparing critically ill patients with and without COVID-19. Furthermore, we wanted to investigate the rates and timing of RRT as well as the outcome of patients, who were treated with RRT. RESULTS A total of 8,678 patients were included, of which 555 (12.8%) in the COVID-19 and 554 (12.8%) in the control group were treated with RRT. In the first week of ICU stay the COVID-19 patients showed a significantly lower probability for RRT initiation (day 1: p < 0.0001, day 2: p = 0.021). However, after day 7 a reversed HR was found. In mechanically ventilated patients the risk was significantly higher for the initiation of RRT over the entire stay. While in non-COVID-19 patients this was a non-significant trend, in COVID-19 patients the risk for RRT was significantly increased. The median delay between initiation of IMV and requirement of RRT was observed to be longer in COVID-19 patients (5 days [IQR: 2-11] vs. 2 days [IQR: 1-5]). The analysis restricted to patients with RRT showed a significantly higher risk for ICU death in patients requiring IMV compared to patients without IMV. CONCLUSION The analysis demonstrated that IMV as well as COVID-19 are associated with an increased risk for initiation of RRT. The association between IMV and risk of RRT initiation was given for all investigated time intervals. Additionally, COVID-19 patients showed an increased risk for RRT initiation during the entire ICU stay within patients admitted to an ICU due to respiratory disease. In COVID-19 patients treated with RRT, the risk of death was significantly higher compared to non-COVID-19 patients.
Collapse
Affiliation(s)
- Fabian Perschinka
- Division of Intensive Care and Emergency Medicine, Department of Internal Medicine, Medical University Innsbruck, Anichstrasse 35, Innsbruck, 6020, Austria
| | - Timo Mayerhöfer
- Division of Intensive Care and Emergency Medicine, Department of Internal Medicine, Medical University Innsbruck, Anichstrasse 35, Innsbruck, 6020, Austria
| | - Teresa Engelbrecht
- Institute of Medical Statistics, Center for Medical Data Science, Medical University of Vienna, Vienna, Austria
| | - Alexandra Graf
- Institute of Medical Statistics, Center for Medical Data Science, Medical University of Vienna, Vienna, Austria
| | - Paul Zajic
- Division of Anaesthesiolgy and Intensive Care 1, Department of Anaesthesiology and Intensive Care, Medical University of Graz, Graz, Austria
| | - Philipp Metnitz
- Division of Anaesthesiolgy and Intensive Care 1, Department of Anaesthesiology and Intensive Care, Medical University of Graz, Graz, Austria
| | - Michael Joannidis
- Division of Intensive Care and Emergency Medicine, Department of Internal Medicine, Medical University Innsbruck, Anichstrasse 35, Innsbruck, 6020, Austria.
| |
Collapse
|
19
|
Cheng HI, Chang KW, Wu BC, Teo MY, Hung WS, Wu HM, Huang ACC, Lin CW, Lin TY, Lin HC, Chiu CH, Lin SM. Comparison of Clinical Characteristics and Mortality Outcome in Critical COVID-19 Patients Infected with Alpha and Omicron Variants. Infect Drug Resist 2025; 18:151-160. [PMID: 39803308 PMCID: PMC11725234 DOI: 10.2147/idr.s479896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Accepted: 12/19/2024] [Indexed: 01/16/2025] Open
Abstract
Objective Early reports have indicated that the Omicron variant of coronavirus disease 2019 (COVID-19) may be associated with low mortality. However, the mortality rate of critical patients in Taiwan with COVID-19 caused by different variants has not been well described. Methods This retrospective cohort study was conducted at the Linkou Branch of Chang Gung Memorial Hospital, Taiwan, from April 2020 to September 2022. Critically ill patients who had confirmed SARS-CoV-2 infection and were on mechanical ventilation (MV) were enrolled. Demographic data, laboratory results, and treatment information were collected and analyzed. In addition, clinical outcomes for different SARS-CoV-2 variants were analyzed. Results This study included 110 critical patients with COVID-19 who required intubation and intensive care unit (ICU) admission. Among these patients, 46 (41.8%) required intensive care during Alpha predominance period and 64 (58.2%) during the Omicron predominance period. The Alpha group had a higher body mass index, had a longer ICU stay, and included more patients with acute respiratory distress syndrome, and the Omicron group included more active smokers, had more comorbidities, had worse initial laboratory data (including higher white blood cell counts, prothrombin time [PT], activated partial prothrombin time, blood urine nitrogen levels, and creatine levels), and had higher in-hospital mortality rates (40.6% vs 15.2%, p = 0.004). The independent risk factors for in-hospital mortality, were Charlson Comorbidity Index (CCI) ≥ 3 and higher PT and creatine levels. Conclusion Our study discovered that CCI ≥ 3, elevated serum creatine levels, and prolonged PT were independently associated with a high mortality rate in patients with critical COVID-19. Patients with those risk factors may require intensive monitoring during their treatment course.
Collapse
Affiliation(s)
- Hsin-I Cheng
- Department of Thoracic Medicine, Chang Gung Memorial Hospital, Linkuo, Taiwan
| | - Ko-Wei Chang
- Department of Thoracic Medicine, Chang Gung Memorial Hospital, Linkuo, Taiwan
| | - Bing-Chen Wu
- Department of Thoracic Medicine, Chang Gung Memorial Hospital, Linkuo, Taiwan
| | - Mei-Yuan Teo
- Department of Thoracic Medicine, Chang Gung Memorial Hospital, Linkuo, Taiwan
| | - Wei-Syun Hung
- Department of Thoracic Medicine, Chang Gung Memorial Hospital, Linkuo, Taiwan
| | - Hao-Ming Wu
- Department of Thoracic Medicine, Chang Gung Memorial Hospital, Linkuo, Taiwan
| | | | - Chang-Wei Lin
- Department of Thoracic Medicine, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Ting-Yu Lin
- Department of Thoracic Medicine, Chang Gung Memorial Hospital, Linkuo, Taiwan
| | - Horng-Chyuan Lin
- Department of Thoracic Medicine, Chang Gung Memorial Hospital, Linkuo, Taiwan
| | - Cheng-Hsun Chiu
- Department of Pediatrics, Chang Gung Children’s Hospital, Chang Gung University College of Medicine, Taoyuan City, Taiwan
- Molecular Infectious Disease Research Center, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Shu-Min Lin
- Department of Thoracic Medicine, Chang Gung Memorial Hospital, Linkuo, Taiwan
- Department of Respiratory Therapy, Chang Gung Memorial Hospital, Linkuo, Taiwan
- School of Medicine, National Tsing Hua University, Hsin-Chu, Taiwan
- School of Medicine, Chang Gung University, Taoyuan, Taiwan
| |
Collapse
|
20
|
Guissouma J, Ben Ali H, Allouche H, Trabelsi I, Hammami O, Yahia Y, Hatem G. Acute Kidney Injury Complicating Critical Forms of COVID-19: risk Factors and Prognostic Impact. F1000Res 2025; 13:497. [PMID: 39839732 PMCID: PMC11747297 DOI: 10.12688/f1000research.144105.2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/23/2024] [Indexed: 01/23/2025] Open
Abstract
Background Severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) mainly affects the respiratory tract, but different organs may be involved including the kidney. Data on acute kidney injury (AKI) in critical forms of coronavirus disease 2019 (COVID-19) are scarce. We aimed to assess the incidence, risk factors and prognostic impact of AKI complicating critical forms of COVID-19. Methods A retrospective descriptive case/control monocentric study conducted in a medical intensive care unit of a tertiary teaching hospital over a period of 18 months. Results We enrolled 144 patients, with a mean age of 58±13 years old and a male predominance (sex-ratio: 1.25). Forty-one (28%) developed AKI within a median of 4 days (Q1: 3, Q3: 8.5) after hospitalization. It was staged KDIGO class 3, in about half of the cases. Thirteen patients underwent renal replacement therapy and renal function improved in seven cases. Diabetes (OR: 6.07; 95% CI: (1,30-28,4); p: 0.022), nephrotoxic antibiotics (OR: 21; 95% CI: (3,2-146); p: 0.002), and shock (OR: 12.21; 95% CI: (2.87-51.85); p: 0.031,) were the three independent risk factors of AKI onset. Mortality was significantly higher in AKI group (HR:12; 95% CI: (5.81-18.18); p:0.041) but AKI didn't appear to be an independent risk factor of poor outcome. In fact, age > 53 years (p: 0.018), septic shock complicating hospital acquired infection (p: 0.003) and mechanical ventilation (p<0.001) were the three prognostic factors in multivariate analysis. Conclusions The incidence of AKI was high in this study and associated to an increased mortality. Diabetes, use of nephrotoxic antibiotics and shock contributed significantly to its occurrence. This underlines the importance of rationalizing antibiotic prescription and providing adequate management of patients with hemodynamic instability in order to prevent consequent AKI.
Collapse
Affiliation(s)
- Jihene Guissouma
- Medical intensive care unit of Bizerte University Hospital, Bizerte, 7021, Tunisia
- University of Tunis El Manar Faculty of medicine of Tunis, Tunis, 1007, Tunisia
| | - Hana Ben Ali
- Medical intensive care unit of Bizerte University Hospital, Bizerte, 7021, Tunisia
- University of Tunis El Manar Faculty of medicine of Tunis, Tunis, 1007, Tunisia
| | - Hend Allouche
- Medical intensive care unit of Bizerte University Hospital, Bizerte, 7021, Tunisia
- University of Tunis El Manar Faculty of medicine of Tunis, Tunis, 1007, Tunisia
| | - Insaf Trabelsi
- Medical intensive care unit of Bizerte University Hospital, Bizerte, 7021, Tunisia
- University of Tunis El Manar Faculty of medicine of Tunis, Tunis, 1007, Tunisia
| | - Olfa Hammami
- University of Tunis El Manar Faculty of medicine of Tunis, Tunis, 1007, Tunisia
- Pediatrics department of Bizerte University Hospital, Bizerte, 7021, Tunisia
| | - Yosra Yahia
- University of Tunis El Manar Faculty of medicine of Tunis, Tunis, 1007, Tunisia
- Emergency department of Rabta University Hospital, Tunis, 1007, Tunisia
| | - Ghadhoune Hatem
- Medical intensive care unit of Bizerte University Hospital, Bizerte, 7021, Tunisia
- University of Tunis El Manar Faculty of medicine of Tunis, Tunis, 1007, Tunisia
| |
Collapse
|
21
|
Chahal S, Raj RG, Kumar R. Risk of Type 1 Diabetes Mellitus in SARS-CoV-2 Patients. Curr Diabetes Rev 2025; 21:e240524230298. [PMID: 38798206 DOI: 10.2174/0115733998290807240522045553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 04/19/2024] [Accepted: 05/09/2024] [Indexed: 05/29/2024]
Abstract
Recent studies have found that a link between people with type 1 diabetes mellitus (T1DM) are at higher risk of morbidity as well as mortality from COVID-19 infection, indicating a need for vaccination. T1DM appears to impair innate and adaptive immunity. The overabundance of pro-inflammatory cytokines produced in COVID-19 illness that is severe and potentially fatal is known as a "cytokine storm." Numerous cohorts have revealed chronic inflammation as a key risk factor for unfavorable COVID-19 outcomes. TNF-α, interleukin (IL)-1a, IL-1, IL-2, IL-6, and other cytokines were found in higher concentrations in patients with T1DM. Even more importantly, oxidative stress contributes significantly to the severity and course of COVID- 19's significant role in the progression and severity of COVID-19 diseases. Severe glucose excursions, a defining characteristic of type 1 diabetes, are widely recognized for their potent role as mediating agents of oxidative stress via several routes, such as heightened production of advanced glycation end products (AGEs) and activation of protein kinase C (PKC). Furthermore, persistent endothelial dysfunction and hypercoagulation found in T1DM may impair microcirculation and endothelium, which could result in the development of various organ failure and acute breathing syndrome.
Collapse
Affiliation(s)
- Shweta Chahal
- Department of Pharmacy Practice, ISF College of Pharmacy, Moga, Punjab, 142001, India
| | - Rojin G Raj
- Department of Pharmacy Practice, ISF College of Pharmacy, Moga, Punjab, 142001, India
| | - Ranjeet Kumar
- Department of Pharmacy Practice, ISF College of Pharmacy, Moga, Punjab, 142001, India
| |
Collapse
|
22
|
Oliva I, Ferré C, Daniel X, Cartanyà M, Villavicencio C, Salgado M, Vidaur L, Papiol E, de Molina FG, Bodí M, Herrera M, Rodríguez A. Risk factors and outcome of acute kidney injury in critically ill patients with SARS-CoV-2 pneumonia: a multicenter study. Med Intensiva 2025; 49:15-24. [PMID: 39003118 DOI: 10.1016/j.medine.2024.06.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2024] [Revised: 05/06/2024] [Accepted: 06/07/2024] [Indexed: 07/15/2024]
Abstract
OBJECTIVE To assess incidence, risk factors and impact of acute kidney injury(AKI) within 48 h of intensive care unit(ICU) admission on ICU mortality in patients with SARS-CoV-2 pneumonia. To assess ICU mortality and risk factors for continuous renal replacement therapy (CRRT) in AKI I and II patients. DESIGN Retrospective observational study. SETTING Sixty-seven ICU from Spain, Andorra, Ireland. PATIENTS 5399 patients March 2020 to April 2022. MAIN VARIABLES OF INTEREST Demographic variables, comorbidities, laboratory data (worst values) during the first two days of ICU admission to generate a logistic regression model describing independent risk factors for AKI and ICU mortality. AKI was defined according to current international guidelines (kidney disease improving global outcomes, KDIGO). RESULTS Of 5399 patients included 1879 (34.8%) developed AKI. These patients had higher ICU mortality and AKI was independently associated with a higher ICU mortality (HR 1.32 CI 1.17-1.48; p < 0.001). Male gender, hypertension, diabetes, obesity, chronic heart failure, myocardial dysfunction, higher severity scores, and procalcitonine were independently associated with the development of AKI. In AKI I and II patients the need for CRRT was 12.6% (217/1710). In these patients, APACHE II, need for mechanical ventilation in the first 24 h after ICU admission and myocardial dysfunction were associated with risk of needing CRRT. AKI I and II patients had a high ICU mortality (38.5%), especially if CRRT were required (64.1% vs. 34,8%; p < 0.001). CONCLUSIONS Critically ill patients with SARS-CoV-2 pneumonia and AKI have a high ICU mortality. Even AKI I and II stages are associated with high risk of needing CRRT and ICU mortality.
Collapse
Affiliation(s)
- Iban Oliva
- Critical Care Department, Joan XXIII University Hospital, Tarragona, Spain.
| | - Cristina Ferré
- Critical Care Department, Joan XXIII University Hospital, Tarragona, Spain
| | - Xavier Daniel
- Critical Care Department, Joan XXIII University Hospital, Tarragona, Spain
| | - Marc Cartanyà
- Critical Care Department, Joan XXIII University Hospital, Tarragona, Spain
| | | | - Melina Salgado
- Critical Care Department, Joan XXIII University Hospital, Tarragona, Spain
| | - Loreto Vidaur
- Critical Care Department, Donostia University Hospital, San Sebastian, Spain
| | - Elisabeth Papiol
- Critical Care Department, Vall d'Hebron Hospital, Barcelona, Spain
| | | | - María Bodí
- Critical Care Department, Joan XXIII University Hospital, Tarragona, Spain
| | - Manuel Herrera
- Critical Care Department, Regional University Hospital of Malaga, Malaga, Spain
| | | |
Collapse
|
23
|
Bach ML, Laftih S, Andresen JK, Pedersen RM, Andersen TE, Madsen LW, Madsen K, Hinrichs GR, Zachar R, Svenningsen P, Lund L, Johansen IS, Hansen LF, Palarasah Y, Jensen BL. ACE2 and TMPRSS2 in human kidney tissue and urine extracellular vesicles with age, sex, and COVID-19. Pflugers Arch 2025; 477:83-98. [PMID: 39382598 PMCID: PMC11711140 DOI: 10.1007/s00424-024-03022-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 09/17/2024] [Accepted: 09/20/2024] [Indexed: 10/10/2024]
Abstract
SARS-CoV-2 virus infects cells by engaging with ACE2 requiring protease TMPRSS2. ACE2 is highly expressed in kidneys. Predictors for severe disease are high age and male sex. We hypothesized that ACE2 and TMPRSS2 proteins are more abundant (1) in males and with increasing age in kidney and (2) in urine and extracellular vesicles (EVs) from male patients with COVID-19 and (3) SARS-CoV-2 is present in urine and EVs during infection. Kidney cortex samples from patients subjected to cancer nephrectomy (male/female; < 50 years/˃75 years, n = 24; ˃80 years, n = 15) were analyzed for ACE2 and TMPRSS2 protein levels. Urine from patients hospitalized with SARS-CoV-2 infection was analyzed for ACE2 and TMPRSS2. uEVs were used for immunoblotting and SARS-CoV-2 mRNA and antigen detection. Tissue ACE2 and TMPRSS2 protein levels did not change with age. ACE2 was not more abundant in male kidneys in any age group. ACE2 protein was associated with proximal tubule apical membranes in cortex. TMPRSS2 was observed predominantly in the medulla. ACE2 was elevated significantly in uEVs and urine from patients with COVID-19 with no sex difference compared with urine from controls w/wo albuminuria. TMPRSS2 was elevated in uEVs from males compared to female. ACE2 and TMPRSS2 did not co-localize in uEVs/apical membranes. SARS-CoV-2 nucleoprotein and mRNA were not detected in urine. Higher kidney ACE2 protein abundance is unlikely to explain higher susceptibility to SARS-CoV-2 infection in males. Kidney tubular cells appear not highly susceptible to SARS-CoV-2 infection. Loss of ACE2 into urine in COVID could impact susceptibility and angiotensin metabolism.
Collapse
Affiliation(s)
- Marie Lykke Bach
- Unit of Cardiovascular and Renal Research, Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark.
| | - Sara Laftih
- Unit of Cardiovascular and Renal Research, Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Jesper K Andresen
- Unit of Cardiovascular and Renal Research, Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Rune M Pedersen
- Department of Clinical Microbiology, Odense University Hospital, and Research Unit for Clinical Microbiology, University of Southern Denmark, Odense, Denmark
| | - Thomas Emil Andersen
- Department of Clinical Microbiology, Odense University Hospital, and Research Unit for Clinical Microbiology, University of Southern Denmark, Odense, Denmark
| | - Lone W Madsen
- Department of Infectious Diseases, Odense University Hospital, and Research Unit for Infectious Diseases, University of Southern Denmark, Odense, Denmark
- Unit for Infectious Diseases, Department of Medicine, Sygehus Lillebælt, Kolding, Denmark
| | - Kirsten Madsen
- Unit of Cardiovascular and Renal Research, Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark
- Department of Pathology, Odense University Hospital, Odense, Denmark
| | - Gitte R Hinrichs
- Unit of Cardiovascular and Renal Research, Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark
- Department of Nephrology, Odense University Hospital, Odense, Denmark
| | - Rikke Zachar
- Department of Nephrology, Odense University Hospital, Odense, Denmark
| | - Per Svenningsen
- Unit of Cardiovascular and Renal Research, Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Lars Lund
- Department of Urology, Odense University Hospital, Odense, Denmark
| | - Isik S Johansen
- Department of Infectious Diseases, Odense University Hospital, and Research Unit for Infectious Diseases, University of Southern Denmark, Odense, Denmark
| | | | - Yaseelan Palarasah
- Unit of Inflammation and Cancer Research, Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Boye L Jensen
- Unit of Cardiovascular and Renal Research, Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
24
|
Alberto RPJ, Benjamin GN, Elizabeth RMJ, Alberto CDL, Eliseo PDB. Understanding COVID-19-related Acute Renal Injury in Elderly Individuals: Preexisting Systemic Inflammation before COVID-19 (SIC). Endocr Metab Immune Disord Drug Targets 2025; 25:300-309. [PMID: 38919086 DOI: 10.2174/0118715303312433240611093855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 05/14/2024] [Accepted: 05/20/2024] [Indexed: 06/27/2024]
Abstract
In this study, we examined preexisting systemic inflammation before COVID-19 (SIC), as assessed through C-reactive protein (CRP) levels, to gain insights into the origins of acute kidney injury (AKI) in adults with comorbidities affected by COVID-19. Although aging is not categorized as a disease, it is characterized by chronic inflammation, and older individuals typically exhibit higher circulating levels of inflammatory molecules, particularly CRP, compared to younger individuals. Conversely, elevated CRP concentrations in older adults have been linked with the development of comorbidities. Simultaneously, these comorbidities contribute to the production of inflammatory molecules, including CRP. Consequently, older adults with comorbidities have higher CRP concentrations than their counterparts without comorbidities or those with fewer comorbidities. Given that CRP levels are correlated with the development and severity of AKI in non-COVID-19 patients, we hypothesized that individuals with greater SIC are more likely to develop AKI during SARS-CoV-2 infection than those with less SIC.
Collapse
Affiliation(s)
- Ruiz-Pacheco Juan Alberto
- Investigador por México-CONAHCYT, Centro de Investigación Biomédica de Occidente, IMSS, Guadalajara, Jalisco, México
| | - Gomez-Navarro Benjamin
- Servicio de Nefrología y Trasplantes, Hospital Country 2000, Guadalajara, Jalisco, México
| | | | - Castillo-Díaz Luis Alberto
- Departamento de Medicina y Ciencias de la Salud, Facultad Interdiciplinaria de Ciencias Biológicas y de la Salud, Universidad de Sonora, Hermosillo, México
| | - Portilla-de Buen Eliseo
- División de Investigación Quirúrgica, Centro de Investigación Biomédica de Occidente, IMSS, Guadalajara, Jalisco, México
| |
Collapse
|
25
|
Sun C, Zhao X, Wang X, Yu Y, Shi H, Tang J, Sun S, Zhu S. Astragalus Polysaccharide Mitigates Rhabdomyolysis-Induced Acute Kidney Injury via Inhibition of M1 Macrophage Polarization and the cGAS-STING Pathway. J Inflamm Res 2024; 17:11505-11527. [PMID: 39735897 PMCID: PMC11675321 DOI: 10.2147/jir.s494819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 12/05/2024] [Indexed: 12/31/2024] Open
Abstract
Purpose This study aimed to examine the impact of APS on acute kidney injury induced by rhabdomyolysis (RIAKI), exploring its association with macrophage M1 polarization and elucidating the underlying mechanisms. Methods C57BL/6J mice were randomly assigned to one of three groups: a normal control group, a RIAKI model group, and an APS treatment group. Techniques such as flow cytometry and immunofluorescence were employed to demonstrate that APS can inhibit the transition of renal macrophages to the M1 phenotype in RIAKI. Furthermore, the raw264.7 macrophage cell line was chosen and induced into the M1 phenotype to further examine the impact of APS on this model and elucidate the underlying mechanism. Results Administration of APS led to a significant decrease in UREA levels by 25.2% and CREA levels by 60.9% within the model group. Also, APS exhibited an inhibitory effect on the infiltration of M1 macrophages and the cGAS-STING pathway in kidneys within the RIAKI, subsequently leading to decreased serum concentrations of IL-1β, IL-6 and TNF-α by 44.5%, 12.9%, and 10.3%, respectively, consistent with the results of in vitro experiments. Furthermore, APS exhibited an anti-apoptotic effect on MPC5 cells when co-cultured with M1 macrophages. Conclusion Astragalus polysaccharide (APS) potentially mitigated rhabdomyolysis-induced renal damage by impeding the M1 polarization of macrophages. This inherent mechanism might involve the suppression of the cGAS-STING pathway activation within macrophages. Furthermore, APS could endow protective effects on podocytes through the inhibition of apoptosis.
Collapse
Affiliation(s)
- Chuanchuan Sun
- Department of Traditional Chinese Medicine, The First Affiliated Hospital of Jinan University, Guangzhou, People’s Republic of China
- Department of Nephrology, The First Affiliated Hospital of Jinan University, Guangzhou, People’s Republic of China
| | - Xinhai Zhao
- Department of Traditional Chinese Medicine, The First Affiliated Hospital of Jinan University, Guangzhou, People’s Republic of China
| | - Xianghong Wang
- Department of Endocrinology and Metabolism, Zhuhai People’s Hospital (Zhuhai Clinical Medical College of Jinan University), Zhuhai, People’s Republic of China
| | - Yeye Yu
- Department of Traditional Chinese Medicine, The First Affiliated Hospital of Jinan University, Guangzhou, People’s Republic of China
| | - Heng Shi
- Department of Gastroenterology, The Central Hospital of Shaoyang, Shaoyang, People’s Republic of China
| | - Jun Tang
- The Fifth Affiliated Hospital of Zunyi Medical University, Zhuhai(Zhuhai Sixth People’s Hospital), Zhuhai, People’s Republic of China
| | - Shengyun Sun
- Department of Traditional Chinese Medicine, The First Affiliated Hospital of Jinan University, Guangzhou, People’s Republic of China
| | - Shiping Zhu
- Department of Traditional Chinese Medicine, The First Affiliated Hospital of Jinan University, Guangzhou, People’s Republic of China
| |
Collapse
|
26
|
Vardhan H, Saigal M, Shyama S, Krishna A. Clinical Features and Outcomes of Patients With COVID-19 Infection and Acute Kidney Injury Requiring Hemodialysis in an Intensive Care Unit: A Retrospective Study From a Tertiary Care Center in Eastern India. Cureus 2024; 16:e75363. [PMID: 39781159 PMCID: PMC11707804 DOI: 10.7759/cureus.75363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/09/2024] [Indexed: 01/12/2025] Open
Abstract
BACKGROUND During the COVID-19 pandemic, it has been observed that acute kidney injury (AKI) especially requiring intervention support of hemodialysis has notably increased mortality rates among COVID-19-positive critically ill patients; however, comprehensive data regarding this from India, especially the eastern territory, remains sparse. This study aims to outline the demographic, clinical, and biochemical characteristics, along with the outcomes, of these patients. METHODS A retrospective study was performed at the All India Institute of Medical Sciences (AIIMS), Patna, from March 1, 2020, to March 31, 2021. Included were patients diagnosed with COVID-19 and AKI necessitating hemodialysis during their intensive care unit (ICU) stay. These patients tested positive for COVID-19 and met the Kidney Disease: Improving Global Outcomes (KDIGO) criteria for AKI stages 1-3, requiring ICU admission and hemodialysis. Medical history, clinical features, laboratory results, comorbidities, and demographic data were collected and analyzed. Patients were tracked from admission to discharge or death. Gaussian-distributed values were compared using the unpaired t-test or Pearson's test, while non-Gaussian continuous variables were analyzed using the Mann-Whitney test or Spearman's test. The study employed the Kolmogorov-Smirnov test to assess Gaussian distribution, while categorical data were compared using the Chi-square test. RESULTS Among 773 patients with positive COVID-19 tests who were admitted to the ICU, 236 patients developed AKI, and among them, 139 patients required hemodialysis. The total mortality rate was 167 (70.7%) among people who had AKI and 102 (77%) in patients with AKI who required hemodialysis. AKI was also a risk factor associated with higher mortality rates in older patients (>45 years) (n=150 (73.2%)), those needing invasive ventilation (n=163 (88.1%)), and patients with elevated total leucocyte count (TLC) (n=130 (79.3%)), lactate dehydrogenase (LDH) (n=159 (72.9%)), interleukin-6 (IL-6) (n=153 (72.2%)), and serum ferritin (n=51 (73.7%)) and hypoalbuminemia (n=152 (73.1%)). CONCLUSION AKI requiring hemodialysis significantly increases mortality risk in COVID-19 patients. Other risk factors for mortality with AKI in COVID-19-positive patients include age, elevated leucocyte count, invasive ventilation, and deranged inflammatory markers.
Collapse
Affiliation(s)
| | - Megha Saigal
- Nephrology, All India Institute of Medical Sciences, Patna, Patna, IND
| | - Shyama Shyama
- Internal Medicine, All India Institute of Medical Sciences, Patna, Patna, IND
| | - Amresh Krishna
- Nephrology, All India Institute of Medical Sciences, Patna, Patna, IND
| |
Collapse
|
27
|
Muthya A, Ekinci EI, Lecamwasam A. What is the spectrum of kidney pathology associated with COVID-19? Intern Med J 2024; 54:1935-1943. [PMID: 39485035 PMCID: PMC11610687 DOI: 10.1111/imj.16540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 08/20/2024] [Indexed: 11/03/2024]
Abstract
Kidney involvement occurs in almost one third of patients hospitalised with coronavirus disease 2019 (COVID-19) and is associated with increased disease severity. This review aims to outline the spectrum of kidney pathology involved in COVID-19. Literature was reviewed systematically on the databases Medline OVID and Scopus in search of case reports, case series, cohort studies and autopsy studies of patients with COVID-19 who underwent kidney biopsies. Studies were published between August 2020 and November 2021. Fourteen studies consisting of 159 patients were included in this review. Acute tubular necrosis is the most common pathology followed by collapsing glomerulopathy, occurring in 40.1% and 28.9% of patients respectively. Of the 46 patients with collapsing glomerulopathy, 44 were of African descent with high-risk apolipoprotein L1 genotypes. Less common glomerular diseases include membranous nephropathy, secondary focal segmental glomerulosclerosis, minimal change disease and primary focal segmental glomerulosclerosis occurring in 5%, 4.4%, 3.1% and 2.5% of patients respectively. Glomerulonephritis occurred in a minority of patients. Direct viral infection has not been found as a definitive aetiology. Acute kidney injury occurs frequently in hospitalised COVID-19 patients and is associated with increased morbidity and mortality. The mechanisms underpinning acute kidney injury are multifactorial. Acute tubular necrosis is the most common. Collapsing glomerulopathy is the most common glomerular injury and is strongly linked to apolipoprotein L1 genotypes. Improved understanding of COVID-19-related kidney pathologies can guide treatment to improve patient outcomes and reduce progression of chronic kidney disease. The longitudinal impact of COVID-19-related kidney disease requires further research.
Collapse
Affiliation(s)
- Ankita Muthya
- Department of MedicineUniversity of MelbourneMelbourneVictoriaAustralia
| | - Elif I. Ekinci
- Department of MedicineUniversity of MelbourneMelbourneVictoriaAustralia
- Department of EndocrinologyAustin HealthMelbourneVictoriaAustralia
| | - Ashani Lecamwasam
- Department of MedicineUniversity of MelbourneMelbourneVictoriaAustralia
- Department of EndocrinologyAustin HealthMelbourneVictoriaAustralia
- Department of NephrologyNorthern HealthMelbourneVictoriaAustralia
| |
Collapse
|
28
|
He Z, Liu JJ, Ma SL. Serum chemokine IL-8 acts as a biomarker for identifying COVID-19-associated persistent severe acute kidney injury. Ren Fail 2024; 46:2311316. [PMID: 38305217 PMCID: PMC10840601 DOI: 10.1080/0886022x.2024.2311316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 01/23/2024] [Indexed: 02/03/2024] Open
Abstract
OBJECTIVES Persistent severe acute kidney injury (PS-AKI) is associated with poor clinical outcomes. Our study attempted to evaluate the diagnostic value of chemokines for early-stage PS-AKI prediction. METHODS According to the KDIGO criteria, 115 COVID-19 patients diagnosed with stage 2/3 AKI were recruited from the intensive care unit between December 2022 and February 2023. Primary clinical outcomes included detecting PS-AKI in the first week (≥ KDIGO stage 2 ≥ 72 h). Cytometric Bead Array was used to detect patient plasma levels (interleukin-8 (IL-8), C-C chemokine ligand 5 (CCL5), chemokine (C-X-C Motif) ligand 9 (CXCL9), and interferon-inducible protein 10 (IP-10)) of chemokines within 24 h of enrollment. RESULTS Of the 115 COVID-19 patients with stage 2/3 AKI, 27 were diagnosed with PS-AKI. Among the four measured chemokines, only the IL-8 level was significantly elevated in the PS-AKI group than in the Non-PS-AKI group. IL-8 was more effective as a biomarker while predicting PS-AKI with an area under the curve of 0.769 (0.675-0.863). This was superior to other biomarkers related to AKI, including serum creatinine. Moreover, plasma IL-8 levels of >32.2 pg/ml on admission could predict PS-AKI risk (sensitivity = 92.6%, specificity = 51.1%). Additionally, the IL-8 level was associated with total protein and IL-6 levels. CONCLUSION Plasma IL-8 is a promising marker for the early identification of PS-AKI among COVID-19 patients. These findings should be validated in further studies with a larger sample size.
Collapse
Affiliation(s)
- Zhi He
- Center of Clinical Laboratory Medicine, Zhongda Hospital, Southeast University, Nanjing, China
| | - Jing-jing Liu
- Center of Clinical Laboratory Medicine, Zhongda Hospital, Southeast University, Nanjing, China
| | - Shao-lei Ma
- Department of Emergency and Critical Care Medicine, Zhongda Hospital, Southeast University, Nanjing, China
| |
Collapse
|
29
|
Nyarko JA, Dogbe PM, Ativi LAE, Wutsika J, Agyenim EB, Awere-Duodu A, Botaeng AT, Ntim NAA. Pathological Sequelae of SARS-CoV-2: A Review for Clinicians. THE YALE JOURNAL OF BIOLOGY AND MEDICINE 2024; 97:431-445. [PMID: 39703609 PMCID: PMC11650917 DOI: 10.59249/dqjh2274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/21/2024]
Abstract
The Coronavirus Disease 2019 (COVID-19) pandemic, driven by the novel coronavirus and its variants, has caused over 518 million infections and 6.25 million deaths globally, leading to a significant health crisis. Beyond its primary respiratory impact, Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2) has been implicated in various extra-pulmonary complications. Research studies reveal that the virus affects multiple organs, including the kidneys, liver, pancreas, and central nervous system (CNS), largely due to the widespread expression of Angiotensin Converting Enzyme-2 (ACE-2) receptors. Clinical evidence shows that the virus can induce diabetes by disrupting pancreatic and liver functions as well as cause acute kidney injury. Additionally, neurological complications, including cognitive impairments and neuroinflammation, have been observed in a significant number of COVID-19 patients. This review discusses the mechanisms linking SARS-CoV-2 to acute kidney injury, Type 1 and Type 2 Diabetes Mellitus (T1DM and T2DM), emphasizing its effects on pancreatic beta cells, insulin resistance, and the regulation of gluconeogenesis. We also explore how SARS-CoV-2 induces neurological complications, detailing the intricate pathways of neuro-invasion and the potential to trigger conditions such as Alzheimer's disease (AD). By elucidating the metabolic and neurological manifestations of COVID-19 and the underlying pathogenic mechanisms, this review underscores the imperative for continued research and the development of effective therapeutic interventions to mitigate the long-term and short-term impacts of SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Joseph Asuam Nyarko
- National Influenza Centre, Noguchi Memorial Institute
for Medical Research, Accra, Ghana
| | - Patience Mawuena Dogbe
- Department of Environmental Science, Kwame Nkrumah
University of Science and Technology, Kumasi, Ghana
| | | | - Jennifer Wutsika
- National Influenza Centre, Noguchi Memorial Institute
for Medical Research, Accra, Ghana
| | | | - Aaron Awere-Duodu
- Department of Medical Microbiology, University of Ghana
Medical School, Accra, Ghana
| | - Anthony Twumasi Botaeng
- Department of Environmental Science, Kwame Nkrumah
University of Science and Technology, Kumasi, Ghana
| | - Nana Afia Asante Ntim
- National Influenza Centre, Noguchi Memorial Institute
for Medical Research, Accra, Ghana
| |
Collapse
|
30
|
Mayamba Nlandu Y, Tannor EK, Bamikefa TA, Rissassi Makulo JR. Kidney damage associated with COVID-19: from the acute to the chronic phase. Ren Fail 2024; 46:2316885. [PMID: 38561236 PMCID: PMC10986440 DOI: 10.1080/0886022x.2024.2316885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 02/05/2024] [Accepted: 02/05/2024] [Indexed: 04/04/2024] Open
Abstract
Severe acute respiratory syndrome coronavirus-2 (SARS-COV-2) infection is well established as a systemic disease including kidney damage. The entry point into the renal cell remains the angiotensin-converting enzyme 2 (ACE-2) receptor and the spectrum of renal lesions is broad, with a clear predominance of structural and functional tubular lesions. The most common form of glomerular injury is collapsing glomerulopathy (CG), which is strongly associated with apolipoprotein L1(APOL-1) risk variants. These acute lesions, which are secondary to the direct or indirect effects of SARS-CoV-2, can progress to chronicity and are specific to long COVID-19 in the absence of any other cause. Residual inflammation associated with SARS-CoV-2 infection, in addition to acute kidney injury (AKI) as a transitional state with or without severe histological lesions, may be responsible for greater kidney function decline in mild-to-moderate COVID-19. This review discusses the evidence for renal histological markers of chronicity in COVID-19 patients and triggers of low-grade inflammation that may explain the decline in kidney function in the post-COVID-19 period.
Collapse
Affiliation(s)
- Yannick Mayamba Nlandu
- Nephrology Unit, Kinshasa University Hospital, Kinshasa, Democratic Republic of the Congo
| | - Elliot Koranteng Tannor
- Department of Medicine, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
- Directorate of Medicine, Komfo Anokye Teaching Hospital, Kumasi, Ghana
| | | | | |
Collapse
|
31
|
Yazılıtaş F, Çakıcı EK, Güngör T, Karakaya D, Çelikkaya E, Şen ZS, Gümüşer R, Tanır NG, Bülbül M. Retrospective evaluation of acute kidney injury in paediatric COVID-19 patients: a tertiary referral hospital experience. J Nephrol 2024; 37:2541-2550. [PMID: 39031240 PMCID: PMC11663816 DOI: 10.1007/s40620-024-01986-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 01/01/2023] [Indexed: 07/22/2024]
Abstract
BACKGROUND Coronavirus disease 2019 (COVID-19) has been recognised as a risk factor for acute kidney injury (AKI). Our aim was to investigate the risk factors contributing to hospitalised and outpatient paediatric COVID-19-associated AKI. METHODS A retrospective observational study was conducted on patients aged 1 month to 18 years with diagnosed COVID-19-associated AKI applied to a tertiary paediatric referral hospital between March 1, 2020 and March 1, 2022. RESULTS A total of 6683 patients were evaluated and 486 patients were included in the study. Acute kidney injury was observed in 3.7% of outpatients and 23.9% of hospitalised patients. Multivariate logistic regression analysis showed that, on admission, a history of contact with a COVID-19 positive person (p < 0.001), age below 12 months (p = 0.004), presence of comorbidities (p < 0.001), abdominal pain (p = 0.008), anorexia (p = 0.003), dyspnoea (p = 0.005), higher lactate dehydrogenase values (p = 0.004), neutrophilia (p < 0.001), higher neutrophil-to-lymphocyte ratio (NLR) (p = 0.003), higher white blood cell counts (p = 0.006), elevated C-reactive protein (CRP) levels (p = 0.002), anaemia (p = 0.015), hypoalbuminaemia (p < 0.001), hyperglycaemia (p = 0.006), and presence of proteinuria (p = 0.003) were independent predictors of AKI. Higher rates of hospitalisation (p < 0.001) and admission to the paediatric intensive care unit (PICU) (p < 0.001), longer length of hospitalisation (p < 0.001), and greater need for mechanical ventilation (p < 0.001) were associated with AKI. CONCLUSIONS This study reveals that not only hospitalised children, but also paediatric patients are at risk for AKI. The presence of comorbidities, abdominal pain, anorexia, dyspnoea, anaemia, inflammation, hypoalbuminaemia, proteinuria and history of contact with a COVID-19 positive person were the main risk factors for AKI. COVID-19-associated AKI was associated with worse outcomes.
Collapse
Affiliation(s)
- Fatma Yazılıtaş
- Dr Sami Ulus Kadin Dogum Cocuk Sagligi ve Hastaliklari Egitim ve Arastirma. Hastanesi, Ankara, Turkey.
| | - Evrim Kargın Çakıcı
- Dr Sami Ulus Kadin Dogum Cocuk Sagligi ve Hastaliklari Egitim ve Arastirma. Hastanesi, Ankara, Turkey
| | - Tülin Güngör
- Dr Sami Ulus Kadin Dogum Cocuk Sagligi ve Hastaliklari Egitim ve Arastirma. Hastanesi, Ankara, Turkey
| | - Deniz Karakaya
- Dr Sami Ulus Kadin Dogum Cocuk Sagligi ve Hastaliklari Egitim ve Arastirma. Hastanesi, Ankara, Turkey
| | - Evra Çelikkaya
- Dr Sami Ulus Kadin Dogum Cocuk Sagligi ve Hastaliklari Egitim ve Arastirma. Hastanesi, Ankara, Turkey
| | - Zeynep Savaş Şen
- Dr Sami Ulus Kadin Dogum Cocuk Sagligi ve Hastaliklari Egitim ve Arastirma. Hastanesi, Ankara, Turkey
| | - Rüveyda Gümüşer
- Dr Sami Ulus Kadin Dogum Cocuk Sagligi ve Hastaliklari Egitim ve Arastirma. Hastanesi, Ankara, Turkey
| | - Naciye Gönül Tanır
- Dr Sami Ulus Kadin Dogum Cocuk Sagligi ve Hastaliklari Egitim ve Arastirma. Hastanesi, Ankara, Turkey
| | - Mehmet Bülbül
- Dr Sami Ulus Kadin Dogum Cocuk Sagligi ve Hastaliklari Egitim ve Arastirma. Hastanesi, Ankara, Turkey
| |
Collapse
|
32
|
Zhang C, Gerzanich V, Cruz-Cosme R, Zhang J, Tsymbalyuk O, Tosun C, Sallapalli BT, Liu D, Keledjian K, Papadimitriou JC, Drachenberg CB, Nasr M, Zhang Y, Tang Q, Simard JM, Zhao RY. SARS-CoV-2 ORF3a induces COVID-19-associated kidney injury through HMGB1-mediated cytokine production. mBio 2024; 15:e0230824. [PMID: 39345136 PMCID: PMC11559048 DOI: 10.1128/mbio.02308-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 08/12/2024] [Indexed: 10/01/2024] Open
Abstract
The primary challenge posed by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection is COVID-19-related mortality, often exacerbated by additional medical complications, such as COVID-19-associated kidney injuries (CAKIs). Up to half of COVID-19 patients experience kidney complications, with those facing acute respiratory failure and kidney injury having the worst overall prognosis. Despite the significant impact of CAKI on COVID-19-related mortality and its enduring effects in long COVID, the underlying causes and molecular mechanisms of CAKI remain elusive. In this study, we identified a functional relationship between the expression of the SARS-CoV-2 ORF3a protein and inflammation-driven apoptotic death of renal tubular epithelial cells in patients with CAKI. We demonstrate in vitro that ORF3a independently induces renal cell-specific apoptotic cell death, as evidenced by the elevation of kidney injury molecule-1 (KIM-1) and the activation of NF-kB-mediated proinflammatory cytokine (TNFα and IL-6) production. By examining kidney tissues of SARS-CoV-2-infected K18-ACE2 transgenic mice, we observed a similar correlation between ORF3a-induced cytopathic changes and kidney injury. This correlation was further validated through reconstitution of the ORF3a effects via direct adenoviral injection into mouse kidneys. Through medicinal analysis, we identified a natural compound, glycyrrhizin (GL4419), which not only blocks viral replication in renal cells, but also mitigates ORF3a-induced renal cell death by inhibiting activation of a high mobility group box 1 (HMGB1) protein, leading to a reduction of KIM-1. Moreover, ORF3a interacts with HMGB1. Overproduction or downregulation of hmgb1 expression results in correlative changes in renal cellular KIM-1 response and respective cytokine production, implicating a crucial role of HMGB1 in ORF3a-inflicted kidney injuries. Our data suggest a direct functional link between ORF3a and kidney injury, highlighting ORF3a as a unique therapeutic target contributing to CAKI. IMPORTANCE The major challenge of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection during the pandemic is COVID-19-related mortality, which has tragically claimed millions of lives. COVID-19-associated morbidity and mortality are often exacerbated by pre-existing medical conditions, such as chronic kidney diseases (CKDs), or the development of acute kidney injury (AKI) due to COVID-19, collectively known as COVID-19-associated kidney injuries (CAKIs). Patients who experience acute respiratory failure with CAKI have the poorest clinical outcomes, including increased mortality. Despite these alarming clinical findings, there is a critical gap in our understanding of the underlying causes of CAKI. Our study establishes a direct correlation between the expression of the SARS-CoV-2 viral ORF3a protein and kidney injury induced by ORF3a linking to CAKI. This functional relationship was initially observed in our clinical studies of COVID-19 patients with AKI and was further validated through animal and in vitro cellular studies, either by expressing ORF3a alone or in the context of viral infection. By elucidating this functional relationship and its underlying mechanistic pathways, our research deepens the understanding of COVID-19-associated kidney diseases and presents potential therapeutic avenues to address the healthcare challenges faced by individuals with underlying conditions.
Collapse
Affiliation(s)
- Chenyu Zhang
- Department of Pathology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Volodymyr Gerzanich
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Ruth Cruz-Cosme
- Department of Microbiology, Howard University College of Medicine, Washington, DC, USA
| | - Jiantao Zhang
- Department of Pathology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Orest Tsymbalyuk
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Cigdem Tosun
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | | | - Dongxiao Liu
- Department of Microbiology, Howard University College of Medicine, Washington, DC, USA
| | - Kaspar Keledjian
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - John C. Papadimitriou
- Department of Pathology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Cinthia B. Drachenberg
- Department of Pathology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Mohamed Nasr
- Division of AIDS, NIAID, NIH, Drug Development and Clinical Sciences Branch, Bethesda, Maryland, USA
| | - Yanjin Zhang
- Department of Veterinary Medicine, University of Maryland, College Park, Maryland, USA
| | - Qiyi Tang
- Department of Microbiology, Howard University College of Medicine, Washington, DC, USA
| | - J. Marc Simard
- Department of Pathology, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Research and Development Service, VA Maryland Health Care System, Baltimore, Maryland, USA
| | - Richard Y. Zhao
- Department of Pathology, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Research and Development Service, VA Maryland Health Care System, Baltimore, Maryland, USA
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Institute of Global Health, University of Maryland School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
33
|
Azim T, Khan AH, Sadiq F, Sulaiman SAS, Khan A, Ain Q. Impact of COVID-19 on nephropathy in diabetes mellitus type-II patients: a systematic literature review and meta-analysis. BMC Nephrol 2024; 25:399. [PMID: 39506723 PMCID: PMC11542412 DOI: 10.1186/s12882-024-03821-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 10/14/2024] [Indexed: 11/08/2024] Open
Abstract
BACKGROUND Recent reports have revealed that nephropathy leading to kidney injury (KI) is a prevalent complication of COVID-19 and is linked to high mortality and morbidity in diabetes mellitus type II (DM-T-II) patients. This systematic literature review and meta-analysis aimed to critically analyze existing studies and evidence on the impact of COVID-19 on nephropathy and kidney injury in diabetes mellitus type II (DM-T-II) patients. METHOD A systematic search was conducted in the Web of Science (WoS), PubMed and Cochrane databases for relevant studies published between March 2020 and July 2023. To ensure the integrity of the systematic literature review and meta-analysis, observational studies that specifically reported post-COVID-19 kidney injury in DM-T2 patients were included, whereas we did not include articles in the press, meta-analyses, case reports, case series, Diabetes Type-I articles or non-English papers. The primary outcome was kidney injury in patients with type II diabetes after contracting COVID-19. The protocol for this study was published on PROSPERO (registration number CRD42023413887). RESULTS Initially, 6,339 articles were included in the search, from which only 6 observational studies were selected by following the 2020 PRISMA statement. The quality of the evidence was assessed by a tool provided by the National Institutes of Health (observational studies). The total number of participants included in the studies was 14,723. Our systematic literature review and meta-analysis provide compelling evidence that kidney injury is a prevalent complication of COVID-19 infection in the type II diabetes population, with a pooled odds ratio of 2.27 (95% CI: 2.05-2.51; p < 0.00001), often necessitating hospitalization and hemodialysis in severe cases. CONCLUSION Covid-19 is associated with a two-fold increase in nephropathy and acute kidney injury in diabetes mellitus type 2 patients compared to non-diabetic patients. This implies that kidney injury is more likely to occur in diabetes mellitus type 2 patients post Covid infection.
Collapse
Affiliation(s)
- Tabinda Azim
- School of Pharmaceutical Sciences, University Sains Malaysia, Penang, 11800, Malaysia.
- Iqra University, H-9 Campus, Islamabad, 44000, Pakistan.
| | - Amer Hayat Khan
- School of Pharmaceutical Sciences, University Sains Malaysia, Penang, 11800, Malaysia.
| | - Fouzia Sadiq
- Directorate of Research, Shifa Tameer-e-Millat University, Islamabad, Pakistan
| | | | - Amjad Khan
- Faculty of Pharmacy, Quaid-e-Azam University, Islamabad, Pakistan
| | - Quratul Ain
- Directorate of Research, Shifa Tameer-e-Millat University, Islamabad, Pakistan
| |
Collapse
|
34
|
Wang G, Yang L, Xu X, Guo W, Sun L, Wang Y, Cheng W, Ye N, Kong L, Zhao X, Cheng H. SARS-CoV-2 Protein Deposition Enhances Renal Complement Activation and Aggravates Kidney Injury in Membranous Nephropathy After COVID-19. Kidney Int Rep 2024; 9:3145-3155. [PMID: 39534192 PMCID: PMC11551107 DOI: 10.1016/j.ekir.2024.08.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 07/22/2024] [Accepted: 08/05/2024] [Indexed: 11/16/2024] Open
Abstract
Introduction COVID-19 has been reported to be associated with the occurrence and recurrence of membranous nephropathy (MN). The clinicopathological characteristics and complement system activation of MN after COVID-19 are unclear. Methods A total of 38 patients with biopsy-proven MN who developed new-onset proteinuria after COVID-19 were enrolled in this study. One hundred patients with primary MN diagnosed before the COVID-19 pandemic were the control. Renal immunohistochemical staining for SARS-CoV-2 nucleocapsid protein was performed in 38 patients with MN after COVID-19. Serum membrane attack complex (MAC) was detected by enzyme-linked immunosorbent assay. Glomerular staining for the complement proteins in different pathways were detected by immunohistochemistry. Results Thirteen of 38 patients had positive staining for SARS-CoV-2 nucleocapsid protein. Compared with the control patients, the clinical manifestations were more severe in patients after COVID-19. Patients with positive SARS-CoV-2 staining had a higher proportion of nephrotic syndrome, lower level of serum albumin, and greater severity of renal interstitial fibrosis than those of patients with negative SARS-CoV-2 staining. Serum MAC level and renal MAC staining intensity of MN after COVID-19 were significantly higher than those of the control patients. MAC expression in MN patients with positive SARS-CoV-2 staining was stronger than that in both control patients and MN after COVID-19 with negative SARS-CoV-2 staining. The expression trend of factor H was consistent with that of MAC. Conclusion Excessive activation of the complement system aggravated symptoms in MN after COVID-19. Therapeutic strategy targeting the complement system may need to be considered.
Collapse
Affiliation(s)
- Guoqin Wang
- Division of Nephrology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Lei Yang
- Division of Nephrology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Xiaoyi Xu
- Division of Nephrology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Weiyi Guo
- Division of Nephrology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Lijun Sun
- Division of Nephrology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Yanyan Wang
- Division of Nephrology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Wenrong Cheng
- Division of Nephrology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Nan Ye
- Division of Nephrology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Lingqiang Kong
- Division of Nephrology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Xiaoyi Zhao
- Division of Nephrology, Affiliated Hospital of Chifeng University, Neimenggu, China
| | - Hong Cheng
- Division of Nephrology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
35
|
Cadamuro M, Lasagni A, Radu CM, Calistri A, Pilan M, Valle C, Bonaffini PA, Vitiello A, Toffanin S, Venturin C, Friòn-Herrera Y, Sironi S, Alessio MG, Previtali G, Seghezzi M, Gianatti A, Strazzabosco M, Strain AJ, Campello E, Spiezia L, Palù G, Frigo AC, Tosoni A, Nebuloni M, Parolin C, Sonzogni A, Simioni P, Fabris L. Procoagulant phenotype of virus-infected pericytes is associated with portal thrombosis and intrapulmonary vascular dilations in fatal COVID-19. J Hepatol 2024; 81:872-885. [PMID: 38908437 DOI: 10.1016/j.jhep.2024.06.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 05/22/2024] [Accepted: 06/05/2024] [Indexed: 06/24/2024]
Abstract
BACKGROUND & AIMS The underlying mechanisms and clinical impact of portal microthrombosis in severe COVID-19 are unknown. Intrapulmonary vascular dilation (IPVD)-related hypoxia has been described in severe liver diseases. We hypothesised that portal microthrombosis is associated with IPVD and fatal respiratory failure in COVID-19. METHODS Ninety-three patients who died from COVID-19 were analysed for portal microvascular damage (histology), IPVD (histology and chest-computed tomography, CT), and hypoxemia (arterial blood gas). Seventeen patients who died from COVID-19-unrelated pneumonia served as controls. Vascular lesions and microthrombi were phenotyped for endothelial (vWF) and pericyte (αSMA/PDGFR-β) markers, tissue factor (TF), viral spike protein and nucleoprotein (SP, NP), fibrinogen, and platelets (CD41a). Viral particles in vascular cells were assessed by transmission electron microscopy. Cultured pericytes were infected with SARS-CoV-2 to measure TF expression and tubulisation of human pulmonary microvascular endothelial cells was assessed upon vWF treatment. RESULTS IPVD was present in 16/66 patients with COVID-19, with available liver and lung histology, and was associated with younger age (62 vs. 78 years-old), longer illness (25 vs. 14 days), worsening hypoxemia (PaO2/FiO2 from 209 to 89), and an increased requirement for ventilatory support (63% vs. 22%) compared to COVID-19/Non-IPVD. IPVD, absent in controls, was confirmed by chest CT. COVID-19/IPVD liver histology showed portal microthrombosis in >82.5% of portal areas, with a thicker wall of αSMA/PDGFR-β+/SP+/NP+ pericytes compared with COVID-19/Non-IPVD. Thrombosed portal venules correlated with αSMA+ area, whereas infected SP+/NP+ pericytes expressed TF. SARS-CoV-2 viral particles were observed in portal pericytes. In vitro SARS-CoV-2 infection of pericytes upregulated TF and induced endothelial cells to overexpress vWF, which expanded human pulmonary microvascular endothelial cell tubules. CONCLUSIONS SARS-CoV-2 infection of liver pericytes elicits a local procoagulant response associated with extensive portal microthrombosis, IPVD and worsening respiratory failure in fatal COVID-19. IMPACT AND IMPLICATIONS Vascular involvement of the liver represents a serious complication of COVID-19 infection that must be considered in the work-up of patients with long-lasting and progressively worsening respiratory failure, as it may associate with the development of intrapulmonary vascular dilations. This clinical picture is associated with a procoagulant phenotype of portal venule pericytes, which is induced by SARS-CoV-2 infection of pericytes. Both observations provide a model that may apply, at least in part, to other vascular disorders of the liver, featuring obliterative portal venopathy, similarly characterised at the clinical level by development of hypoxemia and at the histological level by phlebosclerosis and reduced calibre of the portal vein branches in the absence of cirrhosis. Moreover, our findings shed light on an overlooked player in the pathophysiology of thrombosis, i.e. pericytes, which may present a novel therapeutic target.
Collapse
Affiliation(s)
- Massimiliano Cadamuro
- Department of Medicine DIMED, University of Padua, Padua, Italy; Clinical Medicine 1 and Thrombotic and Haemorrhagic Disease Unit, and Haemophilia Center, Padova University Hospital, Padua, Italy; School of Medicine and Surgery, University of Milan-Bicocca, Milan, Italy
| | - Alberto Lasagni
- Department of Medicine DIMED, University of Padua, Padua, Italy; Clinical Medicine 1 and Thrombotic and Haemorrhagic Disease Unit, and Haemophilia Center, Padova University Hospital, Padua, Italy
| | - Claudia Maria Radu
- Department of Women's & Children's Health (SDB), University of Padua, Italy
| | - Arianna Calistri
- Department of Molecular Medicine (DMM), University of Padua, Padua, Italy
| | - Matteo Pilan
- Department of Medicine DIMED, University of Padua, Padua, Italy; Clinical Medicine 1 and Thrombotic and Haemorrhagic Disease Unit, and Haemophilia Center, Padova University Hospital, Padua, Italy
| | - Clarissa Valle
- Department of Diagnostic Radiology, ASST Papa Giovanni XXIII Hospital, Bergamo, Italy
| | | | - Adriana Vitiello
- Department of Molecular Medicine (DMM), University of Padua, Padua, Italy
| | - Serena Toffanin
- Department of Medicine DIMED, University of Padua, Padua, Italy; Clinical Medicine 1 and Thrombotic and Haemorrhagic Disease Unit, and Haemophilia Center, Padova University Hospital, Padua, Italy
| | - Camilla Venturin
- Department of Medicine DIMED, University of Padua, Padua, Italy; Clinical Medicine 1 and Thrombotic and Haemorrhagic Disease Unit, and Haemophilia Center, Padova University Hospital, Padua, Italy
| | - Yahima Friòn-Herrera
- Department of Medicine DIMED, University of Padua, Padua, Italy; Clinical Medicine 1 and Thrombotic and Haemorrhagic Disease Unit, and Haemophilia Center, Padova University Hospital, Padua, Italy
| | - Sandro Sironi
- Department of Diagnostic Radiology, ASST Papa Giovanni XXIII Hospital, Bergamo, Italy
| | - Maria Grazia Alessio
- Clinical Chemistry Laboratory, ASST Papa Giovanni XXIII Hospital, Bergamo, Italy
| | - Giulia Previtali
- Clinical Chemistry Laboratory, ASST Papa Giovanni XXIII Hospital, Bergamo, Italy
| | - Michela Seghezzi
- Clinical Chemistry Laboratory, ASST Papa Giovanni XXIII Hospital, Bergamo, Italy
| | - Andrea Gianatti
- Department of Pathology, ASST Papa Giovanni XXIII Hospital, Bergamo, Italy
| | - Mario Strazzabosco
- Department of Internal Medicine, Digestive Disease Section, Liver Center, Yale University, New Haven, CT, US
| | | | - Elena Campello
- Department of Medicine DIMED, University of Padua, Padua, Italy; Clinical Medicine 1 and Thrombotic and Haemorrhagic Disease Unit, and Haemophilia Center, Padova University Hospital, Padua, Italy
| | - Luca Spiezia
- Department of Medicine DIMED, University of Padua, Padua, Italy; Clinical Medicine 1 and Thrombotic and Haemorrhagic Disease Unit, and Haemophilia Center, Padova University Hospital, Padua, Italy
| | - Giorgio Palù
- Department of Molecular Medicine (DMM), University of Padua, Padua, Italy
| | - Anna Chiara Frigo
- Department of Cardiac, Thoracic, and Vascular Sciences and Public Health (DCTV), University of Padua, Italy
| | - Antonella Tosoni
- Pathology Unit, L. Sacco Hospital, ASST Fatebenefratelli Sacco, Milan, Italy
| | - Manuela Nebuloni
- Pathology Unit, L. Sacco Hospital, ASST Fatebenefratelli Sacco, Milan, Italy; Pathology Unit, Department of Biomedical and Clinical Sciences, University of Milan, Milan, Italy
| | - Cristina Parolin
- Department of Molecular Medicine (DMM), University of Padua, Padua, Italy
| | | | - Paolo Simioni
- Department of Medicine DIMED, University of Padua, Padua, Italy; Clinical Medicine 1 and Thrombotic and Haemorrhagic Disease Unit, and Haemophilia Center, Padova University Hospital, Padua, Italy.
| | - Luca Fabris
- Department of Medicine DIMED, University of Padua, Padua, Italy; Clinical Medicine 1 and Thrombotic and Haemorrhagic Disease Unit, and Haemophilia Center, Padova University Hospital, Padua, Italy; Department of Internal Medicine, Digestive Disease Section, Liver Center, Yale University, New Haven, CT, US.
| |
Collapse
|
36
|
Hooper JE, Sanchez H, Litovsky S, Lu ZA, Gabrielson EW, Padera RF, Steffensen T, Solomon IH, Gilbert A, Threlkeld KJ, Rapkiewicz AV, Harper H, Kapp ME, Schwerdt MK, Mount S, Wang Y, Lu R, Williamson AK. A Large Postmortem Database of COVID-19 Patients Can Inform Disease Research and Public Policy Decision Making. Arch Pathol Lab Med 2024; 148:e386-e393. [PMID: 38452801 DOI: 10.5858/arpa.2023-0380-oa] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/21/2023] [Indexed: 03/09/2024]
Abstract
CONTEXT.— Autopsies performed on COVID-19 patients have provided critical information about SARS-CoV-2's tropism, mechanisms of tissue injury, and spectrum of disease. OBJECTIVE.— To provide an updated database of postmortem disease in COVID-19 patients, assess relationships among clinical and pathologic variables, evaluate the accuracy of death certification, and correlate disease variables to causes of death. DESIGN.— The 272 postmortem examinations reported in this paper were submitted by 14 pathologists from 9 medical or forensic institutions across the United States. The study spans the eras of the 3 principal COVID-19 strains and incorporates surveyed demographic, clinical, and postmortem data from decedents infected with SARS-CoV-2, including primary and contributing causes of death. It is the largest database of its kind to date. RESULTS.— Demographics of the decedents reported here correspond well to national statistics. Primary causes of death as determined by autopsy and official death certificates were significantly correlated. When specifically cited disease conditions found at autopsy were correlated with COVID-19 versus non-COVID-19 deaths, only lung findings characteristic of SARS-CoV-2 infection or the absence of lung findings were significantly associated. CONCLUSIONS.— Changes in hospitalization and disease likely stem from longer lifespans after COVID-19 diagnosis and alteration in treatment approaches. Although Omicron variants preferentially replicate in the upper airways, autopsied patients who died of COVID-19 in that time period showed the same lung damage as earlier decedents. Most importantly, findings suggest that there are still unelucidated risk factors for death from COVID-19 including possibly genetic susceptibility.
Collapse
Affiliation(s)
- Jody E Hooper
- From the Department of Pathology (Hooper) and the Quantitative Sciences Unit (R Lu, Wang), Stanford Medicine, Stanford, California
| | - Harry Sanchez
- the Department of Pathology, Yale School of Medicine, New Haven, Connecticut (Sanchez)
| | - Silvio Litovsky
- the Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama (Litovsky)
| | - Zhen Arthur Lu
- the Department of Family Medicine, Owensboro Family Medicine Residency Clinic, Owensboro, Kentucky (ZA Lu)
| | - Edward W Gabrielson
- the Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland (Gabrielson)
| | - Robert F Padera
- the Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts (Padera, Solomon)
| | - Thora Steffensen
- the Department of Pathology, Tampa General Hospital, Tampa, Florida (Steffensen)
| | - Isaac H Solomon
- the Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts (Padera, Solomon)
| | - Andrea Gilbert
- the Department of Pathology and Laboratory Medicine, University of Texas Health San Antonio, San Antonio, Texas (Gilbert)
| | - Kirsten J Threlkeld
- the Department of Pathology and Laboratory Medicine, The University of Vermont Medical Center, Burlington, Vermont (Threlkeld, Mount)
| | - Amy V Rapkiewicz
- the Office of the Medical Examiner, Hauppauge, New York (Rapkiewicz)
| | - Holly Harper
- the Department of Pathology, University Hospitals Cleveland Medical Center, Cleveland, Ohio (Harper)
| | - Meghan E Kapp
- the Department of Pathology, University Hospitals and Case Western Reserve University, Cleveland, Ohio (Kapp)
| | - Mary K Schwerdt
- the Office of Chief Medical Examiner of the City of New York, New York, New York (Schwerdt)
| | - Sharon Mount
- the Department of Pathology and Laboratory Medicine, The University of Vermont Medical Center, Burlington, Vermont (Threlkeld, Mount)
| | - Yiwen Wang
- From the Department of Pathology (Hooper) and the Quantitative Sciences Unit (R Lu, Wang), Stanford Medicine, Stanford, California
| | - Rong Lu
- From the Department of Pathology (Hooper) and the Quantitative Sciences Unit (R Lu, Wang), Stanford Medicine, Stanford, California
| | - Alex K Williamson
- the Department of Pathology & Laboratory Medicine, Northwell Health, New Hyde Park, New York (Williamson)
| |
Collapse
|
37
|
Yu P, Jin X, Huang W, Wang J, Zhang S, Ren L, Zhang H, Shi S. Characterization of immortalized human podocytes infected with lentivirus as an in vitro model of viral infection-associated podocytopathy. AMERICAN JOURNAL OF CLINICAL AND EXPERIMENTAL IMMUNOLOGY 2024; 13:204-214. [PMID: 39583339 PMCID: PMC11578807 DOI: 10.62347/bbcx1142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 09/25/2024] [Indexed: 11/26/2024]
Abstract
A large number of studies have shown the association of kidney disease with viral infections in the body. Viral infections cause kidney injury in two manners, the systemic inflammation (cytokine storm) and the direct infection of kidney cells. Concerning direct viral infection of podocytes, the mechanism underlying virus-induced podocyte injury remains largely unknown and requires effective experimental models to facilitate its study. Here, we performed molecular characterization of immortalized human podocyte cell line (HPC) infected with lentivirus by RNA-seq. Bioinformatics analysis revealed a strong innate immune response in the cells, including interferon production and signaling. Meanwhile, activations of ferroptosis pathway and TNF-alpha signaling were also found, consistent with an impaired viability of the cells. Lentiviral infection also upregulated expression of APOL1 as observed in patients with HIV associated nephropathy (HIVAN) and diabetic nephropathy (DN). Interestingly, when the lentiviral infected cells were treated with Adriamycin (ADR), the ADR-associated signaling pathways were not interfered and remained activated as that in the cells treated with ADR only, suggesting that the virus and ADR have distinct mechanisms in damaging podocytes. Thus, the lentivirus-infected HPC cells represent a useful in vitro model of viral infection-associated podocytopathy.
Collapse
Affiliation(s)
- Peng Yu
- National Clinical Research Center for Kidney Diseases, Jinling Hospital, Medical School of Nanjing University Nanjing 210002, Jiangsu, China
| | - Xi Jin
- National Clinical Research Center for Kidney Diseases, Jinling Hospital, Medical School of Nanjing University Nanjing 210002, Jiangsu, China
| | - Weijun Huang
- National Clinical Research Center for Kidney Diseases, Jinling Hospital, Medical School of Nanjing University Nanjing 210002, Jiangsu, China
| | - Jingjing Wang
- National Clinical Research Center for Kidney Diseases, Jinling Hospital, Medical School of Nanjing University Nanjing 210002, Jiangsu, China
| | - Sipang Zhang
- National Clinical Research Center for Kidney Diseases, Jinling Hospital, Medical School of Nanjing University Nanjing 210002, Jiangsu, China
| | - Lu Ren
- National Clinical Research Center for Kidney Diseases, Jinling Hospital, Medical School of Nanjing University Nanjing 210002, Jiangsu, China
| | - Haitao Zhang
- National Clinical Research Center for Kidney Diseases, Jinling Hospital, Medical School of Nanjing University Nanjing 210002, Jiangsu, China
| | - Shaolin Shi
- National Clinical Research Center for Kidney Diseases, Jinling Hospital, Medical School of Nanjing University Nanjing 210002, Jiangsu, China
| |
Collapse
|
38
|
Wendt R, Macholz M, Kalbitz S, Herrmann N, Herbst V, Hammes T, Kai M, Ankersmit HJ, Beige J, Lübbert C, Graf A, Scherberich J. Serum uromodulin associates with kidney function and outcome in a cohort of hospitalised COVID-19 patients. Sci Rep 2024; 14:25420. [PMID: 39455668 PMCID: PMC11512065 DOI: 10.1038/s41598-024-76372-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 10/14/2024] [Indexed: 10/28/2024] Open
Abstract
This study investigates the prevalence and evaluates the prognostic implications of acute kidney injury (AKI) in COVID-19 patients, with a novel emphasis on the evaluation of serum uromodulin (sUmod) as a potential kidney-specific biomarker. A cohort of hospitalised COVID-19 patients (n = 378) was examined for AKI using standard criteria. In addition to traditional urinary biomarkers, sUmod levels were analysed. Univariable and multivariable regression models were employed to evaluate the association of sUmod and AKI and in-hospital mortality. Levels of sUmod were significantly lower in patients with CKD (91.8 ± 60.7 ng/ml) compared to patients with normal kidney function (204.7 ± 91.7 ng/ml; p < 0.001). 151 patients (40.0%) presented with AKI at the time of hospital admission or developed an AKI during hospitalization. 116 patients (76.8%) had an AKI already at the time of hospital admission. COVID-19 patients with AKI had significantly lower levels of sUmod compared to patients without AKI during hospitalisation (124.8 ± 79.5 ng/ml) vs 214.6 ± 92.3 ng/ml; p < 0.001). The in-hospital mortality rate in this cohort of COVID-19 patients was 15.3%. Patients with AKI had a higher probability for in-hospital death (OR 5.6, CI 1.76 to 17.881, p = 0.004). Patients who died during hospital stay, had significantly lower sUmod levels (129.14 ± 89.56 ng/ml) compared to patients surviving hospitalisation (187.71 ± 96,64 ng/ml; p < 0.001). AKI is frequently associated with COVID-19 in hospitalized patients. Serum uromodulin may emerge as a promising biomarker for AKI in COVID-19 patients. Further research is warranted to explore its clinical application and refine risk stratification in this patient population.
Collapse
Affiliation(s)
- Ralph Wendt
- Department of Nephrology, St. Georg Hospital, Delitzscher Str. 141, 04129, Leipzig, Germany.
| | - Martin Macholz
- Department of Infectious Diseases and Tropical Medicine, St. Georg Hospital, Delitzscher Str. 141, 04129, Leipzig, Germany
| | - Sven Kalbitz
- Department of Infectious Diseases and Tropical Medicine, St. Georg Hospital, Delitzscher Str. 141, 04129, Leipzig, Germany
| | - Nadja Herrmann
- Department of Finance and Controlling, St. Georg Hospital, Delitzscher Str. 141, 04129, Leipzig, Germany
| | - Victor Herbst
- Institute for Experimental Immunology, Affiliated to EUROIMMUN Medizinische Labordiagnostika AG, Seekamp 31, 23560, Lübeck, Germany
| | - Tabea Hammes
- Institute for Experimental Immunology, Affiliated to EUROIMMUN Medizinische Labordiagnostika AG, Seekamp 31, 23560, Lübeck, Germany
| | - Marco Kai
- Institute for Experimental Immunology, Affiliated to EUROIMMUN Medizinische Labordiagnostika AG, Seekamp 31, 23560, Lübeck, Germany
| | - Hendrik Jan Ankersmit
- Clinic of Thoracic Surgery, Medical University of Vienna, Spitalgasse 23, 1090, Vienna, Austria
| | - Joachim Beige
- Kuratorium for Dialysis and Transplantation (KfH) Renal Unit, Hospital St. Georg, Delitzscher Str. 141, 04129, Leipzig, Germany
- Martin-Luther University Halle/Wittenberg, Halle, Germany
| | - Christoph Lübbert
- Department of Infectious Diseases and Tropical Medicine, St. Georg Hospital, Delitzscher Str. 141, 04129, Leipzig, Germany
- Division of Infectious Diseases and Tropical Medicine, Leipzig University Hospital, Liebigstr. 20, 04103, Leipzig, Germany
| | - Alexandra Graf
- Institute of Medical Statistics, Center for Medical Data Science, Medical University of Vienna, Spitalgasse 23, 1090, Vienna, Austria
| | - Jürgen Scherberich
- Department of Nephrology and Clinical Immunology, Klinikum München, 81545, Munich, Germany
| |
Collapse
|
39
|
Avdonin PP, Blinova MS, Serkova AA, Komleva LA, Avdonin PV. Immunity and Coagulation in COVID-19. Int J Mol Sci 2024; 25:11267. [PMID: 39457048 PMCID: PMC11508857 DOI: 10.3390/ijms252011267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 09/23/2024] [Accepted: 10/15/2024] [Indexed: 10/28/2024] Open
Abstract
Discovered in late 2019, the SARS-CoV-2 coronavirus has caused the largest pandemic of the 21st century, claiming more than seven million lives. In most cases, the COVID-19 disease caused by the SARS-CoV-2 virus is relatively mild and affects only the upper respiratory tract; it most often manifests itself with fever, chills, cough, and sore throat, but also has less-common mild symptoms. In most cases, patients do not require hospitalization, and fully recover. However, in some cases, infection with the SARS-CoV-2 virus leads to the development of a severe form of COVID-19, which is characterized by the development of life-threatening complications affecting not only the lungs, but also other organs and systems. In particular, various forms of thrombotic complications are common among patients with a severe form of COVID-19. The mechanisms for the development of thrombotic complications in COVID-19 remain unclear. Accumulated data indicate that the pathogenesis of severe COVID-19 is based on disruptions in the functioning of various innate immune systems. The key role in the primary response to a viral infection is assigned to two systems. These are the pattern recognition receptors, primarily members of the toll-like receptor (TLR) family, and the complement system. Both systems are the first to engage in the fight against the virus and launch a whole range of mechanisms aimed at its rapid elimination. Normally, their joint activity leads to the destruction of the pathogen and recovery. However, disruptions in the functioning of these innate immune systems in COVID-19 can cause the development of an excessive inflammatory response that is dangerous for the body. In turn, excessive inflammation entails activation of and damage to the vascular endothelium, as well as the development of the hypercoagulable state observed in patients seriously ill with COVID-19. Activation of the endothelium and hypercoagulation lead to the development of thrombosis and, as a result, damage to organs and tissues. Immune-mediated thrombotic complications are termed "immunothrombosis". In this review, we discuss in detail the features of immunothrombosis associated with SARS-CoV-2 infection and its potential underlying mechanisms.
Collapse
Affiliation(s)
| | | | | | | | - Pavel V. Avdonin
- Koltzov Institute of Developmental Biology RAS, ul. Vavilova, 26, 119334 Moscow, Russia; (P.P.A.)
| |
Collapse
|
40
|
Kurosaki Y, Matsumoto T, Uematsu T, Kawakami F, Kawashima R, Tamaki S, Imai M, Ichikawa T, Ishii N, Kitasato H, Hanaki H, Kubo M. SARS-CoV-2 infection causes a decline in renal megalin expression and affects vitamin D metabolism in the kidney of K18-hACE2 mice. Sci Rep 2024; 14:24313. [PMID: 39414885 PMCID: PMC11484755 DOI: 10.1038/s41598-024-75338-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 10/04/2024] [Indexed: 10/18/2024] Open
Abstract
Patients with coronavirus disease 2019 (COVID-19) often experience acute kidney injury, linked to disease severity or mortality, along with renal tubular dysfunction and megalin loss in proximal tubules. Megalin plays a crucial role in kidney vitamin D metabolism. However, the impact of megalin loss on vitamin D metabolism during COVID-19 is unclear. This study investigated whether severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection reduces megalin expression in proximal tubules and its subsequent effect on vitamin D metabolism in mice expressing human angiotensin converting enzyme 2 (K18-hACE2 mice). Histological and immunohistochemical staining analyses revealed glomerular and capillary congestion, and elevated renal neutrophil gelatinase-associated lipocalin levels, indicative of acute kidney injury in K18-hACE2 mice. In SARS-CoV-2-infected mice, immunohistochemical staining revealed suppressed megalin protein levels. Decreased vitamin D receptor (VDR) localization in the nucleus and increased mRNA expression of VDR, CYP27B1, and CYP24A1 were observed by quantitative PCR in SARS-CoV-2-infected mice. Serum vitamin D levels remained similar in infected and vehicle-treated mice, but an increase in tumor necrosis factor-alpha and a decrease in IL-4 mRNA expression were observed in the kidneys of the SARS-CoV-2 group. These findings suggest that megalin loss in SARS-CoV-2 infection may impact the local role of vitamin D in kidney immunomodulation, even when blood vitamin D levels remain unchanged.
Collapse
Affiliation(s)
- Yoshifumi Kurosaki
- Department of Medical Laboratory Sciences, School of Allied Health Sciences, Kitasato University, 1-15-1 Kitasato, Sagamihara, 252-0373, Japan
- Regenerative Medicine and Cell Design Research Facility, School of Allied Health Sciences, Kitasato University, 1-15-1 Kitasato, Sagamihara, 252-0373, Japan
| | - Toshihide Matsumoto
- Regenerative Medicine and Cell Design Research Facility, School of Allied Health Sciences, Kitasato University, 1-15-1 Kitasato, Sagamihara, 252-0373, Japan
- Department of Pathology, School of Allied Health Sciences, Kitasato University, 1-15-1 Kitasato, Sagamihara, 252-0373, Japan
| | - Takayuki Uematsu
- Biomedical Laboratory, Division of Biomedical Research, Kitasato University Medical Center, 6-100 Arai, Kitamoto, 364-8501, Japan
| | - Fumitaka Kawakami
- Regenerative Medicine and Cell Design Research Facility, School of Allied Health Sciences, Kitasato University, 1-15-1 Kitasato, Sagamihara, 252-0373, Japan
- Department of Health Administration, School of Allied Health Sciences, Kitasato University, 1-15-1 Kitasato, Sagamihara, 252-0373, Japan
| | - Rei Kawashima
- Regenerative Medicine and Cell Design Research Facility, School of Allied Health Sciences, Kitasato University, 1-15-1 Kitasato, Sagamihara, 252-0373, Japan
- Department of Biochemistry, School of Allied Health Sciences, Kitasato University, 1-15-1 Kitasato, Sagamihara, 252-0373, Japan
| | - Shun Tamaki
- Regenerative Medicine and Cell Design Research Facility, School of Allied Health Sciences, Kitasato University, 1-15-1 Kitasato, Sagamihara, 252-0373, Japan
- Department of Biochemistry, School of Allied Health Sciences, Kitasato University, 1-15-1 Kitasato, Sagamihara, 252-0373, Japan
| | - Motoki Imai
- Regenerative Medicine and Cell Design Research Facility, School of Allied Health Sciences, Kitasato University, 1-15-1 Kitasato, Sagamihara, 252-0373, Japan
- Department of Molecular Diagnostics, School of Allied Health Sciences, Kitasato University, 1-15-1 Kitasato, Sagamihara, 252-0373, Japan
| | - Takafumi Ichikawa
- Regenerative Medicine and Cell Design Research Facility, School of Allied Health Sciences, Kitasato University, 1-15-1 Kitasato, Sagamihara, 252-0373, Japan
- Department of Biochemistry, School of Allied Health Sciences, Kitasato University, 1-15-1 Kitasato, Sagamihara, 252-0373, Japan
| | - Naohito Ishii
- Department of Medical Laboratory Sciences, School of Allied Health Sciences, Kitasato University, 1-15-1 Kitasato, Sagamihara, 252-0373, Japan
- Regenerative Medicine and Cell Design Research Facility, School of Allied Health Sciences, Kitasato University, 1-15-1 Kitasato, Sagamihara, 252-0373, Japan
| | - Hidero Kitasato
- Regenerative Medicine and Cell Design Research Facility, School of Allied Health Sciences, Kitasato University, 1-15-1 Kitasato, Sagamihara, 252-0373, Japan
- Department of Environmental Microbiology, Kitasato University Graduate School of Medical Sciences, 1-15-1 Kitasato, Sagamihara, 252-0373, Japan
| | - Hideaki Hanaki
- Infection Control Research Center, Ōmura Satoshi Memorial Institute, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo, 108-8641, Japan
| | - Makoto Kubo
- Regenerative Medicine and Cell Design Research Facility, School of Allied Health Sciences, Kitasato University, 1-15-1 Kitasato, Sagamihara, 252-0373, Japan.
- Department of Environmental Microbiology, Kitasato University Graduate School of Medical Sciences, 1-15-1 Kitasato, Sagamihara, 252-0373, Japan.
| |
Collapse
|
41
|
Mahroum N, Habra M, Alrifaai MA, Shoenfeld Y. Antiphospholipid syndrome in the era of COVID-19 - Two sides of a coin. Autoimmun Rev 2024; 23:103543. [PMID: 38604461 DOI: 10.1016/j.autrev.2024.103543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 04/08/2024] [Accepted: 04/08/2024] [Indexed: 04/13/2024]
Abstract
In addition to the respiratory symptoms associated with COVID-19, the disease has consistently been linked to many autoimmune diseases such as systemic lupus erythematous and antiphospholipid syndrome (APS). APS in particular was of paramount significance due to its devastating clinical sequela. In fact, the hypercoagulable state seen in patients with acute COVID-19 and the critical role of anticoagulant treatment in affected individuals shed light on the possible relatedness between APS and COVID-19. Moreover, the role of autoimmunity in the assumed association is not less important especially with the accumulated data available regarding the autoimmunity-triggering effect of SARS-CoV-2 infection. This is furtherly strengthened at the time patients with COVID-19 manifested antiphospholipid antibodies of different types following infection. Additionally, the severe form of the APS spectrum, catastrophic APS (CAPS), was shown to have overlapping characteristics with severe COVID-19 such as cytokine storm and multi-organ failure. Interestingly, COVID vaccine-induced autoimmune phenomena described in the medical literature have pointed to an association with APS. Whether the antiphospholipid antibodies were present or de novo, COVID vaccine-induced vascular thrombosis in certain individuals necessitates further investigations regarding the possible mechanisms involved. In our current paper, we aimed to focus on the associations mentioned, their implications, importance, and consequences.
Collapse
Affiliation(s)
- Naim Mahroum
- International School of Medicine, Istanbul Medipol University, Istanbul, Turkey.
| | - Mona Habra
- International School of Medicine, Istanbul Medipol University, Istanbul, Turkey
| | | | - Yehuda Shoenfeld
- Zabludowicz Center for autoimmune diseases, Sheba Medical Center, Ramat-Gan, Israel; Reichman University, Herzliya, Israel
| |
Collapse
|
42
|
Akpoviroro O, Sauers NK, Uwandu Q, Castagne M, Akpoviroro OP, Humayun S, Mirza W, Woodard J. Severe COVID-19 infection: An institutional review and literature overview. PLoS One 2024; 19:e0304960. [PMID: 39163410 PMCID: PMC11335168 DOI: 10.1371/journal.pone.0304960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 05/21/2024] [Indexed: 08/22/2024] Open
Abstract
BACKGROUND Our study aimed to describe the group of severe COVID-19 patients at an institutional level, and determine factors associated with different outcomes. METHODS A retrospective chart review of patients admitted with severe acute hypoxic respiratory failure due to COVID-19 infection. Based on outcomes, we categorized 3 groups of severe COVID-19: (1) Favorable outcome: progressive care unit admission and discharge (2) Intermediate outcome: ICU care (3) Poor outcome: in-hospital mortality. RESULTS Eighty-nine patients met our inclusion criteria; 42.7% were female. The average age was 59.7 (standard deviation (SD):13.7). Most of the population were Caucasian (95.5%) and non-Hispanic (91.0%). Age, sex, race, and ethnicity were similar between outcome groups. Medicare and Medicaid patients accounted for 62.9%. The average BMI was 33.5 (SD:8.2). Moderate comorbidity was observed, with an average Charlson Comorbidity index (CCI) of 3.8 (SD:2.6). There were no differences in the average CCI between groups(p = 0.291). Many patients (67.4%) had hypertension, diabetes (42.7%) and chronic lung disease (32.6%). A statistical difference was found when chronic lung disease was evaluated; p = 0.002. The prevalence of chronic lung disease was 19.6%, 27.8%, and 40% in the favorable, intermediate, and poor outcome groups, respectively. Smoking history was associated with poor outcomes (p = 0.04). Only 7.9% were fully vaccinated. Almost half (46.1%) were intubated and mechanically ventilated. Patients spent an average of 12.1 days ventilated (SD:8.5), with an average of 6.0 days from admission to ventilation (SD:5.1). The intermediate group had a shorter average interval from admission to ventilator (77.2 hours, SD:67.6), than the poor group (212.8 hours, SD:126.8); (p = 0.001). The presence of bacterial pneumonia was greatest in the intermediate group (72.2%), compared to the favorable group (17.4%), and the poor group (56%); this was significant (p<0.0001). In-hospital mortality was seen in 28.1%. CONCLUSION Most patients were male, obese, had moderate-level comorbidity, a history of tobacco abuse, and government-funded insurance. Nearly 50% required mechanical ventilation, and about 28% died during hospitalization. Bacterial pneumonia was most prevalent in intubated groups. Patients who were intubated with a good outcome were intubated earlier during their hospital course, with an average difference of 135.6 hours. A history of cigarette smoking and chronic lung disease were associated with poor outcomes.
Collapse
Affiliation(s)
- Ogheneyoma Akpoviroro
- Department of Internal Medicine, Geisinger Wyoming Valley Medical Center, Wilkes-Barre, Pennsylvania, United States of America
| | - Nathan Kyle Sauers
- Department of Engineering, Pennsylvania State University, State College, Pennsylvania, United States of America
| | - Queeneth Uwandu
- Department of Internal Medicine, Geisinger Wyoming Valley Medical Center, Wilkes-Barre, Pennsylvania, United States of America
| | - Myriam Castagne
- Clinical & Translational Science Institute, Boston University, Boston, Massachusetts, United States of America
| | | | - Sara Humayun
- Department of Internal Medicine, Geisinger Wyoming Valley Medical Center, Wilkes-Barre, Pennsylvania, United States of America
| | - Wasique Mirza
- Department of Internal Medicine, Geisinger Wyoming Valley Medical Center, Wilkes-Barre, Pennsylvania, United States of America
| | - Jameson Woodard
- Department of Internal Medicine, Geisinger Wyoming Valley Medical Center, Wilkes-Barre, Pennsylvania, United States of America
| |
Collapse
|
43
|
Yuan L, Mehmood A, Meng L. A meta-analysis of risk factors for acute kidney injury in pneumonia: Effectiveness of nursing interventions. Ther Apher Dial 2024; 28:518-533. [PMID: 38545743 DOI: 10.1111/1744-9987.14124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 01/10/2024] [Accepted: 03/01/2024] [Indexed: 07/05/2024]
Abstract
INTRODUCTION The spread of coronavirus disease 2019 (COVID-19) worldwide since November 2019 is of interest to understand its impact on various organs. COVID-19 patients experience a higher incidence of acute kidney injury (AKI) compared with non-COVID-19 patients. METHODS A systematic literature search was conducted that covered the period from November 1, 2019 to February 28, 2021. RESULTS The analysis incorporated a comprehensive review of 19 studies of 21 362 patients. The older age (mean difference [MDs] = 5.11), cardiovascular disease (CVD) (odds ratio [OR] = 1.94), male sex (OR = 1.55), chronic kidney disease (CKD) (OR = 3.82), hypertension (OR = 2.15), diabetes (OR = 1.71), cancer (OR = 1.16), and chronic obstructive pulmonary disease (COPD) (OR = 1.40), mechanical ventilation (OR = 8.66), and vasopressor (OR = 6.30), were significantly associated with risk factor for AKI (P < 0.05). CONCLUSION The analysis revealed independent risk factors for AKI.
Collapse
Affiliation(s)
- Liangjuan Yuan
- Department of Respiratory, Shandong Provincial Third Hospital, Jinan, China
| | - Arshad Mehmood
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Lei Meng
- Department of Nephrology, The First Affiliated Hospital of Shandong First Medical University (Shandong Provincial Qianfoshan Hospital), Jinan, China
| |
Collapse
|
44
|
Gheban-Roşca IA, Gheban BA, Pop B, Mironescu DC, Siserman VC, Jianu EM, Drugan T, Bolboacă SD. A histopathological analysis of extrapulmonary lesions in fatal coronavirus disease (COVID-19). Pathol Res Pract 2024; 260:155373. [PMID: 38901140 DOI: 10.1016/j.prp.2024.155373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 05/26/2024] [Accepted: 05/28/2024] [Indexed: 06/22/2024]
Abstract
INTRODUCTION The coronavirus disease 2019 (COVID-19) pandemic, caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), presents diverse clinical manifestations and multi-organ involvement. This study aimed to evaluate the extra-pulmonary histopathological patterns underpinning COVID-19-induced lesions in cardiac, hepatic, renal, brainstem, and splenic tissues. MATERIALS AND METHODS The research involved conventional forensic autopsies conducted between April 2020 and April 2021 on individuals with confirmed SARS-CoV-2 infection in Cluj-Napoca, Romania. Tissues were processed and stained for histological examination. Differences in patients with and without diffuse alveolar damage (DAD) were evaluated. RESULTS In our study of 79 COVID-19 autopsies conducted on unvaccinated patients besides lung involvement, the patients had histological changes in at least two out of five (brain, heart, liver, kidney, and spleen) organs. Notable findings include hepatitis observed in 46.8 % of cases, 21.5 % with lobular hepatitis, and 41.8 % with liver steatosis. Additionally, 69.6 % exhibited acute tubular necrosis, and 55.7 % had varying degrees of splenic lymphocyte depletion. Almost 41 % of cases had pericardial effusion, 36.7 % myocarditis, 24.1 % myocardial infarction, and 12.7 % of cases had encephalitis. Acute tubular necrosis (78.6 %) was the most frequent histopathological finding observed in patients with DAD. Myocarditis was described in 45.9 % of the patients without DAD. DISCUSSION The autopsy findings in our cohort of COVID-19 victims align with international scientific literature. Distinguishing viral-induced myocarditis, encephalitis, hepatitis, or systemic inflammatory syndrome remains challenging. CONCLUSION Post-mortem analysis identified lesions associated with SARS-CoV-2 in multiple organs, highlighting the systemic nature of the virus and emphasizing the need for continued research into organ-specific damage and long-term sequelae of COVID-19.
Collapse
Affiliation(s)
- Ioana-Andreea Gheban-Roşca
- Department of Medical Informatics and Biostatistics, Iuliu Hațieganu University of Medicine and Pharmacy, Cluj-Napoca 400347, Romania; Clinical Hospital for Infectious Diseases, Cluj-Napoca 400003, Romania
| | - Bogdan-Alexandru Gheban
- County Emergency Clinical Hospital, Cluj-Napoca 400347, Romania; Department of Histology, Iuliu Hațieganu University of Medicine and Pharmacy, Cluj-Napoca 400347, Romania.
| | - Bogdan Pop
- The Oncology Institute " Prof. Dr. Ion Chiricuță", Cluj-Napoca 400015, Romania; Department of Anatomic Pathology, Iuliu Hațieganu University of Medicine and Pharmacy, Cluj-Napoca 400347, Romania
| | - Daniela-Cristina Mironescu
- Forensic Institute, Cluj-Napoca 400006, Romania; Department of Forensic Medicine, Iuliu Hațieganu University of Medicine and Pharmacy, Cluj-Napoca 400347, Romania
| | - Vasile Costel Siserman
- Forensic Institute, Cluj-Napoca 400006, Romania; Department of Forensic Medicine, Iuliu Hațieganu University of Medicine and Pharmacy, Cluj-Napoca 400347, Romania
| | - Elena Mihaela Jianu
- Department of Histology, Iuliu Hațieganu University of Medicine and Pharmacy, Cluj-Napoca 400347, Romania
| | - Tudor Drugan
- Department of Medical Informatics and Biostatistics, Iuliu Hațieganu University of Medicine and Pharmacy, Cluj-Napoca 400347, Romania
| | - Sorana D Bolboacă
- Department of Medical Informatics and Biostatistics, Iuliu Hațieganu University of Medicine and Pharmacy, Cluj-Napoca 400347, Romania
| |
Collapse
|
45
|
Nova A, McNicholas B, Magliocca A, Laffey M, Zambelli V, Mariani I, Atif M, Giacomini M, Vitale G, Rona R, Foti G, Laffey J, Rezoagli E. Perfusion deficits may underlie lung and kidney injury in severe COVID-19 disease: insights from a multicenter international cohort study. JOURNAL OF ANESTHESIA, ANALGESIA AND CRITICAL CARE 2024; 4:40. [PMID: 38971842 PMCID: PMC11227201 DOI: 10.1186/s44158-024-00175-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Accepted: 06/25/2024] [Indexed: 07/08/2024]
Abstract
BACKGROUND Lung perfusion defects, mainly due to endothelial and coagulation activation, are a key contributor to COVID-19 respiratory failure. COVID-19 patients may also develop acute kidney injury (AKI) because of renal perfusion deficit. We aimed to explore AKI-associated factors and the independent prediction of standardized minute ventilation (MV)-a proxy of alveolar dead space-on AKI onset and persistence in COVID-19 mechanically ventilated patients. METHODS This is a multicenter observational cohort study. We enrolled 157 COVID-19 patients requiring mechanical ventilation and intensive care unit (ICU) admission. We collected clinical information, ventilation, and laboratory data. AKI was defined by the 2012 KDIGO guidelines and classified as transient or persistent according to serum creatinine criteria persistence within 48 h. Ordered univariate and multivariate logistic regression analyses were employed to identify variables associated with AKI onset and persistence. RESULTS Among 157 COVID-19 patients on mechanical ventilation, 47% developed AKI: 10% had transient AKI, and 37% had persistent AKI. The degree of hypoxia was not associated with differences in AKI severity. Across increasing severity of AKI groups, despite similar levels of paCO2, we observed an increased MV and standardized MV, a robust proxy of alveolar dead space. After adjusting for other clinical and laboratory covariates, standardized MV remained an independent predictor of AKI development and persistence. D-dimer levels were higher in patients with persistent AKI. CONCLUSIONS In critically ill COVID-19 patients with respiratory failure, increased wasted ventilation is independently associated with a greater risk of persistent AKI. These hypothesis-generating findings may suggest that perfusion derangements may link the pathophysiology of both wasted ventilation and acute kidney injury in our population.
Collapse
Affiliation(s)
- Alice Nova
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Bairbre McNicholas
- School of Medicine, National University of Ireland Galway, Galway, Ireland
- Department of Anesthesia and Intensive Care Medicine, Galway University Hospitals, Galway, Ireland
| | - Aurora Magliocca
- Department of Anesthesia and Intensive Care Medicine, Gruppo Ospedaliero San Donato, Policlinico San Marco, Zingonia, Bergamo, Italy
- Department of Pathophysiology and Transplants, University of Milan, Milan, Italy
| | - Matthew Laffey
- School of Medicine, Trinity College Dublin, Dublin 2, Ireland
| | - Vanessa Zambelli
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Ilaria Mariani
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Minahel Atif
- School of Medicine, National University of Ireland Galway, Galway, Ireland
- Department of Anesthesia and Intensive Care Medicine, Galway University Hospitals, Galway, Ireland
| | - Matteo Giacomini
- Department of Anesthesia and Intensive Care Medicine, Gruppo Ospedaliero San Donato, Policlinico San Marco, Zingonia, Bergamo, Italy
| | - Giovanni Vitale
- Department of Anesthesia and Intensive Care Medicine, Gruppo Ospedaliero San Donato, Policlinico San Marco, Zingonia, Bergamo, Italy
| | - Roberto Rona
- Department of Emergency and Intensive Care, Fondazione IRCCS San Gerardo dei Tintori Hospital, Monza, Italy
| | - Giuseppe Foti
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
- Department of Emergency and Intensive Care, Fondazione IRCCS San Gerardo dei Tintori Hospital, Monza, Italy
| | - John Laffey
- School of Medicine, National University of Ireland Galway, Galway, Ireland
- Department of Anesthesia and Intensive Care Medicine, Galway University Hospitals, Galway, Ireland
| | - Emanuele Rezoagli
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy.
- Department of Emergency and Intensive Care, Fondazione IRCCS San Gerardo dei Tintori Hospital, Monza, Italy.
| |
Collapse
|
46
|
Fisher M, Ross M, DiFranza L, Reidy K. An Update on Viral Infection-Associated Collapsing Glomerulopathy. ADVANCES IN KIDNEY DISEASE AND HEALTH 2024; 31:317-325. [PMID: 39084757 PMCID: PMC11296492 DOI: 10.1053/j.akdh.2023.12.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Revised: 11/14/2023] [Accepted: 12/14/2023] [Indexed: 08/02/2024]
Abstract
The COVID-19 era has been a reminder to clinicians around the world of the important role that viral infections play in promoting glomerular disease. Several viral infections including human immunodeficiency virus (HIV), severe acute respiratory syndrome coronavirus 2, Epstein-Barr virus, cytomegalovirus, and parvovirus B19 can cause podocyte injury and present with a collapsing glomerulopathy (CG) variant of focal segmental glomerulosclerosis or minimal change disease. CG associated with COVID-19 has been termed COVID-19-associated nephropathy due to its striking resemblance to HIV-associated nephropathy. Host susceptibility is a major determinant of viral infection-associated CG, and the presence of two APOL1 risk variants explains most of the racial predilection to viral-associated CG observed in individuals of African ancestry. Interactions between APOL1 risk variants, viral genes, and the systemic inflammatory response to viral infection all contribute to kidney injury. This review will summarize our current knowledge of viral infection-associated CG, focusing primarily on the clinical presentation, histological features, mechanisms, and disease course of HIV-associated nephropathy and COVID-19-associated nephropathy.
Collapse
Affiliation(s)
- Molly Fisher
- Division of Nephrology, Albert Einstein College of Medicine, Montefiore Health System, Bronx, NY.
| | - Michael Ross
- Division of Nephrology, Albert Einstein College of Medicine, Montefiore Health System, Bronx, NY; Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY
| | - Lanny DiFranza
- Department of Pathology, Albert Einstein College of Medicine, Montefiore Health System, Bronx, NY
| | - Kimberly Reidy
- Division of Pediatric Nephrology, The Children's Hospital at Montefiore/Albert Einstein College of Medicine, Bronx, NY
| |
Collapse
|
47
|
Özdemir BH, Ok Atılgan A, Akyüz Özdemir A, Haberal M. Unmasking the Silent Threat: COVID-19's Pervasive Impact on Renal Allografts. EXP CLIN TRANSPLANT 2024; 22:522-530. [PMID: 39223810 DOI: 10.6002/ect.2023.0352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
OBJECTIVES Growing evidence has highlighted the substantial effects of COVID-19 on kidneys, ranging from mild proteinuria to severe acute kidney injury. However, comprehensive assessments of histopathological features in renal allograft biopsies are lacking. MATERIALS AND METHODS Seventeen kidney transplant recipients with COVID-19 between March 2020 and November 2022 were evaluated. Clinical characteristics, pathological findings, and outcomes were studied. RESULTS Six kidney transplant recipients (35.3%) developed acute kidney injury, leading to the requirement for hemodialysis. COVID-19 severity, as indicated by pneumonia (P = .028) and hospitalization (P = .002), was significantly associated with development of acute kidney injury. Most patients with COVID-19 (82.4%) showed considerably increased proteinuria levels (82.4%), along with presence of new-onset microscopic hematuria (35.3%) and nephrotic syndrome (58.8%). Tubular viral inclusionlike changes were detected in 47.1% of cases and were associated with a higher risk of graft loss (75%). Thrombotic microangiopathy and endothelial cell swelling in glomeruli were prevalent, highlighting extensive endothelial cell injury. Most recipients (88.2%) experienced rejection after COVID-19, with graft loss occurring in 46.7% of these cases. Biopsies revealed collapsing (n = 5), noncollapsing (n = 3), and recurrent (n = 2) focal segmental glomerulosclerosis, as well as acute tubulointerstitial nephritis (n = 3), crescentic glomerulonephritis with immunoglobulin A nephropathy (n = 1), and membranoproliferative glomerulonephritis (n = 1), in 129.7 ± 33 days. Eight patients experienced graft loss (8.2 ± 2 mo posttransplant). Hospitalization (P = .044) and viralinclusion-like nuclear changes in tubules (P = .044) significantly influenced graft survival. Collapsing (60%) and noncollapsing (66.7%) focal segmental glomerulosclerosis increased the risk of graft loss. CONCLUSIONS COVID-19 has had a multifaceted and enduring effect on renal allografts, urging the need for meticulous monitoring and tailored management strategies to mitigate the risk of severe kidney-related complications and graft loss in this vulnerable population.
Collapse
Affiliation(s)
- B Handan Özdemir
- >From the Pathology Department, Faculty of Medicine, Baskent University, Ankara, Türkiye
| | | | | | | |
Collapse
|
48
|
Kamo M, Sogawa R, Shimanoe C. Association of Antiviral Drugs for the Treatment of COVID-19 With Acute Renal Failure. In Vivo 2024; 38:1841-1846. [PMID: 38936945 PMCID: PMC11215623 DOI: 10.21873/invivo.13637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 03/13/2024] [Accepted: 03/14/2024] [Indexed: 06/29/2024]
Abstract
BACKGROUND/AIM Reports regarding the association of remdesivir use for the treatment of Coronavirus disease 2019 (COVID-19) with the development of acute kidney injury (AKI) are inconsistent, and the associations between the use of other antivirals and AKI remain unclear. Therefore, this study investigated whether the use of antiviral drugs for the treatment of COVID-19 is a risk factor for the development of AKI. PATIENTS AND METHODS This study analyzed 176,197 reports submitted to the Japanese Adverse Event Reporting Database between 2020 and 2022. Reporting odds ratios (RORs) and 95% confidence intervals (95%CIs) for AKI that were associated with the use of antiviral drugs in patients with COVID-19 were calculated after adjusting for potential confounders. RESULTS Overall, 5,879 of the reports analyzed were associated with AKI. Signs of AKI were detected with the use of remdesivir [crude ROR (cROR)=2.45; 95%CI=1.91-3.14] and nirmatrelvir/ritonavir (cROR=6.07; 95%CI=4.06-9.06). These results were maintained even after adjusting for potential confounders [remdesivir: adjusted ROR (aROR)=2.18; 95%CI=1.69-2.80, nirmatrelvir/ritonavir: aROR=5.24; 95%CI=3.48-7.90]. However, when analyzing data stratified by reporting year, the association between remdesivir and AKI appeared to diminish over time and was not sustained. CONCLUSION Nirmatrelvir/ritonavir use may be associated with developing AKI. This knowledge may be useful in helping patients with COVID-19 avoid AKI complications.
Collapse
Affiliation(s)
- Masahiro Kamo
- Department of Pharmacy, Saga University Hospital, Saga, Japan
| | - Rintaro Sogawa
- Department of Pharmacy, Saga University Hospital, Saga, Japan
| | | |
Collapse
|
49
|
De Grazia S, Pollicino F, Giannettino C, Errera CM, Veronese N, Giammanco GM, Cacioppo F, Sanfilippo GL, Barbagallo M, COMEPA Study Authors. Factors Associated with Prolonged SARS-CoV-2 Viral Positivity in an Italian Cohort of Hospitalized Patients. Diseases 2024; 12:138. [PMID: 39057109 PMCID: PMC11275323 DOI: 10.3390/diseases12070138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 06/24/2024] [Accepted: 06/26/2024] [Indexed: 07/28/2024] Open
Abstract
Clinical or microbiological factors potentially associated with prolonged COVID-19 PCR positivity are still poorly underexplored, but they could be of importance for public-health and clinical reasons. The objective of our analysis is to explore demographic, clinical, and microbiological factors potentially associated with a prolonged positivity to SARS-CoV-2 among 222 hospitalized patients. Prolonged detection positivity for SARS-CoV-2 RNA in swap samples, defined as positivity more than 21 days, was the outcome of interest. The 56 cases with a prolonged positivity to SARS-CoV-2 were matched for age and sex with 156 controls. The cases reported a significantly higher presence of diabetes mellitus, autoimmune diseases, chronic kidney diseases, and acute coronary syndrome. Moreover, the viral load was significantly higher in a period of prolonged positivity compared to a normal period. In the multivariable analysis, the presence of autoimmune diseases and chronic kidney disease were significantly associated with an increased risk of prolonged positivity as well as medium viral load or high viral load, i.e., low Ct value ≤ 30 indicating high viral load. The results of this study confirmed that in a large population of hospitalized patients with COVID-19 manifestations, the prolonged positivity of SARS-CoV-2 detection with nasopharyngeal swab was mainly related to autoimmune diseases, chronic kidney disease, and to baseline viral load.
Collapse
Affiliation(s)
| | | | | | | | - Nicola Veronese
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties “G. D’Alessandro”, University of Palermo, 90127 Palermo, Italy; (S.D.G.); (F.P.); (C.G.); (C.M.E.); (G.M.G.); (F.C.); (G.L.S.); (M.B.)
| | | | | | | | | | | |
Collapse
|
50
|
Sideratou CM, Papaneophytou C. Persistent Vascular Complications in Long COVID: The Role of ACE2 Deactivation, Microclots, and Uniform Fibrosis. Infect Dis Rep 2024; 16:561-571. [PMID: 39051242 PMCID: PMC11270324 DOI: 10.3390/idr16040042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 06/25/2024] [Accepted: 06/26/2024] [Indexed: 07/27/2024] Open
Abstract
Angiotensin-converting enzyme 2 (ACE2), a key regulator in vasoregulation and the renin-angiotensin system, is hypothesized to be downregulated in patients with COVID-19, leading to a cascade of cardiovascular complications. This deactivation potentially results in increased blood pressure and vessel injury, contributing to the formation and persistence of microclots in the circulation. Herein, we propose a hypothesis regarding the prolonged vascular complications observed in long COVID, focusing on the role of ACE2 deactivation and/or shedding, the persistence of microclots, and the unique pattern of fibrosis induced by severe acute respiratory syndrome-coronavirus-2 (SARS-CoV-2). Furthermore, we propose that the distinctive, uniform fibrosis associated with COVID-19, which is challenging to detect through conventional X-ray imaging, exacerbates vascular injury and impairs oxygenation. The persistence of these microclots and the unique fibrosis pattern are suggested as key factors in the extended duration of vascular complications post-COVID-19 infection, regardless of the initial disease severity. Moreover, plasma ACE2 activity has the potential to serve as prognostic or diagnostic biomarkers for monitoring disease severity and managing long COVID symptoms. Elucidating the role of ACE2 deactivation and the consequent events is vital for understanding the long-term effects of COVID-19. The experimental verification of this hypothesis through in vitro studies, clinical longitudinal studies, and advanced imaging techniques could yield significant insights into the pathophysiological mechanisms underlying long COVID, thereby improving the management of patients, particularly those with cardiovascular complications.
Collapse
Affiliation(s)
| | - Christos Papaneophytou
- Department of Life Sciences, School of Life and Health Sciences, University of Nicosia, 2417 Nicosia, Cyprus;
| |
Collapse
|