51
|
Haghpanah A, Masjedi F, Salehipour M, Hosseinpour A, Roozbeh J, Dehghani A. Is COVID-19 a risk factor for progression of benign prostatic hyperplasia and exacerbation of its related symptoms?: a systematic review. Prostate Cancer Prostatic Dis 2022; 25:27-38. [PMID: 34007019 PMCID: PMC8129694 DOI: 10.1038/s41391-021-00388-3] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 04/28/2021] [Accepted: 04/30/2021] [Indexed: 02/08/2023]
Abstract
BACKGROUND To explore the potential mechanisms of SARS-CoV-2 in targeting the prostate gland, leading to exacerbation of benign prostatic hyperplasia (BPH) symptoms and greater risks of BPH complications such as acute urinary retention. METHODS A categorized and comprehensive search in the literature has been conducted by 10 April 2021 using international databases including PubMed, Embase, Web of Science, Scopus, and Cochrane Library in line with the PRISMA guidelines recommendations. PICO strategy was used to formulate the research question. The following terms were used: urology, COVID-19, coronavirus, BPH, inflammation, androgen receptors, LUTS, IPSS, PSA, and SARS-CoV-2 or a combination of them. Studies with irrelevant purposes and duplicates were excluded. The selected studies were performed on humans and published in English. RESULTS The research revealed 89 articles. After title screening and considering exclusion criteria, 52 papers were included for the systematic review. BPH is a common condition affecting older men. SARS-CoV-2 infects the host cell by binding to angiotensin converting enzyme 2 (ACE2). A hyperactivated RAS system during infection with SARS-CoV-2 may lead to activation of pro-inflammatory pathways and increased cytokine release. Thus, this virus can lead to exacerbation of lower urinary tract symptoms (LUTS) and trigger inflammatory processes in the prostate gland. Since androgen receptors (AR) play an important role in the BPH pathophysiology and infection with SARS-CoV-2 may be androgen-mediated, BPH progression and its related symptoms can be a complication of COVID-19 through AR involvement and metabolic disturbances. CONCLUSIONS Based on the current findings, SARS-CoV-2 can possibly damage the prostate and worsen BPH and its related LUTS through ACE2 signaling, AR-related mechanisms, inflammation, and metabolic derangement. We encourage future studies to investigate the possible role of COVID-19 in the progression of BPH-related LUTS and examine the prostatic status in susceptible patients with relevant available questionnaires (e.g., IPSS) and serum biomarkers (e.g., PSA).
Collapse
Affiliation(s)
- Abdolreza Haghpanah
- grid.412571.40000 0000 8819 4698Shiraz Nephro-Urology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran ,grid.412571.40000 0000 8819 4698Department of Urology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Fatemeh Masjedi
- grid.412571.40000 0000 8819 4698Shiraz Nephro-Urology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mehdi Salehipour
- grid.412571.40000 0000 8819 4698Department of Urology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Alireza Hosseinpour
- grid.412571.40000 0000 8819 4698Shiraz Nephro-Urology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran ,grid.412571.40000 0000 8819 4698Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Jamshid Roozbeh
- grid.412571.40000 0000 8819 4698Shiraz Nephro-Urology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Anahita Dehghani
- grid.412571.40000 0000 8819 4698Shiraz Nephro-Urology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
52
|
Arghiani N, Nissan T, Matin MM. Role of microRNAs in COVID-19 with implications for therapeutics. Biomed Pharmacother 2021; 144:112247. [PMID: 34601190 PMCID: PMC8463393 DOI: 10.1016/j.biopha.2021.112247] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 09/21/2021] [Accepted: 09/22/2021] [Indexed: 02/09/2023] Open
Abstract
COVID-19 is a pneumonia-like disease with highly transmittable and pathogenic properties caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), which infects both animals and humans. Although many efforts are currently underway to test possible therapies, there is no specific FDA approved drug against SARS-CoV-2 yet. miRNA-directed gene regulation controls the majority of biological processes. In addition, the development and progression of several human diseases are associated with dysregulation of miRNAs. In this regard, it has been shown that changes in miRNAs are linked to severity of COVID-19 especially in patients with respiratory diseases, diabetes, heart failure or kidney problems. Therefore, targeting these small noncoding-RNAs could potentially alleviate complications from COVID-19. Here, we will review the roles and importance of host and RNA virus encoded miRNAs in COVID-19 pathogenicity and immune response. Then, we focus on potential miRNA therapeutics in the patients who are at increased risk for severe disease.
Collapse
Affiliation(s)
- Nahid Arghiani
- Department of Molecular Biosciences, the Wenner-Gren Institute, Stockholm University, Stockholm, Sweden; School of Life Science, Department of Biochemistry and Biomedicine, University of Sussex, Brighton, United Kingdom
| | - Tracy Nissan
- Department of Molecular Biosciences, the Wenner-Gren Institute, Stockholm University, Stockholm, Sweden; School of Life Science, Department of Biochemistry and Biomedicine, University of Sussex, Brighton, United Kingdom.
| | - Maryam M Matin
- Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran; Novel Diagnostics and Therapeutics Research Group, Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran; Stem Cell and Regenerative Medicine Research Group, Iranian Academic Center for Education, Culture and Research (ACECR), Khorasan Razavi Branch, Mashhad, Iran.
| |
Collapse
|
53
|
Kalejaiye TD, Bhattacharya R, Burt MA, Travieso T, Okafor AE, Mou X, Blasi M, Musah S. BSG/CD147 and ACE2 receptors facilitate SARS-CoV-2 infection of human iPS cell-derived kidney podocytes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2021. [PMID: 34816259 DOI: 10.1101/2021.11.16.468893] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
BACKGROUND Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) causes the Coronavirus disease 2019 (COVID-19), which was declared a pandemic by the World Health Organization (WHO) in March 2020. The disease has caused more than 5.1 million deaths worldwide. While cells in the respiratory system are frequently the initial target for SARS-CoV-2, clinical studies suggest that COVID-19 can become a multi-organ disease in the most severe cases. Still, the direct affinity of SARS-CoV-2 for cells in other organs such as the kidneys, which are often affected in severe COVID-19, remains poorly understood. METHOD In this study, we employed a human induced pluripotent stem (iPS) cell-derived model to investigate the affinity of SARS-CoV-2 for kidney glomerular podocytes. We studied uptake of the live SARS-CoV-2 virus as well as pseudotyped viral particles by human iPS cell derived podocytes using qPCR, western blot, and immunofluorescence. Global gene expression and qPCR analyses revealed that human iPS cell-derived podocytes express many host factor genes (including ACE2, BSG/CD147, PLS3, ACTR3, DOCK7, TMPRSS2, CTSL CD209, and CD33) associated with SARS-CoV-2 binding and viral processing. RESULT Infection of podocytes with live SARS-CoV-2 or spike-pseudotyped lentiviral particles revealed viral uptake by the cells at low Multiplicity of Infection (MOI of 0.01) as confirmed by RNA quantification and immunofluorescence studies. Our results also indicate that direct infection of human iPS cell-derived podocytes by SARS-CoV-2 virus can cause cell death and podocyte foot process retraction, a hallmark of podocytopathies and progressive glomerular diseases including collapsing glomerulopathy observed in patients with severe COVID-19 disease. Additionally, antibody blocking experiments identified BSG/CD147 and ACE2 receptors as key mediators of spike binding activity in human iPS cell-derived podocytes. CONCLUSION These results show that SARS-CoV-2 can infect kidney glomerular podocytes in vitro . These results also show that the uptake of SARS-CoV-2 by kidney podocytes occurs via multiple binding interactions and partners, which may underlie the high affinity of SARS-CoV-2 for kidney tissues. This stem cell-derived model is potentially useful for kidney-specific antiviral drug screening and mechanistic studies of COVID-19 organotropism. SIGNIFICANT STATEMENT Many patients with COVID19 disease exhibit multiorgan complications, suggesting that SARS-CoV-2 infection can extend beyond the respiratory system. Acute kidney injury is a common COVID-19 complication contributing to increased morbidity and mortality. Still, SARS-Cov-2 affinity for specialized kidney cells remain less clear. By leveraging our protocol for stem cell differentiation, we show that SARS-CoV-2 can directly infect kidney glomerular podocytes by using multiple Spike-binding proteins including ACE2 and BSG/CD147. Our results also indicate that infection by SARS-CoV-2 virus can cause podocyte cell death and foot process effacement, a hallmark of podocytopathies including collapsing glomerulopathy observed in patients with severe COVID-19 disease. This stem cell-derived model is potentially useful for kidney-specific antiviral drug screening and mechanistic studies of COVID-19 organotropism.
Collapse
|
54
|
Elkholi MH, Alrais ZF, Algouhary AR, Al-Taie MS, Sawwan AA, Khalafalla AA, Beniamein MM, Alkhouly AE, Shoaib MI, Alkholy HE, Abdel Hadi AM, Abu Alkhair AT. Acute kidney injury in ventilated patients with coronavirus disease-2019 pneumonia: A single-center retrospective study. Int J Crit Illn Inj Sci 2021; 11:123-133. [PMID: 34760658 PMCID: PMC8547683 DOI: 10.4103/ijciis.ijciis_194_20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Revised: 02/02/2021] [Accepted: 06/07/2021] [Indexed: 11/05/2022] Open
Abstract
Background: Acute kidney injury (AKI) is repeatedly observed in ventilated critically ill patients with coronavirus disease-2019 (COVID-19) pneumonia. This study aimed to determine the incidence, risk factors, and consequences of AKI in the ventilated critically ill adult patients with COVID-19 pneumonia. Methods: This retrospective study included all the ventilated critically ill adult patients with COVID-19 pneumonia from March 1, 2020, to June 1, 2020. Data were collected from the electronic medical system. AKI was diagnosed using the Kidney Disease: Improving Global Outcomes 2012 Clinical Practice definition. Patients were followed 90 days from the intensive care unit (ICU) admission time or to the date when they were discharged from the hospital. Results: AKI occurred in 65.1% of patients, with 26.6% of these started on continuous renal replacement therapy (CRRT). Patients with AKI had higher comorbidity and illness severity scores (P < 0.001). Age and the vasopressor requirements were predictors of AKI (P= 0.016 and P = 0.041) and hypertension predicted AKI (P = 0.099) and its progression (P = 0.05). The renal recovery rate was 86.7% and was associated with the mean arterial pressure on ICU admission in the no-CRRT group (P = 0.014) and the hypoxic index in the CRRT group (P = 0.019). AKI was associated with higher mortality (P = 0.017) and significantly longer ICU length-of-stay (P = 0.001). Additionally, AKI patients were more often discharged to a long-term skilled nursing facility (P = 0.005). Conclusion: COVID-19-associated AKI was common and associated with poor outcome, with the specific mechanisms being the main driving factors.
Collapse
|
55
|
Yanofsky SM, Dugas CM, Katsurada A, Liu J, Saifudeen Z, El-Dahr SS, Satou R. Angiotensin II biphasically regulates cell differentiation in human iPSC-derived kidney organoids. Am J Physiol Renal Physiol 2021; 321:F559-F571. [PMID: 34448643 PMCID: PMC8616599 DOI: 10.1152/ajprenal.00134.2021] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 08/09/2021] [Accepted: 08/25/2021] [Indexed: 12/28/2022] Open
Abstract
Human kidney organoid technology holds promise for novel kidney disease treatment strategies and utility in pharmacological and basic science. Given the crucial roles of the intrarenal renin-angiotensin system (RAS) and angiotensin II (ANG II) in the progression of kidney development and injury, we investigated the expression of RAS components and effects of ANG II on cell differentiation in human kidney organoids. Human induced pluripotent stem cell-derived kidney organoids were induced using a modified 18-day Takasato protocol. Gene expression analysis by digital PCR and immunostaining demonstrated the formation of renal compartments and expression of RAS components. The ANG II type 1 receptor (AT1R) was strongly expressed in the early phase of organoid development (around day 0), whereas ANG II type 2 receptor (AT2R) expression levels peaked on day 5. Thus, the organoids were treated with 100 nM ANG II in the early phase on days 0-5 (ANG II-E) or during the middle phase on days 5-10 (ANG II-M). ANG II-E was observed to decrease levels of marker genes for renal tubules and proximal tubules, and the downregulation of renal tubules was inhibited by an AT1R antagonist. In contrast, ANG II-M increased levels of markers for podocytes, the ureteric tip, and the nephrogenic mesenchyme, and an AT2R blocker attenuated the ANG II-M-induced augmentation of podocyte formation. These findings demonstrate RAS expression and ANG II exertion of biphasic effects on cell differentiation through distinct mediatory roles of AT1R and AT2R, providing a novel strategy to establish and further characterize the developmental potential of human induced pluripotent stem cell-derived kidney organoids.NEW & NOTEWORTHY This study demonstrates angiotensin II exertion of biphasic effects on cell differentiation through distinct mediatory roles of angiotensin II type 1 receptor and type 2 receptor in human induced pluripotent stem cell-derived kidney organoids, providing a novel strategy to establish and further characterize the developmental potential of the human kidney organoids.
Collapse
MESH Headings
- Angiotensin II/pharmacology
- Cell Differentiation/drug effects
- Cell Line
- Gene Expression Regulation, Developmental
- Humans
- Induced Pluripotent Stem Cells/drug effects
- Induced Pluripotent Stem Cells/metabolism
- Kidney/cytology
- Kidney/drug effects
- Kidney/metabolism
- Organoids/cytology
- Organoids/drug effects
- Organoids/metabolism
- Receptor, Angiotensin, Type 1/agonists
- Receptor, Angiotensin, Type 1/genetics
- Receptor, Angiotensin, Type 1/metabolism
- Receptor, Angiotensin, Type 2/agonists
- Receptor, Angiotensin, Type 2/genetics
- Receptor, Angiotensin, Type 2/metabolism
- Renin-Angiotensin System/drug effects
- Signal Transduction
- Time Factors
Collapse
Affiliation(s)
- Stacy M Yanofsky
- Department of Physiology and Hypertension and Renal Center of Excellence, Tulane University School of Medicine, New Orleans, Louisiana
| | - Courtney M Dugas
- Department of Physiology and Hypertension and Renal Center of Excellence, Tulane University School of Medicine, New Orleans, Louisiana
| | - Akemi Katsurada
- Department of Physiology and Hypertension and Renal Center of Excellence, Tulane University School of Medicine, New Orleans, Louisiana
| | - Jiao Liu
- Department of Pediatrics, Tulane University School of Medicine, New Orleans, Louisiana
| | - Zubaida Saifudeen
- Department of Pediatrics, Tulane University School of Medicine, New Orleans, Louisiana
| | - Samir S El-Dahr
- Department of Pediatrics, Tulane University School of Medicine, New Orleans, Louisiana
| | - Ryousuke Satou
- Department of Physiology and Hypertension and Renal Center of Excellence, Tulane University School of Medicine, New Orleans, Louisiana
| |
Collapse
|
56
|
Wang T, Xie F, Li YH, Liang B. Downregulation of ACE2 is associated with advanced pathological features and poor prognosis in clear cell renal cell carcinoma. Future Oncol 2021; 17:5033-5044. [PMID: 34704468 DOI: 10.2217/fon-2020-1164] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Aims: The aim of this study was to explore the alteration in ACE2 expression and correlation between ACE2 expression and immune infiltration in clear cell renal cell carcinoma (ccRCC). Methods: The authors first analyzed the expression profiles and prognostic value of ACE2 in ccRCC patients using The Cancer Genome Atlas public database. The authors used ESTIMATE and CIBERSORT algorithms to analyze the correlation between ACE2 expression and tumor microenvironment in ccRCC samples. Results: ACE2 was correlated with sex, distant metastasis, clinical stage, tumor T stage and histological grade. Moreover, downregulation of ACE2 was correlated with unfavorable prognosis. In addition, ACE2 expression was associated with different immune cell subtypes. Conclusion: The authors' analyses suggest that ACE2 plays an important role in the development and progression of ccRCC and may serve as a potential prognostic biomarker in ccRCC patients.
Collapse
Affiliation(s)
- Tianjiao Wang
- Bioinformatics Department, Key Laboratory of Cell Biology, Ministry of Public Health & Key Laboratory of Medical Cell Biology, Ministry of Education, School of Life Sciences, China Medical University, Shenyang 110122, China
| | - Fang Xie
- Medical Basic Experimental Teaching Center, China Medical University, Shenyang 110122, China
| | - Yun-Hui Li
- Department of Clinical Laboratory, General Hospital of PLA Northern Theater Command, Shenyang 110016, China
| | - Bin Liang
- Bioinformatics Department, Key Laboratory of Cell Biology, Ministry of Public Health & Key Laboratory of Medical Cell Biology, Ministry of Education, School of Life Sciences, China Medical University, Shenyang 110122, China
| |
Collapse
|
57
|
Wan L, Song J, He V, Roman J, Whah G, Peng S, Zhang L, He Y. Development of the International Classification of Diseases Ontology (ICDO) and its application for COVID-19 diagnostic data analysis. BMC Bioinformatics 2021; 22:508. [PMID: 34663204 PMCID: PMC8522253 DOI: 10.1186/s12859-021-04402-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 09/24/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The 10th and 9th revisions of the International Statistical Classification of Diseases and Related Health Problems (ICD10 and ICD9) have been adopted worldwide as a well-recognized norm to share codes for diseases, signs and symptoms, abnormal findings, etc. The international Consortium for Clinical Characterization of COVID-19 by EHR (4CE) website stores diagnosis COVID-19 disease data using ICD10 and ICD9 codes. However, the ICD systems are difficult to decode due to their many shortcomings, which can be addressed using ontology. METHODS An ICD ontology (ICDO) was developed to logically and scientifically represent ICD terms and their relations among different ICD terms. ICDO is also aligned with the Basic Formal Ontology (BFO) and reuses terms from existing ontologies. As a use case, the ICD10 and ICD9 diagnosis data from the 4CE website were extracted, mapped to ICDO, and analyzed using ICDO. RESULTS We have developed the ICDO to ontologize the ICD terms and relations. Different from existing disease ontologies, all ICD diseases in ICDO are defined as disease processes to describe their occurrence with other properties. The ICDO decomposes each disease term into different components, including anatomic entities, process profiles, etiological causes, output phenotype, etc. Over 900 ICD terms have been represented in ICDO. Many ICDO terms are presented in both English and Chinese. The ICD10/ICD9-based diagnosis data of over 27,000 COVID-19 patients from 5 countries were extracted from the 4CE. A total of 917 COVID-19-related disease codes, each of which were associated with 1 or more cases in the 4CE dataset, were mapped to ICDO and further analyzed using the ICDO logical annotations. Our study showed that COVID-19 targeted multiple systems and organs such as the lung, heart, and kidney. Different acute and chronic kidney phenotypes were identified. Some kidney diseases appeared to result from other diseases, such as diabetes. Some of the findings could only be easily found using ICDO instead of ICD9/10. CONCLUSIONS ICDO was developed to ontologize ICD10/10 codes and applied to study COVID-19 patient diagnosis data. Our findings showed that ICDO provides a semantic platform for more accurate detection of disease profiles.
Collapse
Affiliation(s)
- Ling Wan
- University of Michigan Medical School, Ann Arbor, MI 48109 USA
- OntoWise, Nanjing, Jiangsu China
| | - Justin Song
- Cranbrook Kingswood Upper School, Bloomfield Hills, MI 48304 USA
| | | | - Jennifer Roman
- College of Literacy, Science, and Arts, University of Michigan, Ann Arbor, MI 48109 USA
| | - Grace Whah
- College of Engineering, University of Michigan, Ann Arbor, MI 48109 USA
| | - Suyuan Peng
- School of Public Health, Peking University, Beijing, China
- National Institute of Health Data Science, Peking University, Beijing, China
| | - Luxia Zhang
- National Institute of Health Data Science, Peking University, Beijing, China
- Advanced Institute of Information Technology, Peking University, Hangzhou, China
- Renal Division, Department of Medicine, Peking University First Hospital, Peking University Institute of Nephrology, Beijing, China
| | - Yongqun He
- University of Michigan Medical School, Ann Arbor, MI 48109 USA
| |
Collapse
|
58
|
Emerging Role of Neuropilin-1 and Angiotensin-Converting Enzyme-2 in Renal Carcinoma-Associated COVID-19 Pathogenesis. Infect Dis Rep 2021; 13:902-909. [PMID: 34698182 PMCID: PMC8544489 DOI: 10.3390/idr13040081] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 10/14/2021] [Accepted: 10/15/2021] [Indexed: 12/13/2022] Open
Abstract
Neuropilin-1 (NRP1) is a recently identified glycoprotein that is an important host factor for SARS-CoV-2 infection. On the other hand, angiotensin-converting enzyme-2 (ACE2) acts as a receptor for SARS-CoV-2. Additionally, both NRP1 and ACE2 express in the kidney and are associated with various renal diseases, including renal carcinoma. Therefore, the expression profiles of NRP1 and ACE2 in kidney renal clear cell carcinoma (KIRC) and kidney renal papillary cell carcinoma (KIRP) patients from the various cancer databases were investigated along with their impact on patients’ survivability. In addition, coexpression analysis of genes involved in COVID-19, KIRC, and KIRP concerning NRP1 and ACE2 was performed. The results demonstrated that both t NRP1 and ACE2 expressions are upregulated in KIRC and KIRP compared to healthy conditions and are significantly correlated with the survivability rate of KIRC patients. A total of 128 COVID-19-associated genes are coexpressed, which are positively associated with NRP1 and ACE2 both in KIRC and KIRP. Therefore, it might be suggested that, along with the ACE2, high expression of the newly identified host factor NRP1 in renal carcinomas may play a vital role in the increased risk of SARS-CoV-2 infection and survivability of COVID-19 patients suffering from kidney cancers. The findings of this investigation will be helpful for further molecular studies and prevention and/or treatment strategies for COVID-19 patients associated with renal carcinomas.
Collapse
|
59
|
Zangaladze A, Cai CL, Marcelino M, Aranda JV, Beharry KD. Renal biomarkers of acute kidney injury in response to increasing intermittent hypoxia episodes in the neonatal rat. BMC Nephrol 2021; 22:299. [PMID: 34481475 PMCID: PMC8418040 DOI: 10.1186/s12882-021-02507-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Accepted: 08/19/2021] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND We tested the hypotheses that: 1) early exposure to increasing episodes of clinically relevant intermittent hypoxia (IH) is detrimental to the developing kidneys; and 2) there is a critical number of daily IH episodes which will result in irreparable renal damage that may involve angiotensin (Ang) II and endothelin (ET)-1. METHODS At birth (P0), neonatal rat pups were exposed to brief IH episodes from the first day of life (P0) to P7 or from P0-P14. Pups were either euthanized immediately or placed in room air (RA) until P21. RA littermates served as controls. Kidneys were harvested at P7, P14, and P21 for histopathology; angiotensin converting enzyme (ACE), ACE-2, ET-1, big ET-1, and malondialdehyde (MDA) levels; immunoreactivity of ACE, ACE-2, ET-1, ET-2, ET receptors (ETAR, ETBR), and hypoxia inducible factor (HIF)1α; and apoptosis (TUNEL stain). RESULTS Histopathology showed increased renal damage with 8-12 IH episodes/day, and was associated with Ang II, ACE, HIF1α, and apoptosis. ACE-2 was not expressed at P7, and minimally increased at P14. However, a robust ACE-2 response was seen during recovery with maximum levels noted in the groups recovering from 8 IH episodes/day. ET-1, big ET-1, ETAR, ETBR, and MDA increased with increasing levels of neonatal IH. CONCLUSIONS Chronic neonatal IH causes severe damage to the developing kidney with associated elevations in vasoconstrictors, suggesting hypertension, particularly with 8 neonatal IH episodes. ACE-2 is not activated in early postnatal life, and this may contribute to IH-induced vasoconstriction. Therapeutic targeting of ACE and ET-1 may help decrease the risk for kidney injury in the developing neonate to prevent and/or treat neonatal acute kidney injury and/or chronic kidney disease.
Collapse
Affiliation(s)
- Anano Zangaladze
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, State University of New York, Downstate Medical Center, Brooklyn, NY, USA
| | - Charles L Cai
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, State University of New York, Downstate Medical Center, Brooklyn, NY, USA
| | - Matthew Marcelino
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, State University of New York, Downstate Medical Center, Brooklyn, NY, USA
| | - Jacob V Aranda
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, State University of New York, Downstate Medical Center, Brooklyn, NY, USA
- Department of Ophthalmology, State University of New York, Downstate Medical Center, Brooklyn, NY, USA
- SUNY Eye Institute, New York, NY, USA
| | - Kay D Beharry
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, State University of New York, Downstate Medical Center, Brooklyn, NY, USA.
- Department of Ophthalmology, State University of New York, Downstate Medical Center, Brooklyn, NY, USA.
- SUNY Eye Institute, New York, NY, USA.
- Department of Pediatrics & Ophthalmology, Neonatal-Perinatal Medicine Clinical & Translational Research Labs, State University of New York, Downstate Medical Center, 450 Clarkson Avenue, Box 49, Brooklyn, NY, 11203, USA.
| |
Collapse
|
60
|
Azinheira Nobrega Cruz N, Gonçalves de Oliveira LC, Tedesco Silva Junior H, Osmar Medina Pestana J, Casarini DE. Angiotensin-Converting Enzyme 2 in the Pathogenesis of Renal Abnormalities Observed in COVID-19 Patients. Front Physiol 2021; 12:700220. [PMID: 34497535 PMCID: PMC8419418 DOI: 10.3389/fphys.2021.700220] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Accepted: 08/03/2021] [Indexed: 01/08/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19) was first reported in late December 2019 in Wuhan, China. The etiological agent of this disease is severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), and the high transmissibility of the virus led to its rapid global spread and a major pandemic (ongoing at the time of writing this review). The clinical manifestations of COVID-19 can vary widely from non-evident or minor symptoms to severe acute respiratory syndrome and multi-organ damage, causing death. Acute kidney injury (AKI) has been recognized as a common complication of COVID-19 and in many cases, kidney replacement therapy (KRT) is required. The presence of kidney abnormalities on hospital admission and the development of AKI are related to a more severe presentation of COVID-19 with higher mortality rate. The high transmissibility and the broad spectrum of clinical manifestations of COVID-19 are in part due to the high affinity of SARS-CoV-2 for its receptor, angiotensin (Ang)-converting enzyme 2 (ACE2), which is widely expressed in human organs and is especially abundant in the kidneys. A debate on the role of ACE2 in the infectivity and pathogenesis of COVID-19 has emerged: Does the high expression of ACE2 promotes higher infectivity and more severe clinical manifestations or does the interaction of SARS-CoV-2 with ACE2 reduce the bioavailability of the enzyme, depleting its biological activity, which is closely related to two important physiological systems, the renin-angiotensin system (RAS) and the kallikrein-kinin system (KKS), thereby further contributing to pathogenesis. In this review, we discuss the dual role of ACE2 in the infectivity and pathogenesis of COVID-19, highlighting the effects of COVID-19-induced ACE2 depletion in the renal physiology and how it may lead to kidney injury. The ACE2 downstream regulation of KKS, that usually receives less attention, is discussed. Also, a detailed discussion on how the triad of symptoms (respiratory, inflammatory, and coagulation symptoms) of COVID-19 can indirectly promote renal injury is primary aborded.
Collapse
Affiliation(s)
| | | | | | | | - Dulce Elena Casarini
- Nephrology Division, Department of Medicine, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| |
Collapse
|
61
|
Piticchio T, Le Moli R, Tumino D, Frasca F. Relationship between betacoronaviruses and the endocrine system: a new key to understand the COVID-19 pandemic-A comprehensive review. J Endocrinol Invest 2021; 44:1553-1570. [PMID: 33583003 PMCID: PMC7882054 DOI: 10.1007/s40618-020-01486-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 12/11/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND A new harmful respiratory disease, called COVID-19 emerged in China in December 2019 due to the infection of a novel coronavirus, called SARS-Coronavirus 2 (SARS-CoV-2), which belongs to the betacoronavirus genus, including SARS-CoV-1 and MERS-CoV. SARS-CoV-2 shares almost 80% of the genome with SARS-CoV-1 and 50% with MERS-CoV. Moreover, SARS-CoV-2 proteins share a high degree of homology (approximately 95%) with SARS-CoV-1 proteins. Hence, the mechanisms of SARS-Cov-1 and SARS-Cov-2 infection are similar and occur via binding to ACE2 protein, which is widely distributed in the human body, with a predominant expression in endocrine tissues including testis, thyroid, adrenal and pituitary. PURPOSE On the basis of expression pattern of the ACE2 protein among different tissues, similarity between SARS-Cov-1 and SARS-Cov-2 and the pathophysiology of COVID-19 disease, we aimed at discussing, after almost one-year pandemic, about the relationships between COVID-19 infection and the endocrine system. First, we discussed the potential effect of hormones on the susceptibility to COVID-19 infection; second, we examined the evidences regarding the effect of COVID-19 on the endocrine system. When data were available, a comparative discussion between SARS and COVID-19 effects was also performed. METHODS A comprehensive literature search within Pubmed was performed. This review has been conducted according to the PRISMA statements. RESULTS Among 450, 100 articles were selected. Tissue and vascular damages have been shown on thyroid, adrenal, testis and pituitary glands, with multiple alterations of endocrine function. CONCLUSION Hormones may affect patient susceptibility to COVID-19 infection but evidences regarding therapeutic implication of these findings are still missing. SARS and COVID-19 may affect endocrine glands and their dense vascularization, impairing endocrine system function. A possible damage of endocrine system in COVID-19 patients should be investigated in both COVID-19 acute phase and recovery to identify both early and late endocrine complications that may be important for patient's prognosis and well-being after COVID-19 infection.
Collapse
Affiliation(s)
- T Piticchio
- Endocrinology Section, Department of Clinical and Experimental Medicine, Garibaldi Nesima Hospital, University of Catania, Via Palermo 636, 95122, Catania, Italy
| | - R Le Moli
- Endocrinology Section, Department of Clinical and Experimental Medicine, Garibaldi Nesima Hospital, University of Catania, Via Palermo 636, 95122, Catania, Italy
| | - D Tumino
- Endocrinology Section, Department of Clinical and Experimental Medicine, Garibaldi Nesima Hospital, University of Catania, Via Palermo 636, 95122, Catania, Italy
| | - F Frasca
- Endocrinology Section, Department of Clinical and Experimental Medicine, Garibaldi Nesima Hospital, University of Catania, Via Palermo 636, 95122, Catania, Italy.
| |
Collapse
|
62
|
Jankowski J, Lee HK, Wilflingseder J, Hennighausen L. JAK inhibitors dampen activation of interferon-activated transcriptomes and the SARS-CoV-2 receptor ACE2 in human renal proximal tubules. iScience 2021; 24:102928. [PMID: 34345808 PMCID: PMC8321697 DOI: 10.1016/j.isci.2021.102928] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 06/02/2021] [Accepted: 07/23/2021] [Indexed: 12/17/2022] Open
Abstract
SARS-CoV-2 infections initiate cytokine storms and activate genetic programs leading to progressive hyperinflammation in multiple organs of patients with COVID-19. While it is known that COVID-19 impacts kidney function, leading to increased mortality, cytokine response of renal epithelium has not been studied in detail. Here, we report on the genetic programs activated in human primary proximal tubule (HPPT) cells by interferons and their suppression by ruxolitinib, a Janus kinase (JAK) inhibitor used in COVID-19 treatment. Integration of our data with those from patients with acute kidney injury and COVID-19, as well as other tissues, permitted the identification of kidney-specific interferon responses. Additionally, we investigated the regulation of the recently discovered isoform (dACE2) of the angiotensin-converting enzyme 2 (ACE2), the SARS-CoV-2 receptor. Using ChIP-seq, we identified candidate interferon-activated enhancers controlling the ACE2 locus, including the intronic dACE2 promoter. Taken together, our study provides an in-depth understanding of genetic programs activated in kidney cells. We provide transcriptomic and epigenetic data sets for human renal proximal tubules Cytokine stimulation induces distinct genetic pathways in the kidney Short isoform of ACE2, dACE2, is expressed in renal proximal tubules Type I interferons increase dACE2, but not full ACE2 expression
Collapse
Affiliation(s)
- Jakub Jankowski
- Laboratory of Genetics and Physiology, National Institute of Diabetes and Digestive and Kidney Diseases, U.S. National Institutes of Health, Building 8, Room 101, 8 Center Dr, Bethesda, MD 20892, USA.,Department of Physiology and Pathophysiology, University of Veterinary Medicine, Veterinärplatz 1, 1210 Vienna, Austria
| | - Hye Kyung Lee
- Laboratory of Genetics and Physiology, National Institute of Diabetes and Digestive and Kidney Diseases, U.S. National Institutes of Health, Building 8, Room 101, 8 Center Dr, Bethesda, MD 20892, USA
| | - Julia Wilflingseder
- Department of Physiology and Pathophysiology, University of Veterinary Medicine, Veterinärplatz 1, 1210 Vienna, Austria
| | - Lothar Hennighausen
- Laboratory of Genetics and Physiology, National Institute of Diabetes and Digestive and Kidney Diseases, U.S. National Institutes of Health, Building 8, Room 101, 8 Center Dr, Bethesda, MD 20892, USA
| |
Collapse
|
63
|
Sainsily X, Coquerel D, Giguère H, Dumont L, Tran K, Noll C, Ionescu AL, Côté J, Longpré JM, Carpentier A, Marsault É, Lesur O, Sarret P, Auger-Messier M. Elabela Protects Spontaneously Hypertensive Rats From Hypertension and Cardiorenal Dysfunctions Exacerbated by Dietary High-Salt Intake. Front Pharmacol 2021; 12:709467. [PMID: 34385922 PMCID: PMC8353398 DOI: 10.3389/fphar.2021.709467] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 07/14/2021] [Indexed: 12/11/2022] Open
Abstract
Objectives: Arterial hypertension, when exacerbated by excessive dietary salt intake, worsens the morbidity and mortality rates associated with cardiovascular and renal diseases. Stimulation of the apelinergic system appears to protect against several circulatory system diseases, but it remains unknown if such beneficial effects are conserved in severe hypertension. Therefore, we aimed at determining whether continuous infusion of apelinergic ligands (i.e., Apelin-13 and Elabela) exerted cardiorenal protective effects in spontaneously hypertensive (SHR) rats receiving high-salt diet. Methods: A combination of echocardiography, binding assay, histology, and biochemical approaches were used to investigate the cardiovascular and renal effects of Apelin-13 or Elabela infusion over 6 weeks in SHR fed with normal-salt or high-salt chow. Results: High-salt intake upregulated the cardiac and renal expression of APJ receptor in SHR. Importantly, Elabela was more effective than Apelin-13 in reducing high blood pressure, cardiovascular and renal dysfunctions, fibrosis and hypertrophy in high-salt fed SHR. Unlike Apelin-13, the beneficial effects of Elabela were associated with a counter-regulatory role of the ACE/ACE2/neprilysin axis of the renin-angiotensin-aldosterone system (RAAS) in heart and kidneys of salt-loaded SHR. Interestingly, Elabela also displayed higher affinity for APJ in the presence of high salt concentration and better resistance to RAAS enzymes known to cleave Apelin-13. Conclusion: These findings highlight the protective action of the apelinergic system against salt-induced severe hypertension and cardiorenal failure. As compared with Apelin-13, Elabela displays superior pharmacodynamic and pharmacokinetic properties that warrant further investigation of its therapeutic use in cardiovascular and kidney diseases.
Collapse
Affiliation(s)
- Xavier Sainsily
- Département de Médecine, Centre de Recherche du CHUS, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC, Canada.,Département de Pharmacologie et Physiologie, Centre de Recherche du CHUS, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC, Canada.,Institut de Pharmacologie de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - David Coquerel
- Département de Médecine, Centre de Recherche du CHUS, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC, Canada.,Institut de Pharmacologie de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Hugo Giguère
- Département de Médecine, Centre de Recherche du CHUS, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC, Canada.,Institut de Pharmacologie de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Lauralyne Dumont
- Département de Médecine, Centre de Recherche du CHUS, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC, Canada.,Institut de Pharmacologie de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Kien Tran
- Département de Pharmacologie et Physiologie, Centre de Recherche du CHUS, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC, Canada.,Institut de Pharmacologie de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Christophe Noll
- Département de Médecine, Centre de Recherche du CHUS, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Andrei L Ionescu
- Département de Médecine, Centre de Recherche du CHUS, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC, Canada.,Institut de Pharmacologie de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Jérôme Côté
- Département de Pharmacologie et Physiologie, Centre de Recherche du CHUS, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC, Canada.,Institut de Pharmacologie de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Jean-Michel Longpré
- Département de Pharmacologie et Physiologie, Centre de Recherche du CHUS, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC, Canada.,Institut de Pharmacologie de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - André Carpentier
- Département de Médecine, Centre de Recherche du CHUS, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Éric Marsault
- Département de Pharmacologie et Physiologie, Centre de Recherche du CHUS, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC, Canada.,Institut de Pharmacologie de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Olivier Lesur
- Département de Médecine, Centre de Recherche du CHUS, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC, Canada.,Institut de Pharmacologie de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Philippe Sarret
- Département de Pharmacologie et Physiologie, Centre de Recherche du CHUS, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC, Canada.,Institut de Pharmacologie de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Mannix Auger-Messier
- Département de Médecine, Centre de Recherche du CHUS, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC, Canada.,Institut de Pharmacologie de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada
| |
Collapse
|
64
|
Gupta K, Bhurwal A, Law C, Ventre S, Minacapelli CD, Kabaria S, Li Y, Tait C, Catalano C, Rustgi VK. Acute kidney injury and hepatorenal syndrome in cirrhosis. World J Gastroenterol 2021; 27:3984-4003. [PMID: 34326609 PMCID: PMC8311533 DOI: 10.3748/wjg.v27.i26.3984] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 03/19/2021] [Accepted: 06/22/2021] [Indexed: 02/06/2023] Open
Abstract
Acute kidney injury (AKI) in cirrhosis, including hepatorenal syndrome (HRS), is a common and serious complication in cirrhotic patients, leading to significant morbidity and mortality. AKI is separated into two categories, non-HRS AKI and HRS-AKI. The most recent definition and diagnostic criteria of AKI in cirrhosis and HRS have helped diagnose and prognosticate the disease. The pathophysiology behind non-HRS-AKI and HRS is more complicated than once theorized and involves more processes than just splanchnic vasodilation. The common biomarkers clinicians use to assess kidney injury have significant limitations in cirrhosis patients; novel biomarkers being studied have shown promise but require further studies in clinical settings and animal models. The overall management of non-HRS AKI and HRS-AKI requires a systematic approach. Although pharmacological treatments have shown mortality benefit, the ideal HRS treatment option is liver transplantation with or without simultaneous kidney transplantation. Further research is required to optimize pharmacologic and nonpharmacologic approaches to treatment. This article reviews the current guidelines and recommendations of AKI in cirrhosis.
Collapse
Affiliation(s)
- Kapil Gupta
- Division of Gastroenterology and Hepatology, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ 08901, United States
| | - Abhishek Bhurwal
- Division of Gastroenterology and Hepatology, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ 08901, United States
| | - Cindy Law
- Division of Gastroenterology and Hepatology, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ 08901, United States
| | - Scott Ventre
- Department of Internal Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ 08901, United States
| | - Carlos D Minacapelli
- Division of Gastroenterology and Hepatology, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ 08901, United States
| | - Savan Kabaria
- Department of Internal Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ 08901, United States
| | - You Li
- Division of Gastroenterology and Hepatology, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ 08901, United States
| | - Christopher Tait
- Division of Gastroenterology and Hepatology, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ 08901, United States
| | - Carolyn Catalano
- Division of Gastroenterology and Hepatology, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ 08901, United States
| | - Vinod K Rustgi
- Division of Gastroenterology and Hepatology, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ 08901, United States
| |
Collapse
|
65
|
Refaie AA, Shalby AB, Kassem SM, Khalil WKB. DNA Damage and Expression Profile of Genes Associated with Nephrotoxicity Induced by Butralin and Ameliorating Effect of Arabic Gum in Female Rats. Appl Biochem Biotechnol 2021; 193:3454-3468. [PMID: 34240313 DOI: 10.1007/s12010-021-03607-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Accepted: 06/21/2021] [Indexed: 12/16/2022]
Abstract
Nephrotoxicity induced by exposure to environmental pollution, including herbicides, is becoming a global problem. Natural products are the prime alternative scientific research as they express better medicinal activity and minor side effects compared with a variety of synthetic drugs. This study was performed to evaluate the nephroprotective proficiency of Arabic gum against butralin-induced nephrotoxicity. Adult female rats were supplemented with Arabic gum (4.3 g/kg b.wt) and/or butralin (312 mg/L) in drinking water for 30 days. The results found that markers of serum kidney function, oxidative stress biomarkers, DNA damage, and expression of kidney specific genes (Acsm2, Ace, and Ace2) as well as histopathological examination in treated rats were conducted. Butralin-treated rats showed a rise in serum creatinine (41%), BUN (47.3%), and MDA (140.9%) as well as decrease in activity of the antioxidant markers (CAT (-21%); GPx (-70.7%); and TAC (43.2%)) in comparison with the control group. In addition, butralin treatment increased the DNA damage (221%); altered the expression levels of Acsm2, Ace, and Ace2 (-51.6%, 141.6%, and 143% respectively); and elevated histopathological lesions in the kidney tissues. Pretreatment of Arabic gum prevented butralin-prompted degenerative changes of kidney tissues. The results suggested that the protective effect provided by Arabic gum on renal tissues exposed to the herbicide butralin could be attributed to enhancement of antioxidants and increase the free radical scavenging activity in vivo.
Collapse
Affiliation(s)
- Amel A Refaie
- Environmental Toxicology Research Unit (ETRU), Pesticide Chemistry Department, National Research Centre (NRC), 33 El Bohouth Street, P.O. 12622, Dokki, Giza, Egypt
| | - Aziza B Shalby
- Department of Hormones, National Research Centre (NRC), 33 El Bohouth Street, P.O. 12622, Dokki, Giza, Egypt.
| | - Salwa M Kassem
- Department of Cell Biology, National Research Centre, 33-Bohouth St., P.O. 12622, Dokki, Giza, Egypt
| | - Wagdy K B Khalil
- Department of Cell Biology, National Research Centre, 33-Bohouth St., P.O. 12622, Dokki, Giza, Egypt
| |
Collapse
|
66
|
Shimmel A, Shaikhouni S, Mariani L. Current Understanding of Clinical Manifestations of COVID-19 in Glomerular Disease. GLOMERULAR DISEASES 2021; 1:250-264. [PMID: 36747902 PMCID: PMC8450860 DOI: 10.1159/000518276] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 06/26/2021] [Indexed: 12/15/2022]
Abstract
Background The novel coronavirus disease (COVID-19), also known as severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), is an evolving pandemic with significant mortality. Information about the impact of infection on glomerular disease patients in particular has been lacking. Understanding the virus's effect in glomerular disease is constantly changing. This review article summarizes the data published thus far on COVID-19 and its manifestations in pre-existing and de novo glomerular disease. Summary While patients with glomerular disease may be at higher risk of severe COVID-19 due to their immunosuppressed status, some data suggest that a low amount of immunosuppression may be helpful in mitigating the systemic inflammatory response which is associated with high mortality rates in COVID-19. There have been a few case reports on COVID-19 causing glomerular disease relapse in patients. Multiple mechanisms have been proposed for kidney injury, proteinuria, and hematuria in the setting of COVID-19. More commonly, these are caused by direct tubular injury due to hemodynamic instability and hypoxic injury. However, the cytokine storm induced by COVID-19 may trigger common post-viral glomerular disease such as IgA nephropathy, anti-GBM, and ANCA vasculitis that have also been described in COVID-19 patients. Collapsing glomerulopathy, a hallmark of HIV-associated nephropathy, is being reported SARS-CoV-2 cases, particularly in patients with high-risk APOL1 alleles. Direct viral invasion of glomerular structures is hypothesized to cause a podocytopathy due to virus's affinity to ACE2, but evidence for this remains under study. Key Messages Infection with SARS-CoV-2 may cause glomerular disease in certain patients. The mechanism of de novo glomerular disease in the setting of COVID-19 is under study. The management of patients with existing glomerular disease poses unique challenges, especially with regard to immunosuppression management. Further studies are needed to inform clinician decisions about the management of these patients during the COVID-19 pandemic.
Collapse
Affiliation(s)
- Allison Shimmel
- College of Medicine and Life Sciences, University of Toledo, Toledo, Ohio, USA
| | - Salma Shaikhouni
- Department of Nephrology, Michigan Medicine, Ann Arbor, Michigan, USA
| | - Laura Mariani
- Department of Nephrology, Michigan Medicine, Ann Arbor, Michigan, USA,*Laura Mariani,
| |
Collapse
|
67
|
Smarz-Widelska I, Grywalska E, Morawska I, Forma A, Michalski A, Mertowski S, Hrynkiewicz R, Niedźwiedzka-Rystwej P, Korona-Glowniak I, Parczewski M, Załuska W. Pathophysiology and Clinical Manifestations of COVID-19-Related Acute Kidney Injury-The Current State of Knowledge and Future Perspectives. Int J Mol Sci 2021; 22:7082. [PMID: 34209289 PMCID: PMC8268979 DOI: 10.3390/ijms22137082] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 05/26/2021] [Accepted: 06/28/2021] [Indexed: 01/08/2023] Open
Abstract
The continually evolving severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic has resulted in a vast number of either acute or chronic medical impairments of a pathophysiology that is not yet fully understood. SARS-CoV-2 tropism for the organs is associated with bilateral organ cross-talks as well as targeted dysfunctions, among which acute kidney injury (AKI) seems to be highly prevalent in infected patients. The need for efficient management of COVID-related AKI patients is an aspect that is still being investigated by nephrologists; however, another reason for concern is a disturbingly high proportion of various types of kidney dysfunctions in patients who have recovered from COVID-19. Even though the clinical picture of AKI and COVID-related AKI seems to be quite similar, it must be considered that regarding the latter, little is known about both the optimal management and long-term consequences. These discrepancies raise an urgent need for further research aimed at evaluating the molecular mechanisms associated with SARS-CoV-2-induced kidney damage as well as standardized management of COVID-related AKI patients. The following review presents a comprehensive and most-recent insight into the pathophysiology, clinical manifestations, recommended patient management, treatment strategies, and post-mortem findings in patients with COVID-related AKI.
Collapse
Affiliation(s)
- Iwona Smarz-Widelska
- Department of Nephrology, Cardinal Stefan Wyszynski Provincial Hospital in Lublin, 20-718 Lublin, Poland;
| | - Ewelina Grywalska
- Department of Clinical Immunology and Immunotherapy, Medical University of Lublin, 20-093 Lublin, Poland; (I.M.); (A.M.); (S.M.)
| | - Izabela Morawska
- Department of Clinical Immunology and Immunotherapy, Medical University of Lublin, 20-093 Lublin, Poland; (I.M.); (A.M.); (S.M.)
| | - Alicja Forma
- Department of Forensic Medicine, Medical University of Lublin, 20-090 Lublin, Poland;
| | - Adam Michalski
- Department of Clinical Immunology and Immunotherapy, Medical University of Lublin, 20-093 Lublin, Poland; (I.M.); (A.M.); (S.M.)
| | - Sebastian Mertowski
- Department of Clinical Immunology and Immunotherapy, Medical University of Lublin, 20-093 Lublin, Poland; (I.M.); (A.M.); (S.M.)
| | - Rafał Hrynkiewicz
- Institute of Biology, University of Szczecin, 71-412 Szczecin, Poland;
| | | | - Izabela Korona-Glowniak
- Department of Pharmaceutical Microbiology, Medical University of Lublin, Chodźki 1, 20-093 Lublin, Poland;
| | - Miłosz Parczewski
- Department of Infectious, Tropical Diseases and Immune Deficiency, Pomeranian Medical University in Szczecin, 71-455 Szczecin, Poland;
| | - Wojciech Załuska
- Department of Nephrology, Medical University of Lublin, 20-954 Lublin, Poland;
| |
Collapse
|
68
|
Merarchi M, Dudha N, Das BC, Garg M. Natural products and phytochemicals as potential anti-SARS-CoV-2 drugs. Phytother Res 2021; 35:5384-5396. [PMID: 34132421 PMCID: PMC8441929 DOI: 10.1002/ptr.7151] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 04/16/2021] [Accepted: 04/22/2021] [Indexed: 12/12/2022]
Abstract
The current pandemic responsible for the crippling of the health care system is caused by the novel SARS‐CoV‐2 in 2019 and leading to coronavirus disease 2019 (COVID‐19). The virus enters into humans by attachment of its Spike protein (S) to the ACE receptor present on the lung epithelial cell surface followed by cleavage of S protein by the cellular transmembrane serine protease (TMPRSS2). After entry, the SARS‐CoV‐2 RNA genome is released into the cytosol, where it highjacks host replication machinery for viral replication, assemblage, as well as the release of new viral particles. The major drug targets that have been identified for SARS‐CoV‐2 through host‐virus interaction studies include 3CLpro, PLpro, RNA‐dependent RNA polymerase, and S proteins. Several reports of natural compounds along with synthetic products have displayed promising results and some of them are Tripterygium wilfordii, Pudilan Xiaoyan Oral Liquid, Saponin derivates, Artemisia annua, Glycyrrhiza glabra L., Jinhua Qinggan granules, Xuebijing, and Propolis. This review attempts to disclose the natural products identified as anti‐SARS‐CoV‐2 based on in silico prediction and the effect of a variety of phytochemicals either alone and/or in combination with conventional treatments along with their possible molecular mechanisms involved for both prevention and treatment of the SARS‐CoV‐2 disease.
Collapse
Affiliation(s)
- Myriam Merarchi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Namrata Dudha
- Department of Biotechnology and Microbiology, School of Sciences, Noida International University, Gautam Budh Nagar, India
| | - Bhudev C Das
- Amity Institute of Molecular Medicine and Stem cell Research (AIMMSCR), Amity University Uttar Pradesh, Noida, India
| | - Manoj Garg
- Amity Institute of Molecular Medicine and Stem cell Research (AIMMSCR), Amity University Uttar Pradesh, Noida, India
| |
Collapse
|
69
|
Liu LP, Zhang XL, Li J. New perspectives on angiotensin-converting enzyme 2 and its related diseases. World J Diabetes 2021; 12:839-854. [PMID: 34168732 PMCID: PMC8192247 DOI: 10.4239/wjd.v12.i6.839] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 03/30/2021] [Accepted: 04/21/2021] [Indexed: 02/06/2023] Open
Abstract
Since the worldwide outbreak of coronavirus disease 2019, angiotensin-converting enzyme 2 (ACE2) has received widespread attention as the cell receptor of the severe acute respiratory syndrome coronavirus 2 virus. At the same time, as a key enzyme in the renin-angiotensin-system, ACE2 is considered to be an endogenous negative regulator of vasoconstriction, proliferation, fibrosis, and proinflammation caused by the ACE-angiotensin II-angiotensin type 1 receptor axis. ACE2 is now implicated as being closely connected to diabetes, cardiovascular, kidney, and lung diseases, and so on. This review covers the available information on the host factors regulating ACE2 and discusses its role in a variety of pathophysiological conditions in animal models and humans.
Collapse
Affiliation(s)
- Li-Ping Liu
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, School of Medicine, Hunan Normal University, Changsha 410013, Hunan Province, China
| | - Xiao-Li Zhang
- TheFifth Department of Medicine (Nephrology/Endocrinology/Rheumatology), University Medical Centre Mannheim, University of Heidelberg, Heidelberg 68135, Baden-Württemberg, Germany
| | - Jian Li
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, School of Medicine, Hunan Normal University, Changsha 410013, Hunan Province, China
| |
Collapse
|
70
|
Giani JF, Veiras LC, Shen JZY, Bernstein EA, Cao D, Okwan-Duodu D, Khan Z, Gonzalez-Villalobos RA, Bernstein KE. Novel roles of the renal angiotensin-converting enzyme. Mol Cell Endocrinol 2021; 529:111257. [PMID: 33781839 PMCID: PMC8127398 DOI: 10.1016/j.mce.2021.111257] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 02/03/2021] [Accepted: 03/20/2021] [Indexed: 12/14/2022]
Abstract
The observation that all components of the renin angiotensin system (RAS) are expressed in the kidney and the fact that intratubular angiotensin (Ang) II levels greatly exceed the plasma concentration suggest that the synthesis of renal Ang II occurs independently of the circulating RAS. One of the main components of this so-called intrarenal RAS is angiotensin-converting enzyme (ACE). Although the role of ACE in renal disease is demonstrated by the therapeutic effectiveness of ACE inhibitors in treating several conditions, the exact contribution of intrarenal versus systemic ACE in renal disease remains unknown. Using genetically modified mouse models, our group demonstrated that renal ACE plays a key role in the development of several forms of hypertension. Specifically, although ACE is expressed in different cell types within the kidney, its expression in renal proximal tubular cells is essential for the development of high blood pressure. Besides hypertension, ACE is involved in several other renal diseases such as diabetic kidney disease, or acute kidney injury even when blood pressure is normal. In addition, studies suggest that ACE might mediate at least part of its effect through mechanisms that are independent of the Ang I conversion into Ang II and involve other substrates such as N-acetyl-seryl-aspartyl-lysyl-proline (AcSDKP), Ang-(1-7), and bradykinin, among others. In this review, we summarize the recent advances in understanding the contribution of intrarenal ACE to different pathological conditions and provide insight into the many roles of ACE besides the well-known synthesis of Ang II.
Collapse
Affiliation(s)
- Jorge F Giani
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA; Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
| | - Luciana C Veiras
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Justin Z Y Shen
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Ellen A Bernstein
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - DuoYao Cao
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Derick Okwan-Duodu
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Zakir Khan
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA; Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | | | - Kenneth E Bernstein
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA; Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| |
Collapse
|
71
|
Mülling N, Rohn H. Angiotensinkonvertierendes Enzym 2 (ACE2): Rolle in der Pathogenese von Erkrankungen außerhalb von COVID-19. DER NEPHROLOGE 2021; 16:185-188. [PMID: 33868494 PMCID: PMC8034869 DOI: 10.1007/s11560-021-00507-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Accepted: 03/31/2021] [Indexed: 12/02/2022]
Affiliation(s)
- N. Mülling
- Klinik für Nephrologie, Universitätsklinikum Essen, Universität Duisburg-Essen, Hufelandstraße 55, 45122 Essen, Deutschland
| | - H. Rohn
- Klinik für Infektiologie, Westdeutsches Zentrum für Infektiologie, Universitätsklinikum Essen, Universität Duisburg-Essen, Essen, Deutschland
| |
Collapse
|
72
|
Beyerstedt S, Casaro EB, Rangel ÉB. COVID-19: angiotensin-converting enzyme 2 (ACE2) expression and tissue susceptibility to SARS-CoV-2 infection. Eur J Clin Microbiol Infect Dis 2021; 40:905-919. [PMID: 33389262 PMCID: PMC7778857 DOI: 10.1007/s10096-020-04138-6] [Citation(s) in RCA: 481] [Impact Index Per Article: 120.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Accepted: 12/20/2020] [Indexed: 02/06/2023]
Abstract
COVID-19 pandemic is caused by the novel coronavirus SARS-CoV-2. Angiotensin-converting enzyme 2 (ACE2) is not only an enzyme but also a functional receptor on cell surfaces through which SARS-CoV-2 enters the host cells and is highly expressed in the heart, kidneys, and lungs and shed into the plasma. ACE2 is a key regulator of the renin-angiotensin-aldosterone system (RAAS). SARS-CoV-2 causes ACE/ACE2 balance disruption and RAAS activation, which leads ultimately to COVID-19 progression, especially in patients with comorbidities, such as hypertension, diabetes mellitus, and cardiovascular disease. Therefore, ACE2 expression may have paradoxical effects, aiding SARS-CoV-2 pathogenicity, yet conversely limiting viral infection. This article reviews the existing literature and knowledge of ACE2 in COVID-19 setting and focuses on its pathophysiologic involvement in disease progression, clinical outcomes, and therapeutic potential.
Collapse
Affiliation(s)
- Stephany Beyerstedt
- Albert Einstein Research and Education Institute, Hospital Israelita Albert Einstein, 627 Albert Einstein Avenue, Building A, Morumbi, São Paulo, SP, Brazil
| | - Expedito Barbosa Casaro
- Albert Einstein Research and Education Institute, Hospital Israelita Albert Einstein, 627 Albert Einstein Avenue, Building A, Morumbi, São Paulo, SP, Brazil
| | - Érika Bevilaqua Rangel
- Albert Einstein Research and Education Institute, Hospital Israelita Albert Einstein, 627 Albert Einstein Avenue, Building A, Morumbi, São Paulo, SP, Brazil.
- Nephrology Division, Federal University of São Paulo, São Paulo, SP, Brazil.
| |
Collapse
|
73
|
You J, Seok JH, Joo M, Bae JY, Kim JI, Park MS, Kim K. Multifactorial Traits of SARS-CoV-2 Cell Entry Related to Diverse Host Proteases and Proteins. Biomol Ther (Seoul) 2021; 29:249-262. [PMID: 33875625 PMCID: PMC8094071 DOI: 10.4062/biomolther.2021.048] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 03/26/2021] [Accepted: 03/29/2021] [Indexed: 11/05/2022] Open
Abstract
The most effective way to control newly emerging infectious disease, such as the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic, is to strengthen preventative or therapeutic public health strategies before the infection spreads worldwide. However, global health systems remain at the early stages in anticipating effective therapeutics or vaccines to combat the SARS-CoV-2 pandemic. While maintaining social distance is the most crucial metric to avoid spreading the virus, symptomatic therapy given to patients on the clinical manifestations helps save lives. The molecular properties of SARS-CoV-2 infection have been quickly elucidated, paving the way to therapeutics, vaccine development, and other medical interventions. Despite this progress, the detailed biomolecular mechanism of SARS-CoV-2 infection remains elusive. Given virus invasion of cells is a determining factor for virulence, understanding the viral entry process can be a mainstay in controlling newly emerged viruses. Since viral entry is mediated by selective cellular proteases or proteins associated with receptors, identification and functional analysis of these proteins could provide a way to disrupt virus propagation. This review comprehensively discusses cellular machinery necessary for SARS-CoV-2 infection. Understanding multifactorial traits of the virus entry will provide a substantial guide to facilitate antiviral drug development.
Collapse
Affiliation(s)
- Jaehwan You
- Department of Microbiology, Institute for Viral Diseases, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - Jong Hyeon Seok
- Department of Microbiology, Institute for Viral Diseases, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - Myungsoo Joo
- School of Korean Medicine, Pusan National University, Pusan 50612, Republic of Korea
| | - Joon-Yong Bae
- Department of Microbiology, Institute for Viral Diseases, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - Jin Il Kim
- Department of Microbiology, Institute for Viral Diseases, Korea University College of Medicine, Seoul 02841, Republic of Korea
- Biosafety Center, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - Man-Seong Park
- Department of Microbiology, Institute for Viral Diseases, Korea University College of Medicine, Seoul 02841, Republic of Korea
- Biosafety Center, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - Kisoon Kim
- Department of Microbiology, Institute for Viral Diseases, Korea University College of Medicine, Seoul 02841, Republic of Korea
| |
Collapse
|
74
|
Luzes R, Muzi-Filho H, Pereira-Acácio A, Crisóstomo T, Vieyra A. Angiotensin-(3-4) modulates the overweight- and undernutrition-induced ACE2 downregulation in renal proximal tubule cells: implications for COVID-19? EXPLORATION OF MEDICINE 2021. [DOI: 10.37349/emed.2021.00038] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Aim: The renal lesions–including severe acute kidney injury–are severe outcomes in severe acute respiratory syndrome coronavirus 2 infections. There are no reports regarding the influence of the nutritional status on the severity and progress of these lesions. Ageing is also an important risk factor.
Methods: In the present study we compared the influence of overweight and undernutrition on the levels of renal angiotensin converting enzymes 1 and 2 (ACE and ACE2), which were evaluated by Western blotting. Since the renin-angiotensin-aldosterone system (RAAS) has been implicated in the progress of kidney failure during coronavirus disease 2019, the influence of Angiotensin-(3-4) [Ang-(3-4)] was investigated. Ang-(3-4) is the shortest angiotensin-derived peptide, which is considered the physiological antagonist of several Ang II effects.
Results: Both overweight and undernutrition downregulate the levels of ACE2 without influence on the levels of ACE in proximal tubules from kidney rats. Administration of Ang-(3-4) upregulates ACE2 to levels above the control in overweight but not in undernourished rats.
Conclusions: Chronic undernourishment and overnourishment conditions play a central role in the renal ACE/ACE2 balance, and that the role of RAAS is also different in overweight and undernutrition.
Collapse
Affiliation(s)
- Rafael Luzes
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, 21941-170 Rio de Janeiro, Brazil 2Graduate Program of Translational Biomedicine/BIOTRANS, Unigranrio University, 25071-202 Duque de Caxias, Brazil 3National Center of Structural Biology and Bioimaging/CENABIO, Federal University of Rio de Janeiro, 21941-902 Rio de Janeiro, Brazil
| | - Humberto Muzi-Filho
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, 21941-170 Rio de Janeiro, Brazil 3National Center of Structural Biology and Bioimaging/CENABIO, Federal University of Rio de Janeiro, 21941-902 Rio de Janeiro, Brazil
| | - Amaury Pereira-Acácio
- Graduate Program of Translational Biomedicine/BIOTRANS, Unigranrio University, 25071-202 Duque de Caxias, Brazil 3National Center of Structural Biology and Bioimaging/CENABIO, Federal University of Rio de Janeiro, 21941-902 Rio de Janeiro, Brazil
| | - Thuany Crisóstomo
- National Center of Structural Biology and Bioimaging/CENABIO, Federal University of Rio de Janeiro, 21941-902 Rio de Janeiro, Brazil 4Leopoldo de Meis Institute of Medical Biochemistry, Federal University of Rio de Janeiro, 21941-902 Rio de Janeiro, Brazil
| | - Adalberto Vieyra
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, 21941-170 Rio de Janeiro, Brazil 2Graduate Program of Translational Biomedicine/BIOTRANS, Unigranrio University, 25071-202 Duque de Caxias, Brazil 3National Center of Structural Biology and Bioimaging/CENABIO, Federal University of Rio de Janeiro, 21941-902 Rio de Janeiro, Brazil
| |
Collapse
|
75
|
Lin W, Fan J, Hu LF, Zhang Y, Ooi JD, Meng T, Jin P, Ding X, Peng LK, Song L, Tang R, Xiao Z, Ao X, Xiao XC, Zhou QL, Xiao P, Zhong Y. Single-cell analysis of angiotensin-converting enzyme II expression in human kidneys and bladders reveals a potential route of 2019 novel coronavirus infection. Chin Med J (Engl) 2021; 134:935-943. [PMID: 33879756 PMCID: PMC8078266 DOI: 10.1097/cm9.0000000000001439] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Since 2019, a novel coronavirus named 2019 novel coronavirus (2019-nCoV) has emerged worldwide. Apart from fever and respiratory complications, acute kidney injury has been observed in a few patients with coronavirus disease 2019. Furthermore, according to recent findings, the virus has been detected in urine. Angiotensin-converting enzyme II (ACE2) has been proposed to serve as the receptor for the entry of 2019-nCoV, which is the same as that for the severe acute respiratory syndrome. This study aimed to investigate the possible cause of kidney damage and the potential route of 2019-nCoV infection in the urinary system. METHODS We used both published kidney and bladder cell atlas data and new independent kidney single-cell RNA sequencing data generated in-house to evaluate ACE2 gene expression in all cell types in healthy kidneys and bladders. The Pearson correlation coefficients between ACE2 and all other genes were first generated. Then, genes with r values larger than 0.1 and P values smaller than 0.01 were deemed significant co-expression genes with ACE2. RESULTS Our results showed the enriched expression of ACE2 in all subtypes of proximal tubule (PT) cells of the kidney. ACE2 expression was found in 5.12%, 5.80%, and 14.38% of the proximal convoluted tubule cells, PT cells, and proximal straight tubule cells, respectively, in three published kidney cell atlas datasets. In addition, ACE2 expression was also confirmed in 12.05%, 6.80%, and 10.20% of cells of the proximal convoluted tubule, PT, and proximal straight tubule, respectively, in our own two healthy kidney samples. For the analysis of public data from three bladder samples, ACE2 expression was low but detectable in bladder epithelial cells. Only 0.25% and 1.28% of intermediate cells and umbrella cells, respectively, had ACE2 expression. CONCLUSION This study has provided bioinformatics evidence of the potential route of 2019-nCoV infection in the urinary system.
Collapse
Affiliation(s)
- Wei Lin
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Jue Fan
- Department of Bioinformatics and Data Science, Singleron Biotechnologies, Nanjing, Jiangsu 210032, China
| | - Long-Fei Hu
- Department of Bioinformatics and Data Science, Singleron Biotechnologies, Nanjing, Jiangsu 210032, China
| | - Yan Zhang
- Department of Bioinformatics and Data Science, Singleron Biotechnologies, Nanjing, Jiangsu 210032, China
| | - Joshua D. Ooi
- Centre for Inflammatory Diseases, Monash University Department of Medicine, Monash Medical Centre, Clayton, VIC 3168, Australia
| | - Ting Meng
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Peng Jin
- Department of Organ Transplantation, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Xiang Ding
- Department of Organ Transplantation, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Long-Kai Peng
- Department of Kidney Transplantation, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Lei Song
- Department of Kidney Transplantation, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Rong Tang
- Department of Kidney Transplantation, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Zhou Xiao
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Xiang Ao
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Xiang-Cheng Xiao
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Qiao-Ling Zhou
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Ping Xiao
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Yong Zhong
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| |
Collapse
|
76
|
Chen J, Wang W, Tang Y, Huang XR, Yu X, Lan HY. Inflammatory stress in SARS-COV-2 associated Acute Kidney Injury. Int J Biol Sci 2021; 17:1497-1506. [PMID: 33907513 PMCID: PMC8071761 DOI: 10.7150/ijbs.58791] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 03/01/2021] [Indexed: 01/08/2023] Open
Abstract
Increasing clinical evidence shows that acute kidney injury (AKI) is a common and severe complication in critically ill COVID-19 patients. The older age, the severity of COVID-19 infection, the ethnicity, and the history of smoking, diabetes, hypertension, and cardiovascular disease are the risk factor for AKI in COVID-19 patients. Of them, inflammation may be a key player in the pathogenesis of AKI in patients with COVID-19. It is highly possible that SARS-COV-2 infection may trigger the activation of multiple inflammatory pathways including angiotensin II, cytokine storm such as interleukin-6 (IL-6), C-reactive protein (CRP), TGF-β signaling, complement activation, and lung-kidney crosstalk to cause AKI. Thus, treatments by targeting these inflammatory molecules and pathways with a monoclonal antibody against IL-6 (Tocilizumab), C3 inhibitor AMY-101, anti-C5 antibody, anti-TGF-β OT-101, and the use of CRRT in critically ill patients may represent as novel and specific therapies for AKI in COVID-19 patients.
Collapse
Affiliation(s)
- Junzhe Chen
- Departments of Medicine & Therapeutics, Li Ka Shing Institute of Health Sciences, and Lui Che Woo Institute of Innovative Medicine, The Chinese University of Hong Kong, Hong Kong, China
- Department of Nephrology, The Third Affiliated hospital, Southern Medical university, Guangzhou, China
| | - Wenbiao Wang
- Departments of Medicine & Therapeutics, Li Ka Shing Institute of Health Sciences, and Lui Che Woo Institute of Innovative Medicine, The Chinese University of Hong Kong, Hong Kong, China
- Guangdong Key Laboratory of Virology, Institute of Medical Microbiology, Jinan University, Guangzhou, China
| | - Ying Tang
- Department of Nephrology, The Third Affiliated hospital, Southern Medical university, Guangzhou, China
| | - Xiao-ru Huang
- Departments of Medicine & Therapeutics, Li Ka Shing Institute of Health Sciences, and Lui Che Woo Institute of Innovative Medicine, The Chinese University of Hong Kong, Hong Kong, China
- Guangdong-Hong Kong Joint Laboratory for Immunity and Genetics of Chronic Kidney Disease, Guangdong Academy of Medical Science, Guangdong Provincial People's Hospital, Guangzhou, China
| | - Xueqing Yu
- Guangdong-Hong Kong Joint Laboratory for Immunity and Genetics of Chronic Kidney Disease, Guangdong Academy of Medical Science, Guangdong Provincial People's Hospital, Guangzhou, China
| | - Hui-Yao Lan
- Departments of Medicine & Therapeutics, Li Ka Shing Institute of Health Sciences, and Lui Che Woo Institute of Innovative Medicine, The Chinese University of Hong Kong, Hong Kong, China
- Guangdong-Hong Kong Joint Laboratory for Immunity and Genetics of Chronic Kidney Disease, The Chinese University of Hong Kong, Hong Kong, China
| |
Collapse
|
77
|
Chen Y, Ouyang L, Bao FS, Li Q, Han L, Zhang H, Zhu B, Ge Y, Robinson P, Xu M, Liu J, Chen S. A Multimodality Machine Learning Approach to Differentiate Severe and Nonsevere COVID-19: Model Development and Validation. J Med Internet Res 2021; 23:e23948. [PMID: 33714935 PMCID: PMC8030658 DOI: 10.2196/23948] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Revised: 10/11/2020] [Accepted: 03/11/2021] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND Effectively and efficiently diagnosing patients who have COVID-19 with the accurate clinical type of the disease is essential to achieve optimal outcomes for the patients as well as to reduce the risk of overloading the health care system. Currently, severe and nonsevere COVID-19 types are differentiated by only a few features, which do not comprehensively characterize the complicated pathological, physiological, and immunological responses to SARS-CoV-2 infection in the different disease types. In addition, these type-defining features may not be readily testable at the time of diagnosis. OBJECTIVE In this study, we aimed to use a machine learning approach to understand COVID-19 more comprehensively, accurately differentiate severe and nonsevere COVID-19 clinical types based on multiple medical features, and provide reliable predictions of the clinical type of the disease. METHODS For this study, we recruited 214 confirmed patients with nonsevere COVID-19 and 148 patients with severe COVID-19. The clinical characteristics (26 features) and laboratory test results (26 features) upon admission were acquired as two input modalities. Exploratory analyses demonstrated that these features differed substantially between two clinical types. Machine learning random forest models based on all the features in each modality as well as on the top 5 features in each modality combined were developed and validated to differentiate COVID-19 clinical types. RESULTS Using clinical and laboratory results independently as input, the random forest models achieved >90% and >95% predictive accuracy, respectively. The importance scores of the input features were further evaluated, and the top 5 features from each modality were identified (age, hypertension, cardiovascular disease, gender, and diabetes for the clinical features modality, and dimerized plasmin fragment D, high sensitivity troponin I, absolute neutrophil count, interleukin 6, and lactate dehydrogenase for the laboratory testing modality, in descending order). Using these top 10 multimodal features as the only input instead of all 52 features combined, the random forest model was able to achieve 97% predictive accuracy. CONCLUSIONS Our findings shed light on how the human body reacts to SARS-CoV-2 infection as a unit and provide insights on effectively evaluating the disease severity of patients with COVID-19 based on more common medical features when gold standard features are not available. We suggest that clinical information can be used as an initial screening tool for self-evaluation and triage, while laboratory test results should be applied when accuracy is the priority.
Collapse
Affiliation(s)
- Yuanfang Chen
- Public Health Research Institute of Jiangsu Province, Nanjing, China
- Institute of HIV/AIDS/STI Prevention and Control, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, China
| | - Liu Ouyang
- Department of Orthopaedics, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Forrest S Bao
- Department of Computer Science, Iowa State University, Ames, IA, United States
| | - Qian Li
- Department of Pediatrics, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, China
| | - Lei Han
- Public Health Research Institute of Jiangsu Province, Nanjing, China
- Department of Occupational Disease Prevention, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, China
| | - Hengdong Zhang
- Public Health Research Institute of Jiangsu Province, Nanjing, China
- Department of Occupational Disease Prevention, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, China
| | - Baoli Zhu
- Public Health Research Institute of Jiangsu Province, Nanjing, China
- Department of Occupational Disease Prevention, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, China
- School of Public health, Nanjing Medical University, Nanjing, China
| | - Yaorong Ge
- Department of Software and Information Systems, University of North Carolina at Charlotte, Charlotte, NC, United States
| | - Patrick Robinson
- Department of Public Health Sciences, University of North Carolina at Charlotte, Charlotte, NC, United States
| | - Ming Xu
- Public Health Research Institute of Jiangsu Province, Nanjing, China
- Department of Occupational Disease Prevention, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, China
- Department of Public Health Sciences, University of North Carolina at Charlotte, Charlotte, NC, United States
| | - Jie Liu
- Department of Radiology, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Shi Chen
- Department of Public Health Sciences, University of North Carolina at Charlotte, Charlotte, NC, United States
- School of Data Science, University of North Carolina at Charlotte, Charlotte, NC, United States
| |
Collapse
|
78
|
Chong WH, Saha BK. Relationship Between Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) and the Etiology of Acute Kidney Injury (AKI). Am J Med Sci 2021; 361:287-296. [PMID: 33358501 PMCID: PMC7590839 DOI: 10.1016/j.amjms.2020.10.025] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 10/23/2020] [Indexed: 01/08/2023]
Abstract
Coronavirus disease 2019 (COVID-19) is caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Since it was first recognized in December 2019, it has resulted in the ongoing worldwide pandemic. Although acute hypoxic respiratory failure (AHRF) and acute respiratory distress syndrome (ARDS) are the main features of the disease, the involvement of other organs needs to be explored. There has been a growing concern regarding the association between acute kidney injury (AKI) and poor outcomes in SARS-CoV-2 patients. Based on current observational data, AKI is the 2nd most common cause of morbidity and mortality behind ARDS in SARS-CoV-2 patients. Angiotensin-converting enzyme 2 (ACE2) receptor has been shown to be the cornerstone of SARS-CoV-2 infection and possibly plays a significant role in the occurrence of renal injury. The pathogenesis of AKI is likely multifactorial that involves not only direct viral invasion but also dysregulated immune response in the form of cytokine storm, ischemia to kidneys, hypercoagulable state, and rhabdomyolysis, among others. We performed a literature search of the Pubmed and Google Scholar database from 1996 to 2020 using the following keywords: severe acute respiratory syndrome coronavirus 2, coronavirus disease 2019, angiotensin-converting enzyme 2 receptor, and acute kidney injury to find the most pertinent and highest-quality of evidence. Any cited references were reviewed to identify relevant literature. The purpose of this review is to discuss, explore, and summarize the relationship between AKI in SARS-CoV-2 patients, with a focus on its epidemiology, association with ACE2 receptors, and pathophysiology of AKI.
Collapse
Affiliation(s)
- Woon H Chong
- Department of Pulmonary and Critical Care Medicine; Albany Medical Center, Albany, New York, USA.
| | - Biplab K Saha
- Department of Pulmonary and Critical Care; Ozarks Medical Center, West Plains, Missouri, USA
| |
Collapse
|
79
|
Perico L, Benigni A, Remuzzi G. Angiotensin-converting enzyme 2: from a vasoactive peptide to the gatekeeper of a global pandemic. Curr Opin Nephrol Hypertens 2021; 30:252-263. [PMID: 33395036 DOI: 10.1097/mnh.0000000000000692] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
PURPOSE OF REVIEW We provide a comprehensive overview of angiotensin-converting enzyme 2 (ACE2) as a possible candidate for pharmacological approaches to halt inflammatory processes in different pathogenic conditions. RECENT FINDINGS ACE2 has quickly gained prominence in basic research as it has been identified as the main entry receptor for Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2). This novel pathogen causes Coronavirus Disease 2019 (COVID-19), a pathogenic condition that reached pandemic proportion and is associated with unprecedented morbidity and mortality. SUMMARY The renin-angiotensin system is a complex, coordinated hormonal cascade that plays a pivotal role in controlling individual cell behaviour and multiple organ functions. ACE2 acts as an endogenous counter-regulator to the pro-inflammatory and pro-fibrotic pathways triggered by ACE through the conversion of Ang II into the vasodilatory peptide Ang 1-7. We discuss the structure, function and expression of ACE2 in different tissues. We also briefly describe the role of ACE2 as a pivotal driver across a wide spectrum of pathogenic conditions, such as cardiac and renal diseases. Furthermore, we provide the most recent data concerning the possible role of ACE2 in mediating SARS-CoV-2 infection and dictating COVID-19 severity.
Collapse
Affiliation(s)
- Luca Perico
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| | - Ariela Benigni
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| | - Giuseppe Remuzzi
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
- Department of Biomedical and Clinical Sciences, University of Milan, Milan, Italy
| |
Collapse
|
80
|
Muciño-Bermejo MJ. Mechanisms of kidney dysfunction in the cirrhotic patient: Non-hepatorenal acute-on-chronic kidney damage considerations. Ann Hepatol 2021; 19:145-152. [PMID: 31594758 DOI: 10.1016/j.aohep.2019.06.022] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 06/22/2019] [Accepted: 06/25/2019] [Indexed: 02/04/2023]
Abstract
Renal dysfunction is a common finding in cirrhotic patients and has a great physiologic, and therefore, prognostic relevance. The combination of liver disease and renal dysfunction can occur as a result of systemic conditions that affect both the liver and the kidney, although primary disorders of the liver complicated by renal dysfunction are much more common. As most of the renal dysfunction scenarios in cirrhotic patients correspond to either prerenal azotemia or hepatorenal syndrome (HRS), physicians tend to conceive renal dysfunction in cirrhotic patients as mainly HRS. However, there are many systemic conditions that may cause both a "baseline" chronic kidney damage and a superimposed kidney dysfunction when this systemic condition worsens. The main aim of this article is to review some of the most important non prerenal non-HRS considerations regarding acute on chronic kidney dysfunction in cirrhotic patients, including renal manifestation of related to non-alcoholic steatohepatitis (NASH) viral hepatitis, the effect of cardiorenal syndrome in cirrhotics and corticosteroid-deficiency associated renal dysfunction.
Collapse
Affiliation(s)
- María-Jimena Muciño-Bermejo
- Medica Sur Clinical Foundation, Mexico City, Mexico; The American British Cowdray Medical Center, Mexico City, Mexico; International Renal Research Institute of Vicenza (IRRIV), Vicenza, Italy.
| |
Collapse
|
81
|
Bhattacharyya N, Singh S, Halder A, Adhikari A, Ghosh R, Shikha D, Tripathi SK, Mallick AK, Mondal P, Pal SK. An Energy-Resolved Optical Non-invasive Device Detects Essential Electrolyte Balance in Humans at Point-of-Care. TRANSACTIONS OF THE INDIAN NATIONAL ACADEMY OF ENGINEERING : AN INTERNATIONAL JOURNAL OF ENGINEERING AND TECHNOLOGY 2021; 6:355-364. [PMID: 35837575 PMCID: PMC7878167 DOI: 10.1007/s41403-021-00204-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Accepted: 01/21/2021] [Indexed: 06/15/2023]
Abstract
Regular monitoring of electrolyte balance is essential for patients suffering from chronic kidney disease (CKD), particularly those undergoing dialysis. In the context of the recent COVID-19 pandemic, more severe forms of infection are observed in elderly individuals and patients having co-morbidities like CKD. The repeated blood tests for the monitoring of electrolyte balance predispose them not only to COVID-19 but also other to hospital-acquired infections (HAI). Therefore, a non-invasive method for easy detection of essential electrolyte (K+ and Na+) levels is urgently needed. In this study, we developed an optical emission spectroscopy-based non-invasive device for simultaneous monitoring of salivary Na+ and K+ levels in a fast and reliable way. The device consisted of a closed spark chamber, micro-spectrometer, high voltage spark generator, electronic circuits, optical fiber, and an indigenously developed software based on the LabVIEW platform. The optical emission originating from the biological sample (i.e., saliva) due to recombination of ions energized by impingement of electrons returning from high voltage spark provides necessary information about the concentration of electrolytes. A small-scale clinical trial on 30 healthy human subjects shows the potential of the indigenously developed device in determining salivary Na + and K+ concentration. The low-cost, portable, point-of-care device requires only 2 mL of sample, and can simultaneously measure 1.0-190.0 mM Na+, and 1.0-270.9 mM K+ . To our understanding, the present work will find its relevance in combating COVID-19 morbidities, along with regular CKD patient-care.
Collapse
Affiliation(s)
- Neha Bhattacharyya
- Department of Radio Physics and Electronics, University of Calcutta, 92, Acharya Prafulla Chandra Rd, Machuabazar, Kolkata, 700009 India
- Technical Research Centre, S. N. Bose National Centre for Basic Sciences, Block JD, Sector III, Salt Lake, Kolkata, 700106 India
| | - Soumendra Singh
- Technical Research Centre, S. N. Bose National Centre for Basic Sciences, Block JD, Sector III, Salt Lake, Kolkata, 700106 India
| | - Animesh Halder
- Technical Research Centre, S. N. Bose National Centre for Basic Sciences, Block JD, Sector III, Salt Lake, Kolkata, 700106 India
- Department of Applied Optics and Photonics, University of Calcutta, Block JD, Sector III, Salt Lake, Kolkata, 700106 India
| | - Aniruddha Adhikari
- Department of Chemical, Biological and Macromolecular Sciences, S. N. Bose National Centre for Basic Sciences, Block JD, Sector III, Salt Lake, Kolkata, 700106 India
| | - Ria Ghosh
- Technical Research Centre, S. N. Bose National Centre for Basic Sciences, Block JD, Sector III, Salt Lake, Kolkata, 700106 India
| | - Deep Shikha
- Technical Research Centre, S. N. Bose National Centre for Basic Sciences, Block JD, Sector III, Salt Lake, Kolkata, 700106 India
| | - Santanu Kumar Tripathi
- Department of Clinical and Experimental Pharmacology, Calcutta School of Tropical Medicine, 108, Chittaranjan Avenue, Kolkata, 700073 India
| | - Asim Kumar Mallick
- Department of Paediatric Medicine, Nil Ratan Sircar Medical College and Hospital, 138, AJC Bose Road, Sealdah, Raja Bazar, Kolkata, 700014 India
| | - Pulak Mondal
- Department of Radio Physics and Electronics, University of Calcutta, 92, Acharya Prafulla Chandra Rd, Machuabazar, Kolkata, 700009 India
| | - Samir Kumar Pal
- Technical Research Centre, S. N. Bose National Centre for Basic Sciences, Block JD, Sector III, Salt Lake, Kolkata, 700106 India
- Department of Chemical, Biological and Macromolecular Sciences, S. N. Bose National Centre for Basic Sciences, Block JD, Sector III, Salt Lake, Kolkata, 700106 India
| |
Collapse
|
82
|
Soltani Zangbar H, Gorji A, Ghadiri T. A Review on the Neurological Manifestations of COVID-19 Infection: a Mechanistic View. Mol Neurobiol 2021; 58:536-549. [PMID: 32981023 PMCID: PMC7519857 DOI: 10.1007/s12035-020-02149-0] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Accepted: 09/23/2020] [Indexed: 01/08/2023]
Abstract
There is increasing evidence of neurological manifestations and complications in patients with coronavirus disease 19 (COVID-19). More than one-quarter of patients with COVID-19 developed various neurological symptoms, ranging from headache and dizziness to more serious medical conditions, such as seizures and stroke. The recent investigations introduced hyposmia as a potential early criterion of infection with COVID-19. Despite the high mortality and morbidity rate of COVID-19, its exact mechanism of action and pathogenesis is not well characterized. The spike protein of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) could interact with angiotensin-converting enzyme 2 (ACE2) in the endothelial, neural, and glial cells. In the present study, we reviewed the most common neurological manifestations and complications that emerged after infection with the SARS-CoV-2 and discussed their possible relation to the expression and function of ACE2. Comprehensive and detailed studies are required to uncover how this virus invades the neural system as well as other critical organs.
Collapse
Affiliation(s)
- Hamid Soltani Zangbar
- Department of Neuroscience and Cognition, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Gorji
- Epilepsy Research Center, Department of Neurology and Institute for Translational Neurology, Westfälische Wilhelms-Universität Münster, Münster, Germany
- Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran, Iran
| | - Tahereh Ghadiri
- Department of Neuroscience and Cognition, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
- Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran, Iran.
| |
Collapse
|
83
|
Parit R, Jayavel S. Association of ACE inhibitors and angiotensin type II blockers with ACE2 overexpression in COVID-19 comorbidities: A pathway-based analytical study. Eur J Pharmacol 2021; 896:173899. [PMID: 33508281 PMCID: PMC7839513 DOI: 10.1016/j.ejphar.2021.173899] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Revised: 01/10/2021] [Accepted: 01/19/2021] [Indexed: 01/08/2023]
Abstract
Severe Acute Respiratory Syndrome Corona Virus 2 (SARS-CoV-2) outbreak is a major public health concern, which has accounted for >1.7 million deaths across the world. A surge in the case fatality ratio as compared with the infection ratio has been observed in most of the countries. The novel Coronavirus SARS-CoV-2 shares the most common sequence with SARS-CoV, but it has a higher rate of transmission. The SARS-CoV-2 pathogenesis is initiated by the binding of viral spike protein with the target receptor Angiotensin-Converting Enzyme 2 (ACE2) facilitating virus internalization within host cells. SARS-CoV-2 mainly causes alveolar damage ranging from mild to severe clinical respiratory manifestations. Most of the cases have revealed the association of Coronavirus disease with patients having earlier comorbidities like Hypertension, Diabetes mellitus, and Cerebrovascular diseases. Pharmacological investigation of the SARS-Cov-2 patients has revealed the frequent use of drugs belongs to Angiotensin-converting enzyme inhibitors (ACEi) and/or Angiotensin II type I receptor blockers (ARBs). Interestingly, a significant increase in ACE2 expression was noticed in patients routinely treated with the above group of drugs were also reported. To date, the association of ACEi and/or ARBs with the up-regulation of ACE2 expression has not been defined distinctively. The proposed review will focus on the pathways which are responsible for the upregulation of ACE2 and its impact on gravity of SARS-CoV-2 disease.
Collapse
Affiliation(s)
- Rahul Parit
- Department of Biotechnology (DDE), Madurai Kamaraj University, Madurai, 625021, Tamilnadu, India
| | - Sridhar Jayavel
- Department of Biotechnology (DDE), Madurai Kamaraj University, Madurai, 625021, Tamilnadu, India.
| |
Collapse
|
84
|
Hanifah N, Achmad YF, Humaira A, Salasia SIO. Red ginger-extract nanoemulsion modulates high blood pressure in rats by regulating angiotensin-converting enzyme production. Vet World 2021; 14:176-181. [PMID: 33642802 PMCID: PMC7896892 DOI: 10.14202/vetworld.2021.176-181] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Accepted: 12/01/2020] [Indexed: 01/18/2023] Open
Abstract
Background and Aim: Red ginger (RG) has reportedly been used in folk medicine for the management and prevention of hypertension. One of the hypertension study models in experimental animals is the unilateral ureteral obstruction (UUO). This study aimed at evaluating the effect of RG-extract (RGE) nanoemulsion on UUO-induced hypertension and angiotensin-converting enzyme (ACE) production in rats. Materials and Methods: RG was extracted using ethanol, combined with virgin coconut oil, polysorbate 80, and polyethylene glycol 400 to form the oil phase. The particle sizes of RGE nanoemulsions were analyzed using a particle size analyzer. The UUO method was used to induce chronic kidney disease in rats (504 mg/200 g and 360 mg/200 g b/w per oral for 7 days). The systolic and diastolic blood pressure was determined non-invasively in conscious state by tail plethysmography using an automated blood pressure monitor. ACE in serum was measured using enzyme-linked immunosorbent assay. Results: The RGE nanoemulsions exhibited a particle size of32.8 nm and a polydispersity index (PI) of 0.268, indicating a homogenous nanoemulsion. UUO rats treated with RGE nanoemulsion (360 mg/200 g b/w) experienced a significant decrease in both their systolic blood pressure (p<0.05) from 142±1 mmHg to 107±6 mmHg and their diastolic blood pressure from 106±1 mmHg to 84±4 mmHg. Furthermore, treatment with RGE resulted in a 10.80% decrease in the level of ACE. Conclusion: The size and the PI of the RGE used in this study suggest a stable and effective distribution of the particle size in the emulsions. RGE nanoemulsions at the dose of 360 mg/200 g bw can be used as potential ACE inhibitors because they were found to decrease the blood pressure of hypertensive UUO rats.
Collapse
Affiliation(s)
- Nada Hanifah
- Department of Clinical Pathology, Faculty of Veterinary Medicine, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Yusuf Farid Achmad
- Department of Clinical Pathology, Faculty of Veterinary Medicine, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Aida Humaira
- Faculty of Pharmacy, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Siti Isrina Oktavia Salasia
- Department of Clinical Pathology, Faculty of Veterinary Medicine, Universitas Gadjah Mada, Yogyakarta, Indonesia
| |
Collapse
|
85
|
Yanes Cardozo LL, Rezq S, Pruett JE, Romero DG. Androgens, the kidney, and COVID-19: an opportunity for translational research. Am J Physiol Renal Physiol 2021; 320:F243-F248. [PMID: 33464168 DOI: 10.1152/ajprenal.00601.2020] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19) has reached pandemic proportions, affecting millions of people worldwide. Severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) is the causative agent of COVID-19. Epidemiological reports have shown that the severity of SARS-CoV-2 infection is associated with preexisting comorbidities such as hypertension, diabetes mellitus, cardiovascular diseases, and chronic kidney diseases, all of which are also risk factors for acute kidney injury (AKI). The kidney has emerged as a key organ affected by SARS-CoV-2. AKI is associated with increased morbidity and mortality in patients with COVID-19. Male sex is an independent predictor for AKI, and an increased death rate has been reported in male patients with COVID-19 worldwide. The mechanism(s) that mediate the sex discrepancy in mortality due to COVID-19 remain(s) unknown. Angiotensin-converting enzyme (ACE)2 is the receptor for SARS-CoV-2. Alterations in the ACE-to-ACE2 ratio have been implicated in renal diseases. This perspective aims to discuss data that suggest that androgens, via alterations in the intrarenal renin-angiotensin system, impair renal hemodynamics, predisposing patients to AKI during COVID-19 infection, which could explain the higher mortality observed in men with COVID-19. Clinicians should ensure early and effective cardiometabolic control for all patients to ameliorate the compensatory elevation of ACE2 and alterations in the ACE-to-ACE2 ratio. A better understanding of the role of androgens in SARS-CoV-2-associated AKI and mortality is imperative. The kidney could constitute a key organ that may explain the sex disparities of the higher mortality and worst outcomes associated with COVID-19 in men.
Collapse
Affiliation(s)
- Licy L Yanes Cardozo
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, Mississippi.,Department of Medicine, University of Mississippi Medical Center, Jackson, Mississippi.,Mississippi Center for Excellence in Perinatal Research, University of Mississippi Medical Center, Jackson, Mississippi.,Women's Health Research Center, University of Mississippi Medical Center, Jackson, Mississippi.,Cardio Renal Research Center, University of Mississippi Medical Center, Jackson, Mississippi
| | - Samar Rezq
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, Mississippi.,Mississippi Center for Excellence in Perinatal Research, University of Mississippi Medical Center, Jackson, Mississippi.,Women's Health Research Center, University of Mississippi Medical Center, Jackson, Mississippi.,Cardio Renal Research Center, University of Mississippi Medical Center, Jackson, Mississippi
| | - Jacob E Pruett
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, Mississippi.,Mississippi Center for Excellence in Perinatal Research, University of Mississippi Medical Center, Jackson, Mississippi.,Women's Health Research Center, University of Mississippi Medical Center, Jackson, Mississippi.,Cardio Renal Research Center, University of Mississippi Medical Center, Jackson, Mississippi
| | - Damian G Romero
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, Mississippi.,Mississippi Center for Excellence in Perinatal Research, University of Mississippi Medical Center, Jackson, Mississippi.,Women's Health Research Center, University of Mississippi Medical Center, Jackson, Mississippi.,Cardio Renal Research Center, University of Mississippi Medical Center, Jackson, Mississippi
| |
Collapse
|
86
|
Jankowski J, Lee HK, Wilflingseder J, Hennighausen L. Interferon-regulated genetic programs and JAK/STAT pathway activate the intronic promoter of the short ACE2 isoform in renal proximal tubules. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2021:2021.01.15.426908. [PMID: 33501441 PMCID: PMC7836111 DOI: 10.1101/2021.01.15.426908] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Recently, a short, interferon-inducible isoform of Angiotensin-Converting Enzyme 2 (ACE2), dACE2 was identified. ACE2 is a SARS-Cov-2 receptor and changes in its renal expression have been linked to several human nephropathies. These changes were never analyzed in context of dACE2, as its expression was not investigated in the kidney. We used Human Primary Proximal Tubule (HPPT) cells to show genome-wide gene expression patterns after cytokine stimulation, with emphasis on the ACE2/dACE2 locus. Putative regulatory elements controlling dACE2 expression were identified using ChIP-seq and RNA-seq. qRT-PCR differentiating between ACE2 and dACE2 revealed 300- and 600-fold upregulation of dACE2 by IFNα and IFNβ, respectively, while full length ACE2 expression was almost unchanged. JAK inhibitor ruxolitinib ablated STAT1 and dACE2 expression after interferon treatment. Finally, with RNA-seq, we identified a set of genes, largely immune-related, induced by cytokine treatment. These gene expression profiles provide new insights into cytokine response of proximal tubule cells.
Collapse
Affiliation(s)
- Jakub Jankowski
- Laboratory of Genetics and Physiology, National Institute of Diabetes and Digestive and Kidney Diseases, U.S. National Institutes of Health, Bethesda, MD 20892, USA
- Department of Physiology and Pathophysiology, University of Veterinary Medicine, Veterinärplatz 1, 1210, Vienna, Austria
| | - Hye Kyung Lee
- Laboratory of Genetics and Physiology, National Institute of Diabetes and Digestive and Kidney Diseases, U.S. National Institutes of Health, Bethesda, MD 20892, USA
| | - Julia Wilflingseder
- Department of Physiology and Pathophysiology, University of Veterinary Medicine, Veterinärplatz 1, 1210, Vienna, Austria
| | - Lothar Hennighausen
- Laboratory of Genetics and Physiology, National Institute of Diabetes and Digestive and Kidney Diseases, U.S. National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
87
|
Cabbab ILN, Manalo RVM. Anti-inflammatory drugs and the renin-angiotensin-aldosterone system: Current knowledge and potential effects on early SARS-CoV-2 infection. Virus Res 2021; 291:198190. [PMID: 33039544 PMCID: PMC7543703 DOI: 10.1016/j.virusres.2020.198190] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 09/21/2020] [Accepted: 10/05/2020] [Indexed: 12/20/2022]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the etiologic agent of coronavirus disease 19 (COVID-19), and is genetically related to the 2003 SARS and Middle East respiratory syndrome (MERS-CoV) coronaviruses. Recent studies have reported that similar to SARS-CoV, this strain expresses a spike protein (S) with a receptor binding domain (RBD) that binds to angiotensin-converting enzyme 2 (ACE2) - an enzyme expressed mostly in the endothelium, kidneys, heart, gastrointestinal tract and lungs - to facilitate viral entry and intracellular replication. Incidentally, the renin-angiotensin-aldosterone system (RAAS) is integral to physiologic control of both ACE and ACE2 expression, and is an essential system utilized by SARS-CoV-2, albeit with varying schools of thought on how it can affect viral entry. In this paper, we will review current knowledge on the RAAS and how it can be affected by non-steroidal anti-inflammatory drugs (NSAIDs) and corticosteroid use at the organ and cellular levels. We will then discuss the relevance of these interactions on organ-specific ACE2 expression, and provide scientific insights on how this mechanism can potentially affect SARS-CoV-2 infection in the early phases of disease. From the standpoint of other known viruses, we will then aim to discuss the potential uses or restrictions of these drugs in viral infection, and provide an update on relevant studies about COVID-19.
Collapse
Affiliation(s)
- Iris Louise N Cabbab
- College of Medicine - William Quasha H. Memorial, St. Luke's Medical Center, Quezon City, 1112, Philippines; National Institutes of Health, University of the Philippines Manila, Ermita, Manila, 1000, Philippines
| | - Rafael Vincent M Manalo
- College of Medicine, University of the Philippines Manila, Ermita, Manila, 1000, Philippines.
| |
Collapse
|
88
|
Yao Y, Lawrence DA. Susceptibility to COVID-19 in populations with health disparities: Posited involvement of mitochondrial disorder, socioeconomic stress, and pollutants. J Biochem Mol Toxicol 2021; 35:e22626. [PMID: 32905655 PMCID: PMC9340490 DOI: 10.1002/jbt.22626] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 07/30/2020] [Accepted: 08/25/2020] [Indexed: 12/18/2022]
Abstract
SARS-CoV-2 is a novel betacoronavirus that has caused the global health crisis known as COVID-19. The implications of mitochondrial dysfunction with COVID-19 are discussed as well as deregulated mitochondria and inter-organelle functions as a posited comorbidity enhancing detrimental outcomes. Many environmental chemicals (ECs) and endocrine-disrupting chemicals can do damage to mitochondria and cause mitochondrial dysfunction. During infection, SARS-CoV-2 via its binding target ACE2 and TMPRSS2 can disrupt mitochondrial function. Viral genomic RNA and structural proteins may also affect the normal function of the mitochondria-endoplasmic reticulum-Golgi apparatus. Drugs considered for treatment of COVID-19 should consider effects on organelles including mitochondria functions. Mitochondrial self-balance and clearance via mitophagy are important in SARS-CoV-2 infection, which indicate monitoring and protection of mitochondria against SARS-CoV-2 are important. Mitochondrial metabolomic analysis may provide new indicators of COVID-19 prognosis. A better understanding of the role of mitochondria during SARS-CoV-2 infection may help to improve intervention therapies and better protect mitochondrial disease patients from pathogens as well as people living with poor nutrition and elevated levels of socioeconomic stress and ECs.
Collapse
Affiliation(s)
- Yunyi Yao
- Wadsworth Center, New York State Department of Health, Center for Medical Science, Albany, New York
| | - David A Lawrence
- Wadsworth Center, New York State Department of Health, Center for Medical Science, Albany, New York
- Department of Environmental Health Sciences, University at Albany School of Public Health, Rensselaer, New York
| |
Collapse
|
89
|
Naljayan M, Yazdi F, Struthers S, Sharshir M, Williamson A, Simon EE. COVID-19 in New Orleans: A Nephrology Clinical and Education Perspective and Lessons Learned. Kidney Med 2021; 3:99-104. [PMID: 33283183 PMCID: PMC7708233 DOI: 10.1016/j.xkme.2020.09.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
New Orleans' first case of coronavirus disease 2019 (COVID-19) was reported on March 9, 2020, with a subsequent rapid increase in the number of cases throughout the state of Louisiana. Traditional educational efforts were no longer viable with social distancing and stay-at-home orders; therefore, virtual didactics were integrated into our curriculum. Due to an exponential increase in the number of patients with acute kidney injury requiring kidney replacement therapy, the nephrology sections at Louisiana State University School of Medicine and Tulane University School of Medicine adapted their clinical workflows to accommodate these increased clinical volumes by using prolonged intermittent kidney replacement therapies and acute peritoneal dialysis, as well as other strategies to mitigate nursing burnout and decrease scarce resource use. Telehealth was implemented in outpatient clinics and dialysis units to protect vulnerable patients with kidney disease while maintaining access to care. Lessons learned from this pandemic and subsequent response may be used for future responses in similar situations.
Collapse
Affiliation(s)
- Mihran Naljayan
- Section of Nephrology and Hypertension, LSU School of Medicine, New Orleans, LA
| | - Farshid Yazdi
- Section of Nephrology and Hypertension, LSU School of Medicine, New Orleans, LA
| | - Sarah Struthers
- Section of Nephrology and Hypertension, LSU School of Medicine, New Orleans, LA
| | - Moh’d Sharshir
- Section of Nephrology and Hypertension, Tulane School of Medicine and Southeast Louisiana Veterans Healthcare System, New Orleans, LA
| | | | - Eric E. Simon
- Section of Nephrology and Hypertension, Tulane School of Medicine and Southeast Louisiana Veterans Healthcare System, New Orleans, LA
| |
Collapse
|
90
|
Giusti S, Chazin S, Vaitla P, Atiemo K, Atari M, Paramesh A, Jeon H, Torres-Ortiz A, Thimmisetty R, Garces J. Observational Study of the Clinical Characteristics and Short-Term Outcomes of Kidney Transplant Recipients Diagnosed With COVID-19 Infection (SARS-CoV-2) Requiring Hospitalization in New Orleans. Ochsner J 2021; 21:329-334. [PMID: 34984045 PMCID: PMC8675612 DOI: 10.31486/toj.21.0008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Background: Kidney transplant recipients are at increased risk of severe disease and death caused by coronavirus disease 2019 (COVID-19) infection. The role of immunosuppressive medications in the clinical presentation, disease course, and outcomes is not well understood. Methods: We analyzed kidney transplant recipients diagnosed with COVID-19 and requiring hospitalization during the initial infection surge at 2 large transplant centers in New Orleans, Louisiana, between February 1, 2020 and April 30, 2020. Patient presentation, clinical course, kidney transplant function, and postdischarge details are included in this analysis. Results: Twenty-three kidney transplant recipients hospitalized with COVID-19 were included in the study. The majority of patients were Black (95.7%). Diabetes, hypertension, and obesity were present in more than 50% of the patients. The most common presenting symptom was fever, present in 52.2% of patients. All patients were managed with reduction in immunosuppression. Patients received azithromycin (60.9%), hydroxychloroquine (47.8%), remdesivir (8.7%), and intravenous methylprednisolone pulse (8.7%). The average length of stay was 4.5 days (range, 2-18 days). In this study population, 73.9% of the patients sustained acute kidney injury, with an average peak serum creatinine of 3.81 mg/dL. Twenty-six percent of the patients required renal replacement therapy. Seventy-seven percent of patients developed proteinuria (at least 1+ proteinuria on urinalysis). Of the patients in this population who required mechanical ventilation (39.1%), 77.8% died. Overall, 30.4% of patients died of COVID-19-related complications during admission. Of the 16 patients discharged, the average serum creatinine at discharge was 2.09 mg/dL compared with an average preadmission serum creatinine of 1.8 mg/dL. Conclusion: During the initial COVID-19 infection surge in New Orleans, we noted that kidney transplant recipients had initial symptoms similar to the general population. However, we recorded a higher incidence of acute kidney injury and death compared to nontransplant patients. Patients who required mechanical ventilation had a high mortality rate. Black patients are overrepresented in our study.
Collapse
Affiliation(s)
- Sixto Giusti
- Department of Medicine, Section of Nephrology and Hypertension, Tulane University School of Medicine, New Orleans, LA
- Department of Surgery, Kidney and Pancreas Transplant Program, Tulane University School of Medicine, New Orleans, LA
| | - Shai Chazin
- Department of Surgery, Kidney and Pancreas Transplant Program, Tulane University School of Medicine, New Orleans, LA
| | - Pradeep Vaitla
- Department of Medicine, Section of Nephrology and Transplant, University of Mississippi Medical Center, Jackson, MS
| | - Kofi Atiemo
- Department of Surgery, Kidney and Pancreas Transplant Program, Tulane University School of Medicine, New Orleans, LA
| | - Mohammad Atari
- Department of Medicine, Section of Nephrology and Hypertension, Tulane University School of Medicine, New Orleans, LA
| | - Anil Paramesh
- Department of Surgery, Kidney and Pancreas Transplant Program, Tulane University School of Medicine, New Orleans, LA
| | - Hoonbae Jeon
- Department of Surgery, Kidney and Pancreas Transplant Program, Tulane University School of Medicine, New Orleans, LA
| | - Aldo Torres-Ortiz
- Department of Internal Medicine, Ochsner Clinic Foundation, New Orleans, LA
| | - Ravi Thimmisetty
- Multi-Organ Transplant Institute, Ochsner Clinic Foundation, New Orleans, LA
| | - Jorge Garces
- Multi-Organ Transplant Institute, Ochsner Clinic Foundation, New Orleans, LA
- The University of Queensland Faculty of Medicine, Ochsner Clinic Foundation, New Orleans, LA
| |
Collapse
|
91
|
Neto MLR, da Silva CGL, do Socorro Vieira dos Santos M, Cândido EL, de Lima MAP, de França Lacerda Pinheiro S, Junior RFFP, Teixeira CS, Machado SSF, Pinheiro LFG, de Sousa GO, Galvão LMA, Gomes KGS, Medeiros KA, Diniz LA, de Oliveira ÍGP, Santana JRP, Rocha MAB, Damasceno IA, Cordeiro TL, da Silva Sales W. Epidemiology and Etiopathogeny of COVID-19. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1352:45-71. [DOI: 10.1007/978-3-030-85109-5_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
92
|
Fabrizi F, Alfieri CM, Cerutti R, Lunghi G, Messa P. COVID-19 and Acute Kidney Injury: A Systematic Review and Meta-Analysis. Pathogens 2020; 9:pathogens9121052. [PMID: 33334023 PMCID: PMC7765425 DOI: 10.3390/pathogens9121052] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 12/07/2020] [Accepted: 12/11/2020] [Indexed: 02/07/2023] Open
Abstract
Background: coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome—coronavirus-2 (SARS-CoV-2)—is an ongoing pandemic with high morbidity and mortality rates. Preliminary evidence suggests that acute kidney injury (AKI) is uncommon in patients with COVID-19 and associated with poor outcomes. Study aims and design: we performed a systematic review of the literature with a meta-analysis of clinical studies to evaluate the frequency of AKI and dialysis requirement in patients who underwent hospitalization due to COVID-19. The incidence of AKI according to the death risk was calculated in these patients. The random-effects model of DerSimonian and Laird was adopted, with heterogeneity and stratified analyses. Results: thirty-nine clinical studies (n = 25,566 unique patients) were retrieved. The pooled incidence of AKI was 0.154 (95% CI, 0.107; 0.201; p < 0.0001) across the studies. Significant heterogeneity was found (p = 0.0001). The overall frequency of COVID-19-positive patients who underwent renal replacement therapy (RRT) was 0.043 (95% CI, 0.031; 0.055; p < 0.0001); no publication bias was found (Egger’s test, p = 0.11). The pooled estimate of AKI incidence in patients with severe COVID-19 was 0.53 (95% CI, 0.427; 0.633) and heterogeneity occurred (Q = 621.08, I2 = 97.26, p = 0.0001). According to our meta-regression, age (p < 0.007) and arterial hypertension (p < 0.001) were associated with AKI occurrence in hospitalized COVID-19 positive patients. The odds ratio (OR) for the incidence of AKI in deceased COVID-19 positive patients was greater than among survivors, 15.4 (95% CI, 20.99; 11.4; p < 0.001). Conclusions: AKI is a common complication in hospitalized COVID-19 positive patients. Additional studies are under way to assess the risk of AKI in COVID-19 patients and to deepen the mechanisms of kidney injury.
Collapse
Affiliation(s)
- Fabrizio Fabrizi
- Division of Nephrology, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milano, Italy; (C.M.A.); (R.C.); (P.M.)
- Correspondence: ; Tel.: +39-2-55034552; Fax: +39-2-55034550
| | - Carlo M. Alfieri
- Division of Nephrology, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milano, Italy; (C.M.A.); (R.C.); (P.M.)
- Department of Clinical Sciences and Community Health, University of Milan, 20133 Milan, Italy
| | - Roberta Cerutti
- Division of Nephrology, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milano, Italy; (C.M.A.); (R.C.); (P.M.)
| | - Giovanna Lunghi
- Virology Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milano, Italy;
| | - Piergiorgio Messa
- Division of Nephrology, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milano, Italy; (C.M.A.); (R.C.); (P.M.)
- Department of Clinical Sciences and Community Health, University of Milan, 20133 Milan, Italy
| |
Collapse
|
93
|
Wang F, Ran L, Qian C, Hua J, Luo Z, Ding M, Zhang X, Guo W, Gao S, Gao W, Li C, Liu Z, Li Q, Ronco C. Epidemiology and Outcomes of Acute Kidney Injury in COVID-19 Patients with Acute Respiratory Distress Syndrome: A Multicenter Retrospective Study. Blood Purif 2020; 50:499-505. [PMID: 33291098 PMCID: PMC7801962 DOI: 10.1159/000512371] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 10/19/2020] [Indexed: 11/26/2022]
Abstract
Background Acute kidney injury (AKI) is associated with increased mortality in patients with acute respiratory distress syndrome (ARDS). However, the epidemiological features and outcomes of AKI among COVID-19 patients with ARDS are unknown. Methods We retrospectively recruited consecutive adult COVID-19 patients who were diagnosed with ARDS according to Berlin definition from 13 designated intensive care units in the city of Wuhan, China. Potential risk factors of AKI as well as the relation between AKI and in-hospital mortality were investigated. Results A total of 275 COVID-19 patients with ARDS were included in the study, and 49.5% of them developed AKI during their hospital stay. In comparison with patients without AKI, patients who developed AKI were older, tended to have chronic kidney disease, had higher Sepsis-Related Organ Failure Assessment score on day 1, and were more likely to receive invasive ventilation and develop acute organ dysfunction. Multivariate analysis showed that age, history of chronic kidney disease, neutrophil-to-lymphocyte ratio, and albumin level were independently associated with the occurrence of AKI. Importantly, increasing AKI severity was associated with increased in-hospital mortality when adjusted for other potential variables: odds ratio of stage 1 = 5.374 (95% CI: 2.147–13.452; p < 0.001), stage 2 = 6.216 (95% CI: 2.011–19.210; p = 0.002), and stage 3 = 34.033 (95% CI: 9.723–119.129; p < 0.001). Conclusion In this multicenter retrospective study, we found that nearly half of COVID-19 patients with ARDS experienced AKI during their hospital stay. The coexistence of AKI significantly increased the mortality of these patients.
Collapse
Affiliation(s)
- Feilong Wang
- Department of Pulmonary and Critical Care Medicine, Shanghai East Hospital, Tongji University, Shanghai, China,
| | - Linyu Ran
- Department of Pulmonary and Critical Care Medicine, Shanghai East Hospital, Tongji University, Shanghai, China
| | - Chenchen Qian
- Department of Internal Medicine, UPMC Pinnacle Hospital, Harrisburg, Pennsylvania, USA
| | - Jing Hua
- Department of Pulmonary and Critical Care Medicine, Shanghai East Hospital, Tongji University, Shanghai, China
| | - Zhibing Luo
- Department of Pulmonary and Critical Care Medicine, Shanghai East Hospital, Tongji University, Shanghai, China
| | - Min Ding
- Department of Pulmonary Medicine, Fengcheng Hospital, Fengcheng, China
| | - Xing Zhang
- Department of Pulmonary and Critical Care Medicine, People's Liberation Army Joint Logistic Support Force 920th Hospital, Kunming, China
| | - Wei Guo
- Trauma Center, Peking University People's Hospital, Beijing, China
| | - Shaoyong Gao
- Department of Pulmonary and Critical Care Medicine, Shanghai East Hospital, Tongji University, Shanghai, China
| | - Weibo Gao
- Department of Emergency, Peking University People's Hospital, Beijing, China
| | - Chaoping Li
- Department of Radiology, Fengcheng Hospital, Fengcheng, China
| | - Zhongmin Liu
- Department of Pulmonary and Critical Care Medicine, Shanghai East Hospital, Tongji University, Shanghai, China
| | - Qiang Li
- Department of Pulmonary and Critical Care Medicine, Shanghai East Hospital, Tongji University, Shanghai, China
| | - Claudio Ronco
- Department of Medicine (DIMED), University of Padova, Padova, Italy.,Department of Nephrology Dialysis and Transplantation, International Renal Research Institute of Vicenza, San Bortolo Hospital, Vicenza, Italy
| |
Collapse
|
94
|
Sahu SN, Mishra B, Sahu R, Pattanayak SK. Molecular dynamics simulation perception study of the binding affinity performance for main protease of SARS-CoV-2. J Biomol Struct Dyn 2020; 40:2444-2459. [PMID: 33228481 PMCID: PMC7754937 DOI: 10.1080/07391102.2020.1850362] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Like common cold and flu, SARC-CoV-2 virus spreads by droplets of sneezes or coughs which virus affects people of various age groups. Today, this virus is almost distributed all over the world. Since binding process plays a crucial role between host and receptor, therefore, we studied the molecules intended toward inhibition process through molecular docking and molecular dynamics simulation process. From the molecular docking study, it is noteworthy that remdesivir shows better binding affinity toward the main protease of SARS-CoV2 compared to other studied drugs. Within studied phytochemicals, carnosic acid shows better binding poses toward main protease of SARS-CoV2 among studied phytochemicals. The amino acid residues GLN110 and PHE294 were almost found in all the studied interactions of drugs and phytochemicals with main protease of SARS-CoV-2. Furthermore, the results show a larger contribution of the Van der Waals energies as compared to others like electrostatic energies suggesting that ligands at the binding pocket are predominantly stabilized by hydrophobic interactions. The conformational change during ligand binding was predicted from Gibbs free energy landscape analysis through molecular dynamics simulation. We observed that, there were two main free energy basins for both docked carnosic acid complex and for docked remdesivir complex, only one main free energy basin was found in the global free energy minimum region. Communicated by Ramaswamy H. Sarma
Collapse
Affiliation(s)
- Satya Narayan Sahu
- School of Applied Sciences, Kalinga Institute of Industrial Technology (KIIT), Deemed to be University, Bhubaneswar, India
| | - Biswajit Mishra
- School of Applied Sciences, Kalinga Institute of Industrial Technology (KIIT), Deemed to be University, Bhubaneswar, India
| | - Rojalin Sahu
- School of Applied Sciences, Kalinga Institute of Industrial Technology (KIIT), Deemed to be University, Bhubaneswar, India
| | | |
Collapse
|
95
|
Adverse impact of renin-angiotensin system blockade on the clinical course in hospitalized patients with severe COVID-19: a retrospective cohort study. Sci Rep 2020; 10:20250. [PMID: 33219294 PMCID: PMC7680105 DOI: 10.1038/s41598-020-76915-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 11/03/2020] [Indexed: 01/08/2023] Open
Abstract
The association between angiotensin-converting enzyme inhibitor (ACE-I) or angiotensin II receptor blocker (ARB) and the risk of mortality in hospitalized patients with severe coronavirus disease 2019 (COVID-19) was investigated. This retrospective cohort study was performed in all hospitalized patients with COVID-19 in tertiary hospitals in Daegu, Korea. Patients were classified based on whether they received ACE-I or ARB before COVID-19 diagnosis. The analysis of the primary outcome, in-hospital mortality, was performed using the Cox proportional hazards regression model. Of 130 patients with COVID-19, 30 (23.1%) who received ACE-I or ARB exhibited an increased risk of in-hospital mortality (adjusted hazard ratio, 2.20; 95% confidence interval [CI], 1.10–4.38; P = 0.025). ACE-I or ARB was also associated with severe complications, such as acute respiratory distress syndrome (ARDS) (adjusted odds ratio [aOR], 2.58; 95% CI, 1.02–6.51; P = 0.045) and acute kidney injury (AKI) (aOR, 3.06; 95% CI, 1.15–8.15; P = 0.026). Among the patients with ACE-I or ARB therapy, 8 patients (26.7%) used high equivalent doses of ACE-I or ARB and they had higher in-hospital mortality and an increased risk of ARDS and AKI (all, P < 0.05). ACE-I or ARB therapy in patients with severe COVID-19 was associated with the occurrence of severe complications and increased in-hospital mortality. The potentially harmful effect of ACE-I or ARB therapy may be higher in patients who received high doses.
Collapse
|
96
|
Li Z, Xu X, Yang M, Feng J, Liu C, Yang C. Role of angiotensin-converting enzyme 2 in neurodegenerative diseases during the COVID-19 pandemic. Aging (Albany NY) 2020; 12:24453-24461. [PMID: 33197881 PMCID: PMC7762468 DOI: 10.18632/aging.103993] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 08/15/2020] [Indexed: 12/13/2022]
Abstract
SARS-CoV-2 (severe acute respiratory syndrome coronavirus 2) uses the angiotensin-converting enzyme 2 (ACE2) receptor for infecting and spreading in humans. Studies have shown that the widespread expression of ACE2 in human tissues may be associated with organ function damage (e.g., lung, kidney, and stomach) in patients with coronavirus disease 2019 (COVID-19). However, in neurodegenerative diseases, whose pathogenesis is closely related to advanced age, ACE2 plays a neurotrophic and protective role by activating the ACE2/Ang-(1-7)/Mas axis, thus inhibiting cognitive impairment. Early reports have revealed that the elderly are more susceptible to COVID-19 and that elderly patients with COVID-19 have faster disease progression and higher mortality. Therefore, during the COVID-19 pandemic, it is crucial to understand the role of ACE2 in neurodegenerative diseases. In this paper, we review the relationship between COVID-19, neurodegenerative diseases, and ACE2, as well as provide recommendations for the protection of elderly patients with neurodegenerative diseases during the COVID-19 pandemic.
Collapse
Affiliation(s)
- Zhenyu Li
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Xiaolin Xu
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Wuhan 430030, China
| | - Meiling Yang
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Jianping Feng
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Cunming Liu
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Chun Yang
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| |
Collapse
|
97
|
Gan R, Rosoman NP, Henshaw DJE, Noble EP, Georgius P, Sommerfeld N. COVID-19 as a viral functional ACE2 deficiency disorder with ACE2 related multi-organ disease. Med Hypotheses 2020; 144:110024. [PMID: 32758871 PMCID: PMC7308773 DOI: 10.1016/j.mehy.2020.110024] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 06/19/2020] [Indexed: 12/31/2022]
Abstract
SARS-CoV-2, the agent of COVID-19, shares a lineage with SARS-CoV-1, and a common fatal pulmonary profile but with striking differences in presentation, clinical course, and response to treatment. In contrast to SARS-CoV-1 (SARS), COVID-19 has presented as an often bi-phasic, multi-organ pathology, with a proclivity for severe disease in the elderly and those with hypertension, diabetes and cardiovascular disease. Whilst death is usually related to respiratory collapse, autopsy reveals multi-organ pathology. Chronic pulmonary disease is underrepresented in the group with severe COVID-19. A commonality of aberrant renin angiotensin system (RAS) is suggested in the at-risk group. The identification of angiotensin-converting-enzyme 2 (ACE2) as the receptor allowing viral entry to cells precipitated our interest in the role of ACE2 in COVID-19 pathogenesis. We propose that COVID-19 is a viral multisystem disease, with dominant vascular pathology, mediated by global reduction in ACE2 function, pronounced in disease conditions with RAS bias toward angiotensin-converting-enzyme (ACE) over ACE2. It is further complicated by organ specific pathology related to loss of ACE2 expressing cells particularly affecting the endothelium, alveolus, glomerulus and cardiac microvasculature. The possible upregulation in ACE2 receptor expression may predispose individuals with aberrant RAS status to higher viral load on infection and relatively more cell loss. Relative ACE2 deficiency leads to enhanced and protracted tissue, and vessel exposure to angiotensin II, characterised by vasoconstriction, enhanced thrombosis, cell proliferation and recruitment, increased tissue permeability, and cytokine production (including IL-6) resulting in inflammation. Additionally, there is a profound loss of the "protective" angiotensin (1-7), a vasodilator with anti-inflammatory, anti-thrombotic, antiproliferative, antifibrotic, anti-arrhythmic, and antioxidant activity. Our model predicts global vascular insult related to direct endothelial cell damage, vasoconstriction and thrombosis with a disease specific cytokine profile related to angiotensin II rather than "cytokine storm". Our proposed mechanism of lung injury provides an explanation for early hypoxia without reduction in lung compliance and suggests a need for revision of treatment protocols to address vasoconstriction, thromboprophylaxis, and to minimize additional small airways and alveolar trauma via ventilation choice. Our model predicts long term sequelae of scarring/fibrosis in vessels, lungs, renal and cardiac tissue with protracted illness in at-risk individuals. It is hoped that our model stimulates review of current diagnostic and therapeutic intervention protocols, particularly with respect to early anticoagulation, vasodilatation and revision of ventilatory support choices.
Collapse
Affiliation(s)
- Rosemary Gan
- Nambour Selangor Private Hospital, Nambour, QLD, Australia.
| | | | | | - Euan P Noble
- Sunshine Coast University Private Hospital, Birtinya, QLD, Australia
| | | | | |
Collapse
|
98
|
Abstract
Despite myriad improvements in the care of COVID-19 patients, atypical manifestations are least appreciated during the current pandemic. Because COVID-19 is primarily manifesting as an acute respiratory illness with interstitial and alveolar pneumonia, the possibility of viral invasions into the other organs cannot be disregarded. Acute kidney injury (AKI) has been associated with various viral infections including dengue, chikungunya, Zika, and HIV. The prevalence and risks of AKI during the course of COVID-19 have been described in few studies. However, the existing literature demonstrate great disparity across findings amid variations in methodology and population. This article underscores the propensity of AKI among COVID-19 patients, limitations of the exiting evidence, and importance of timely identification during the case management. The prevalence of AKI is variable across the studies ranging from 4.7% to 81%. Evidence suggest old age, comorbidities, ventilator support, use of vasopressors, black race, severe infection, and elevated levels of baseline serum creatinine and d-dimers are independent risk factors of COVID-19 associated with AKI. COVID-19 patients with AKI also showed unsatisfactory renal recovery and higher mortality rate as compared with patients without AKI. These findings underscore that AKI frequently occurs during the course of COVID-19 infection and requires early stratification and management.
Collapse
Affiliation(s)
- Tauqeer Hussain Mallhi
- Department of Clinical Pharmacy, College of Pharmacy, Jouf University, Sakaka, Kingdom of Saudi Arabia
| | - Yusra Habib Khan
- Department of Clinical Pharmacy, College of Pharmacy, Jouf University, Sakaka, Kingdom of Saudi Arabia
| | - Azreen Syazril Adnan
- Chronic Kidney Disease Resource Center, School of Medical Sciences, Hospital Universiti Sains Malaysia, Kota Bharu, Malaysia.,Department of Nephrology, MSU Medical Centre, Management and Science University Shah Alam, Malaysia
| |
Collapse
|
99
|
Neil D, Moran L, Horsfield C, Curtis E, Swann O, Barclay W, Hanley B, Hollinshead M, Roufosse C. Ultrastructure of cell trafficking pathways and coronavirus: how to recognise the wolf amongst the sheep. J Pathol 2020; 252:346-357. [PMID: 32918747 DOI: 10.1002/path.5547] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 08/10/2020] [Accepted: 09/06/2020] [Indexed: 12/15/2022]
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic has resulted in an urgent need to understand the pathophysiology of SARS-CoV-2 infection, to assist in the identification of treatment strategies. Viral tissue tropism is an active area of investigation, one approach to which is identification of virus within tissues by electron microscopy of post-mortem and surgical specimens. Most diagnostic histopathologists have limited understanding of the ultrastructural features of normal cell trafficking pathways, which can resemble intra- and extracellular coronavirus; in addition, viral replication pathways make use of these trafficking pathways. Herein, we review these pathways and their ultrastructural appearances, with emphasis on structures which may be confused with coronavirus. In particular, we draw attention to the fact that, when using routine fixation and processing, the typical 'crown' that characterises a coronavirus is not readily identified on intracellular virions, which are located in membrane-bound vacuoles. In addition, the viral nucleocapsid is seen as black dots within the virion and is more discriminatory in differentiating virions from other cellular structures. The identification of the viral replication organelle, a collection of membranous structures (convoluted membranes) seen at a relatively low scanning power, may help to draw attention to infected cells, which can be sparse. © 2020 The Authors. The Journal of Pathology published by John Wiley & Sons, Ltd. on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Desley Neil
- Department of Cellular Pathology, Queen Elizabeth Hospital Birmingham, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK.,School of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Linda Moran
- North West London Pathology, Imperial College Healthcare NHS Trust, London, UK.,Department of Immunology and Inflammation, Centre for Inflammatory Diseases, Faculty of Medicine, Imperial College London, London, UK
| | - Catherine Horsfield
- Department of Histopathology, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Elizabeth Curtis
- Department of Cellular Pathology, Queen Elizabeth Hospital Birmingham, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Olivia Swann
- Department of Infectious Disease, Faculty of Medicine, Imperial College London, London, UK
| | - Wendy Barclay
- Department of Infectious Disease, Faculty of Medicine, Imperial College London, London, UK
| | - Brian Hanley
- North West London Pathology, Imperial College Healthcare NHS Trust, London, UK.,Department of Immunology and Inflammation, Centre for Inflammatory Diseases, Faculty of Medicine, Imperial College London, London, UK
| | | | - Candice Roufosse
- North West London Pathology, Imperial College Healthcare NHS Trust, London, UK.,Department of Immunology and Inflammation, Centre for Inflammatory Diseases, Faculty of Medicine, Imperial College London, London, UK
| |
Collapse
|
100
|
Gao YL, Du Y, Zhang C, Cheng C, Yang HY, Jin YF, Duan GC, Chen SY. Role of Renin-Angiotensin System in Acute Lung Injury Caused by Viral Infection. Infect Drug Resist 2020; 13:3715-3725. [PMID: 33116692 PMCID: PMC7585866 DOI: 10.2147/idr.s265718] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 09/12/2020] [Indexed: 12/14/2022] Open
Abstract
The renin-angiotensin system (RAS) is the most important regulatory system of electrolyte homeostasis and blood pressure and acts through angiotensin-converting enzyme (ACE)/angiotensin II (Ang II)/Ang II type 1 (AT1) receptor axis and angiotensin-converting enzyme 2 (ACE2)/angiotensin (1-7)/MAS receptor axis. RAS dysfunction is related to the occurrence and development of acute lung injury (ALI) and acute respiratory distress syndrome (ARDS) and causes a serious prognosis and even death. ALI/ARDS can be induced by various ways, one of which is viral infections, such as SARS-CoV, SARS-CoV-2, H5N1, H7N9, and EV71. This article reviews the specific mechanism on how RAS dysfunction affects ALI/ARDs caused by viral infections. SARS-CoV and SARS-CoV-2 enter the host cells by binding with ACE2. H5N1 and H7N9 avian influenza viruses reduce the ACE2 level in the body, and EV71 increases Ang II concentration. Treatment with angiotensin-converting enzyme inhibitor and angiotensin AT1 receptor blocker can alleviate ALI/ARDS symptoms. This review provides suggestions for the treatment of lung injury caused by viral infections.
Collapse
Affiliation(s)
- Yan-Lei Gao
- Department of Epidemiology, College of Public Health, Zhengzhou University, Zhengzhou, Henan, People’s Republic of China
| | - Yue Du
- Department of Epidemiology, College of Public Health, Zhengzhou University, Zhengzhou, Henan, People’s Republic of China
| | - Chao Zhang
- Department of Epidemiology, College of Public Health, Zhengzhou University, Zhengzhou, Henan, People’s Republic of China
| | - Cheng Cheng
- Department of Epidemiology, College of Public Health, Zhengzhou University, Zhengzhou, Henan, People’s Republic of China
| | - Hai-Yan Yang
- Department of Epidemiology, College of Public Health, Zhengzhou University, Zhengzhou, Henan, People’s Republic of China
| | - Yue-Fei Jin
- Department of Epidemiology, College of Public Health, Zhengzhou University, Zhengzhou, Henan, People’s Republic of China
| | - Guang-Cai Duan
- Department of Epidemiology, College of Public Health, Zhengzhou University, Zhengzhou, Henan, People’s Republic of China
| | - Shuai-Yin Chen
- Department of Epidemiology, College of Public Health, Zhengzhou University, Zhengzhou, Henan, People’s Republic of China
| |
Collapse
|