1
|
Ren Z, Li G. A comprehensive review on the association and prevention of long-term COVID-induced heart failure: A review. Medicine (Baltimore) 2024; 103:e38736. [PMID: 38968497 PMCID: PMC11224805 DOI: 10.1097/md.0000000000038736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Accepted: 06/07/2024] [Indexed: 07/07/2024] Open
Abstract
The coronavirus disease 2019 (COVID-19) disease caused by the severe acute respiratory syndrome coronavirus 2 has had a widespread global impact. In addition to the main respiratory symptoms, research has found significant effects of this virus on the cardiovascular system. This article comprehensively explores the phenomenon of "long-term COVID-19" or postacute sequelae of severe acute respiratory syndrome coronavirus 2 infection, wherein some recovered patients continue to experience long-term health issues after the resolution of acute illness. We delve into the potential reasons behind these symptoms, including increased risk of heart disease, myocardial injury, abnormal inflammatory responses, thrombosis formation, and immune system dysfunction, among others. Furthermore, this paper highlights the potential association between long-term COVID-19 and HF (heart failure), and proposes corresponding preventive strategies. To address this, we advocate for a collaborative approach involving interdisciplinary teams for treatment and management.
Collapse
Affiliation(s)
- Zhangqing Ren
- Department of General Practice, Shaoxing People’s Hospital, Shaoxing, China
| | - Gang Li
- Department of General Practice, Shaoxing People’s Hospital, Shaoxing, China
| |
Collapse
|
2
|
Prato M, Tiberti N, Mazzi C, Gobbi F, Piubelli C, Longoni SS. The Renin-Angiotensin System (RAS) in COVID-19 Disease: Where We Are 3 Years after the Beginning of the Pandemic. Microorganisms 2024; 12:583. [PMID: 38543635 PMCID: PMC10975343 DOI: 10.3390/microorganisms12030583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 03/04/2024] [Accepted: 03/08/2024] [Indexed: 11/12/2024] Open
Abstract
The RAS is a hormonal system playing a pivotal role in the control of blood pressure and electrolyte homeostasis, the alteration of which is associated with different pathologies, including acute respiratory distress syndrome (ARDS). As such, it is not surprising that a number of studies have attempted to elucidate the role and balance of the renin-angiotensin system (RAS) in COVID-19. In this review article, we will describe the evidence collected regarding the two main enzymes of the RAS (i.e., ACE and ACE2) and their principal molecular products (i.e., AngII and Ang1-7) in SARS-CoV-2 infection, with the overarching goal of drawing conclusions on their possible role as clinical markers in association with disease severity, progression, and outcome. Moreover, we will bring into the picture new experimental data regarding the systemic activity of ACE and ACE2 as well as the concentration of AngII and Ang1-7 in a cohort of 47 COVID-19 patients hospitalized at the IRCCS Sacro Cuore-Don Calabria Hospital (Negrar, Italy) between March and April 2020. Finally, we will discuss the possibility of considering this systemic pathway as a clinical marker for COVID-19.
Collapse
Affiliation(s)
- Marco Prato
- Department of Infectious, Tropical Diseases and Microbiology, IRCCS Sacro Cuore Don Calabria Hospital, Negrar di Valpolicella, 37024 Verona, Italy
| | - Natalia Tiberti
- Department of Infectious, Tropical Diseases and Microbiology, IRCCS Sacro Cuore Don Calabria Hospital, Negrar di Valpolicella, 37024 Verona, Italy
| | - Cristina Mazzi
- Centre for Clinical Research, IRCCS Sacro Cuore Don Calabria Hospital, Negrar di Valpolicella, 37024 Verona, Italy
| | - Federico Gobbi
- Department of Infectious, Tropical Diseases and Microbiology, IRCCS Sacro Cuore Don Calabria Hospital, Negrar di Valpolicella, 37024 Verona, Italy
| | - Chiara Piubelli
- Department of Infectious, Tropical Diseases and Microbiology, IRCCS Sacro Cuore Don Calabria Hospital, Negrar di Valpolicella, 37024 Verona, Italy
| | - Silvia Stefania Longoni
- Department of Infectious, Tropical Diseases and Microbiology, IRCCS Sacro Cuore Don Calabria Hospital, Negrar di Valpolicella, 37024 Verona, Italy
| |
Collapse
|
3
|
Li X, Li X, Kang B, Eom Y, Lee HK, Kim DH, Zhong J, Song JS. Effects of particulate matter exposure on the expression of the SARS-CoV-2 ACE2 receptor in ocular surface tissues and cells. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2024; 31:8768-8780. [PMID: 38180673 DOI: 10.1007/s11356-023-31607-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 12/14/2023] [Indexed: 01/06/2024]
Abstract
Particulate matter (PM) has been reported to be one of the risk factor for COVID-19 caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). However, although the ocular surface is deeply affected by both PM exposure and SARS-COV-2 infection, no studies have investigated the effects of PM exposure on the ocular route of SARS-COV-2 infection. To this end, we explored the effects of PM on the expression of SARS-COV-2-associated receptors and proteins in ocular surface. Herein, short- and long-term PM-exposed rat models were established by topically administering PM for 3 and 10 days, respectively. Immortalized human corneal epithelial cells (HCECs) and human conjunctival epithelial cells (HCjECs) were exposed to PM. ACE2, TMPRSS2, CD147, and ADAM17 expression levels were measured by western blot analysis. Our results show that short-term PM exposure had little effect on the expressions of ACE2, TMPRSS2, and CD147 in ocular surface tissues. However, long-term PM exposure decreased the ACE2 expression in conjunctival tissues and increased the CD147 expression in corneal or conjunctival tissues. PM exposure reduced the ACE2 expression by increasing the ADAM17 expression and ACE2 shedding level in HCECs and HCjECs. Our findings suggest that long-term PM exposure down-regulate the expression of the SARS-CoV-2 receptor ACE2 in conjunctival tissues through ADAM17-dependent ACE2 shedding. However, long-term PM exposure up-regulates the expression of another SARS-CoV-2 receptor CD147 in ocular surface tissues, accompanied by ocular surface damage and cytotoxicity. This study provides a new insight into uncovering potential risk factors for infection with SARS-CoV-2 via the ocular route.
Collapse
Affiliation(s)
- Xiangzhe Li
- Department of Ophthalmology, The First Affiliated Hospital of Ningbo University, Ningbo, China
| | - Xuemin Li
- Department of Ophthalmology, Korea University College of Medicine, 80, Guro-Dong, Guro-Gu, Seoul, 152-703, South Korea
| | - Boram Kang
- Department of Ophthalmology, Korea University College of Medicine, 80, Guro-Dong, Guro-Gu, Seoul, 152-703, South Korea
| | - Youngsub Eom
- Department of Ophthalmology, Korea University College of Medicine, 80, Guro-Dong, Guro-Gu, Seoul, 152-703, South Korea
| | - Hyung Keun Lee
- Institute of Vision Research, Department of Ophthalmology, Yonsei University College of Medicine, Seoul, South Korea
| | - Dong Hyun Kim
- Department of Ophthalmology, Korea University College of Medicine, 80, Guro-Dong, Guro-Gu, Seoul, 152-703, South Korea
| | - Jingxiang Zhong
- Department of Ophthalmology, The Sixth Affiliated Hospital of Jinan University, Dongguan, China
- Department of Ophthalmology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Jong Suk Song
- Department of Ophthalmology, Korea University College of Medicine, 80, Guro-Dong, Guro-Gu, Seoul, 152-703, South Korea.
| |
Collapse
|
4
|
Deng Y, Ding W, Peng Q, Wang W, Duan R, Zhang Y. Advancement in Beneficial Effects of AVE 0991: A Brief Review. Mini Rev Med Chem 2024; 24:139-158. [PMID: 36998128 DOI: 10.2174/1389557523666230328134932] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 01/08/2023] [Accepted: 01/11/2023] [Indexed: 04/01/2023]
Abstract
AVE 0991, a non-peptide analogue of Angiotensin-(1-7) [Ang-(1-7)], is orally active and physiologically well tolerated. Several studies have demonstrated that AVE 0991 improves glucose and lipid metabolism, and contains anti-inflammatory, anti-apoptotic, anti-fibrosis, and anti-oxidant effects. Numerous preclinical studies have also reported that AVE 0991 appears to have beneficial effects on a variety of systemic diseases, including cardiovascular, liver, kidney, cancer, diabetes, and nervous system diseases. This study searched multiple literature databases, including PubMed, Web of Science, EMBASE, Google Scholar, Cochrane Library, and the ClinicalTrials.gov website from the establishment to October 2022, using AVE 0991 as a keyword. This literature search revealed that AVE 0991 could play different roles via various signaling pathways. However, the potential mechanisms of these effects need further elucidation. This review summarizes the benefits of AVE 0991 in several medical problems, including the COVID-19 pandemic. The paper also describes the underlying mechanisms of AVE 0991, giving in-depth insights and perspectives on the pharmaceutical value of AVE 0991 in drug discovery and development.
Collapse
Affiliation(s)
- Yang Deng
- School of Basic Medicine & Clinical Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - Wangli Ding
- School of Basic Medicine & Clinical Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - Qiang Peng
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China
| | - Wei Wang
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China
| | - Rui Duan
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China
| | - Yingdong Zhang
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China
| |
Collapse
|
5
|
Khan NA, Elsori D, Rashid G, Tamanna S, Chakraborty A, Farooqi A, Kar A, Sambyal N, Kamal MA. Unraveling the relationship between the renin-angiotensin system and endometrial cancer: a comprehensive review. Front Oncol 2023; 13:1235418. [PMID: 37869088 PMCID: PMC10585148 DOI: 10.3389/fonc.2023.1235418] [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: 06/06/2023] [Accepted: 09/04/2023] [Indexed: 10/24/2023] Open
Abstract
Endometrial cancer (EC), the most common adenocarcinoma, represents 90% of uterine cancer in women with an increased incidence of occurrence attributed to age, obesity, hypertension, and hypoestrogenism. Being the most common gynecological malignancy in women, it shows a relation with the activation of different components of the renin-angiotensin system (RAS), which is predominantly involved in maintaining blood pressure, salt, water, and aldosterone secretion, thereby playing a significant role in the etiology of hypertension. The components of the RAS, i.e., ACE-I, ACE-II, AT1R, AT2R, and Pro(renin) receptor, are widely expressed in both glandular and stromal cells of the endometrium, with varying levels throughout the different phases of the menstrual cycle. This causes the endometrial RAS to implicate angiogenesis, neovascularization, and cell proliferation. Thus, dysfunctioning of the endometrial RAS could predispose the growth and spread of EC. Interestingly, the increased expression of AngII, AGTR1, and AGTR2 showed advancement in the stages and progression of EC via the prorenin/ATP6AP2 and AngII/AGTR1 pathway. Therefore, this review corresponds to unraveling the relationship between the progression and development of endometrial cancer with the dysfunction in the expression of various components associated with RAS in maintaining blood pressure.
Collapse
Affiliation(s)
- Nihad Ashraf Khan
- Department of Biosciences, Faculty of Natural Sciences, Jamia Millia Islamia, Delhi, India
| | - Deena Elsori
- Faculty of Resillience, Deans Office Rabdan Academy, Abu Dhabi, United Arab Emirates
| | - Gowhar Rashid
- Amity Medical School, Amity University, Gurgaon, Haryana, India
| | - Sonia Tamanna
- Department of Biochemistry and Molecular Biology, University of Dhaka, Dhaka, Bangladesh
| | - Ananya Chakraborty
- Department of Biotechnology, Adamas University, Kolkata, West Bengal, India
| | - Adeeba Farooqi
- Department of Biotechnology, Central University of Kashmir, Ganderbal, India
| | - Ayman Kar
- Department of Biotechnology, Central University of Kashmir, Ganderbal, India
| | - Niti Sambyal
- Department of Biotechnology, Shri Mata Vashino Devi University, Katra, Jammu, India
| | - Mohammad Azhar Kamal
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Alkharj, Saudi Arabia
| |
Collapse
|
6
|
Zhang J, Bishir M, Barbhuiya S, Chang SL. Meta-Analysis of the Mechanisms Underlying COVID-19 Modulation of Parkinson's Disease. Int J Mol Sci 2023; 24:13554. [PMID: 37686360 PMCID: PMC10487929 DOI: 10.3390/ijms241713554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 08/25/2023] [Accepted: 08/28/2023] [Indexed: 09/10/2023] Open
Abstract
Coronavirus disease-19 (COVID-19) is caused by the infection of severe acute respiratory syndrome-coronavirus-2 (SARS-CoV-2). The virus enters host cells through receptor-mediated endocytosis of angiotensin-converting enzyme-2 (ACE2), leading to systemic inflammation, also known as a "cytokine storm", and neuroinflammation. COVID-19's upstream regulator, interferon-gamma (IFNG), is downregulated upon the infection of SARS-CoV-2, which leads to the downregulation of ACE2. The neuroinflammation signaling pathway (NISP) can lead to neurodegenerative diseases, such as Parkinson's disease (PD), which is characterized by the formation of Lewy bodies made primarily of the α-synuclein protein encoded by the synuclein alpha (SNCA) gene. We hypothesize that COVID-19 may modulate PD progression through neuroinflammation induced by cytokine storms. This study aimed to elucidate the possible mechanisms and signaling pathways involved in COVID-19-triggered pathology associated with neurodegenerative diseases like PD. This study presents the analysis of the pathways involved in the downregulation of ACE2 following SARS-CoV-2 infection and its effect on PD progression. Through QIAGEN's Ingenuity Pathway Analysis (IPA), the study identified the NISP as a top-five canonical pathway/signaling pathway and SNCA as a top-five upstream regulator. Core Analysis was also conducted on the associated molecules between COVID-19 and SNCA to construct a network connectivity map. The Molecule Activity Predictor tool was used to simulate the infection of SARS-CoV-2 by downregulating IFNG, which leads to the predicted activation of SNCA, and subsequently PD, through a dataset of intermediary molecules. Downstream effect analysis was further used to quantify the downregulation of ACE2 on SNCA activation.
Collapse
Affiliation(s)
- Jonathan Zhang
- Institute of NeuroImmune Pharmacology, South Orange, NJ 07079, USA; (J.Z.); (M.B.); (S.B.)
- Department of Biological Sciences, Seton Hall University, South Orange, NJ 07079, USA
| | - Muhammed Bishir
- Institute of NeuroImmune Pharmacology, South Orange, NJ 07079, USA; (J.Z.); (M.B.); (S.B.)
- Department of Biological Sciences, Seton Hall University, South Orange, NJ 07079, USA
| | - Sharman Barbhuiya
- Institute of NeuroImmune Pharmacology, South Orange, NJ 07079, USA; (J.Z.); (M.B.); (S.B.)
- Department of Biological Sciences, Seton Hall University, South Orange, NJ 07079, USA
| | - Sulie L. Chang
- Institute of NeuroImmune Pharmacology, South Orange, NJ 07079, USA; (J.Z.); (M.B.); (S.B.)
- Department of Biological Sciences, Seton Hall University, South Orange, NJ 07079, USA
| |
Collapse
|
7
|
Emslander Q, Krey K, Hamad S, Maidl S, Oubraham L, Hesse J, Henrici A, Austen K, Mergner J, Grass V, Pichlmair A. MDM2 Influences ACE2 Stability and SARS-CoV-2 Uptake. Viruses 2023; 15:1763. [PMID: 37632105 PMCID: PMC10459000 DOI: 10.3390/v15081763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 07/20/2023] [Accepted: 08/07/2023] [Indexed: 08/27/2023] Open
Abstract
Angiotensin-converting enzyme 2 (ACE2) is the central entry receptor for SARS-CoV-2. However, surprisingly little is known about the effects of host regulators on ACE2 localization, expression, and the associated influence on SARS-CoV-2 infection. Here we identify that ACE2 expression levels are regulated by the E3 ligase MDM2 and that MDM2 levels indirectly influence infection with SARS-CoV-2. Genetic depletion of MDM2 elevated ACE2 expression levels, which strongly promoted infection with all SARS-CoV-2 isolates tested. SARS-CoV-2 spike-pseudotyped viruses and the uptake of non-replication-competent virus-like particles showed that MDM2 affects the viral uptake process. MDM2 ubiquitinates Lysine 788 of ACE2 to induce proteasomal degradation, and degradation of this residue led to higher ACE2 expression levels and superior virus particle uptake. Our study illustrates that cellular regulators of ACE2 stability, such as MDM2, play an important role in defining the infection capabilities of SARS-CoV-2.
Collapse
Affiliation(s)
- Quirin Emslander
- Institute of Virology, School of Medicine, Technical University of Munich (TUM), 81675 Munich, Germany (S.H.)
| | - Karsten Krey
- Institute of Virology, School of Medicine, Technical University of Munich (TUM), 81675 Munich, Germany (S.H.)
| | - Sabri Hamad
- Institute of Virology, School of Medicine, Technical University of Munich (TUM), 81675 Munich, Germany (S.H.)
| | - Susanne Maidl
- Institute of Virology, School of Medicine, Technical University of Munich (TUM), 81675 Munich, Germany (S.H.)
| | - Lila Oubraham
- Institute of Virology, School of Medicine, Technical University of Munich (TUM), 81675 Munich, Germany (S.H.)
| | - Joshua Hesse
- Institute of Virology, School of Medicine, Technical University of Munich (TUM), 81675 Munich, Germany (S.H.)
| | - Alexander Henrici
- Institute of Virology, School of Medicine, Technical University of Munich (TUM), 81675 Munich, Germany (S.H.)
| | - Katharina Austen
- Institute of Virology, School of Medicine, Technical University of Munich (TUM), 81675 Munich, Germany (S.H.)
| | - Julia Mergner
- BayBioMS@MRI—Bavarian Center for Biomolecular Mass Spectrometry at Klinikum Rechts der Isar, Technical University of Munich, 81675 Munich, Germany
| | - Vincent Grass
- Institute of Virology, School of Medicine, Technical University of Munich (TUM), 81675 Munich, Germany (S.H.)
| | - Andreas Pichlmair
- Institute of Virology, School of Medicine, Technical University of Munich (TUM), 81675 Munich, Germany (S.H.)
- German Centre for Infection Research (DZIF), Partner site Munich, 81675 Munich, Germany
- Center of Immunology of Viral Infection (CiViA), Aarhus University, 8000 Aarhus, Denmark
| |
Collapse
|
8
|
Boccatonda A, Campello E, Simion C, Simioni P. Long-term hypercoagulability, endotheliopathy and inflammation following acute SARS-CoV-2 infection. Expert Rev Hematol 2023; 16:1035-1048. [PMID: 38018136 DOI: 10.1080/17474086.2023.2288154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 11/22/2023] [Indexed: 11/30/2023]
Abstract
INTRODUCTION both symptomatic and asymptomatic SARS-CoV-2 infections - coined Coronavirus disease 2019 (COVID-19) - have been linked to a higher risk of cardiovascular events after recovery. AREAS COVERED our review aims to summarize the latest evidence on the increased thrombotic and cardiovascular risk in recovered COVID-19 patients and to examine the pathophysiological mechanisms underlying the interplay among endothelial dysfunction, inflammatory response and coagulation in long-COVID. We performed a systematic search of studies on hypercoagulability, endothelial dysfunction and inflammation after SARS-CoV-2 infection. EXPERT OPINION endothelial dysfunction is a major pathophysiological mechanism responsible for most clinical manifestations in COVID-19. The pathological activation of endothelial cells by a virus infection results in a pro-adhesive and chemokine-secreting phenotype, which in turn promotes the recruitment of circulating leukocytes. Cardiovascular events after COVID-19 appear to be related to persistent immune dysregulation. Patients with long-lasting symptoms display higher amounts of proinflammatory molecules such as tumor necrosis factor-α, interferon γ and interleukins 2 and 6. Immune dysregulation can trigger the activation of the coagulation pathway. The formation of extensive microclots in vivo, both during acute COVID-19 and in long-COVID-19, appears to be a relevant mechanism responsible for persistent symptoms and cardiovascular events.
Collapse
Affiliation(s)
- Andrea Boccatonda
- Internal Medicine, Bentivoglio Hospital, AUSL Bologna, Bentivoglio, Italy
| | - Elena Campello
- General Medicine and Thrombotic and Hemorrhagic Diseases Unit, Department of Medicine, University Hospital of Padova, Padova, Italy
| | - Chiara Simion
- General Medicine and Thrombotic and Hemorrhagic Diseases Unit, Department of Medicine, University Hospital of Padova, Padova, Italy
| | - Paolo Simioni
- General Medicine and Thrombotic and Hemorrhagic Diseases Unit, Department of Medicine, University Hospital of Padova, Padova, Italy
| |
Collapse
|
9
|
Importance of ACE2 for SARS-CoV-2 Infection of Kidney Cells. Biomolecules 2023; 13:biom13030472. [PMID: 36979408 PMCID: PMC10046276 DOI: 10.3390/biom13030472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 02/27/2023] [Accepted: 03/01/2023] [Indexed: 03/08/2023] Open
Abstract
In late 2019, the novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) as the causative agent of coronavirus disease 2019 (COVID-19) emerged in China and spread rapidly around the world, causing an ongoing pandemic of global concern. COVID-19 proceeds with moderate symptoms in most patients, whereas others experience serious respiratory illness that requires intensive care treatment and may end in death. The severity of COVID-19 is linked to several risk factors including male sex, comorbidities, and advanced age. Apart from respiratory complications, further impairments by COVID-19 affecting other tissues of the human body are observed. In this respect, the human kidney is one of the most frequently affected extrapulmonary organs and acute kidney injury (AKI) is known as a direct or indirect complication of SARS-CoV-2 infection. The aim of this work was to investigate the importance of the protein angiotensin-converting enzyme 2 (ACE2) for a possible cell entry of SARS-CoV-2 into human kidney cells. First, the expression of the cellular receptor ACE2 was demonstrated to be decisive for viral SARS-CoV-2 cell entry in human AB8 podocytes, whereas the presence of the transmembrane protease serine 2 (TMPRSS2) was dispensable. Moreover, the ACE2 protein amount was well detectable by mass spectrometry analysis in human kidneys, while TMPRSS2 could be detected only in a few samples. Additionally, a negative correlation of the ACE2 protein abundance to male sex and elderly aged females in human kidney tissues was demonstrated in this work. Last, the possibility of a direct infection of kidney tubular renal structures by SARS-CoV-2 was demonstrated.
Collapse
|
10
|
El-Sayed Marei Y, Abdallah Bayoumy A, Mohamed Abulazm Nassar H, Mansour B, Bakeir Hamady A. The Relation between ACE Gene Polymorphism and the Severity of COVID-19 Infection. Int J Microbiol 2023; 2023:4540287. [PMID: 36644496 PMCID: PMC9833908 DOI: 10.1155/2023/4540287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 11/25/2022] [Accepted: 12/09/2022] [Indexed: 01/06/2023] Open
Abstract
Introduction The coronavirus disease 2019 (COVID-19) pandemic, which emerged in China at the end of 2019, rapidly spread worldwide. The angiotensin-converting enzyme (ACE) gene contains an insertion/deletion (I/D) polymorphism that leads to a higher serum ACE level which is associated with several diseases and also with a high mortality rate in SARS. Therefore, this study aimed at assessing the association between ACE gene polymorphism and the risk and severity of COVID-19 disease in patients. Methodology. Forty-five SARS-CoV-2 infected patients and another random control group of 45 healthy individuals were included. The detection of ACE I/D gene polymorphism in both groups was done by PCR. Results 53% of infected patients with SARS-CoV-2 had an ACE deletion/deletion genotype (D/D), 27% had an ACE deletion/insertion genotype (D/I), and 20% had an ACE insertion/insertion genotype (I/I). On the one hand, the D/D variant was significantly detected in the COVID-19 patients compared to the control subjects, whereas the I/I variant was significantly detected in the control subjects compared to the COVID-19 patients (p = 0.004). The D/D variant subgroup showed the lowest lymphocytic count compared to the D/I or I/I subgroups. In addition, the C-reactive protein was significantly higher and the oxygen saturation was significantly lower in patients with the D/D allele compared to the other subgroups. Conclusions ACE gene polymorphism, particularly the DD genotype, was observed to affect the severity of COVID-19 infection.
Collapse
Affiliation(s)
- Yara El-Sayed Marei
- Medical Microbiology and Immunology Department, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | - Ahmed Abdallah Bayoumy
- Chest Unit Internal Medicine Department, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | | | - Bassam Mansour
- Infection and Endemic Disease Department, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | - Asmaa Bakeir Hamady
- Medical Microbiology and Immunology Department, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| |
Collapse
|
11
|
Golpour-Hamedani S, Pourmasoumi M, Askari G, Bagherniya M, Majeed M, Guest PC, Sahebkar A. Antiviral Mechanisms of Curcumin and Its Derivatives in Prevention and Treatment of COVID-19: A Review. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1412:397-411. [PMID: 37378779 DOI: 10.1007/978-3-031-28012-2_21] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 06/29/2023]
Abstract
The COVID-19 pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has now plagued the world for almost 3 years. Although vaccines are now available, the severity of the pandemic and the current dearth of approved effective medications have prompted the need for novel treatment approaches. Curcumin, as a food nutraceutical with anti-inflammatory and antioxidant effects, is now under consideration for the prevention and treatment of COVID-19. Curcumin has been demonstrated to retard the entrance of SARS-CoV-2 into cells, interfere with its proliferation inside cells, and curb the hyperinflammatory state caused by the virus by modulating immune system regulators, minimizing the cytokine storm effect, and modulating the renin-angiotensin system. This chapter discusses the role of curcumin and its derivatives in the prevention and treatment of COVID-19 infection, considering the molecular mechanisms involved. It will also focus on the molecular and cellular profiling techniques as essential tools in this research, as these can be used in the identification and development of new biomarkers, drug targets, and therapeutic approaches for improved patient care.
Collapse
Affiliation(s)
- Sahar Golpour-Hamedani
- Department of Community Nutrition, School of Nutrition and Food Science, Isfahan University of Medical Science, Isfahan, Iran
| | - Makan Pourmasoumi
- Gastrointestinal & Liver Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran
| | - Gholamreza Askari
- Nutrition and Food Security Research Center and Department of Community Nutrition, School of Nutrition and Food Science, Isfahan University of Medical Sciences, Isfahan, Iran.
- Anesthesia and Critical Care Research Center, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Mohammad Bagherniya
- Nutrition and Food Security Research Center and Department of Community Nutrition, School of Nutrition and Food Science, Isfahan University of Medical Sciences, Isfahan, Iran
- Anesthesia and Critical Care Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | | | - Paul C Guest
- Department of Psychiatry, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
- Laboratory of Translational Psychiatry, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, Brazil
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- School of Medicine, The University of Western Australia, Perth, Australia
| |
Collapse
|
12
|
Dieter C, de Almeida Brondani L, Lemos NE, Schaeffer AF, Zanotto C, Ramos DT, Girardi E, Pellenz FM, Camargo JL, Moresco KS, da Silva LL, Aubin MR, de Oliveira MS, Rech TH, Canani LH, Gerchman F, Leitão CB, Crispim D. Polymorphisms in ACE1, TMPRSS2, IFIH1, IFNAR2, and TYK2 Genes Are Associated with Worse Clinical Outcomes in COVID-19. Genes (Basel) 2022; 14:genes14010029. [PMID: 36672770 PMCID: PMC9858252 DOI: 10.3390/genes14010029] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 11/29/2022] [Accepted: 12/10/2022] [Indexed: 12/25/2022] Open
Abstract
Although advanced age, male sex, and some comorbidities impact the clinical course of COVID-19, these factors only partially explain the inter-individual variability in disease severity. Some studies have shown that genetic polymorphisms contribute to COVID-19 severity; however, the results are inconclusive. Thus, we investigated the association between polymorphisms in ACE1, ACE2, DPP9, IFIH1, IFNAR2, IFNL4, TLR3, TMPRSS2, and TYK2 and the clinical course of COVID-19. A total of 694 patients with COVID-19 were categorized as: (1) ward inpatients (moderate symptoms) or patients admitted at the intensive care unit (ICU; severe symptoms); and (2) survivors or non-survivors. In females, the rs1990760/IFIH1 T/T genotype was associated with risk of ICU admission and death. Moreover, the rs1799752/ACE1 Ins and rs12329760/TMPRSS2 T alleles were associated with risk of ICU admission. In non-white patients, the rs2236757/IFNAR2 A/A genotype was associated with risk of ICU admission, while the rs1799752/ACE1 Ins/Ins genotype, rs2236757/IFNAR2 A/A genotype, and rs12329760/TMPRSS2 T allele were associated with risk of death. Moreover, some of the analyzed polymorphisms interact in the risk of worse COVID-19 outcomes. In conclusion, this study shows an association of rs1799752/ACE1, rs1990760/IFIH1, rs2236757/IFNAR2, rs12329760/TMPRSS2, and rs2304256/TYK2 polymorphisms with worse COVID-19 outcomes, especially among female and non-white patients.
Collapse
Affiliation(s)
- Cristine Dieter
- Endocrine Division, Hospital de Clínicas de Porto Alegre, Porto Alegre 90035-903, RS, Brazil
- Post-Graduate Program in Medical Sciences, Endocrinology, Department of Internal Medicine, Faculty of Medicine, Universidade Federal do Rio Grande do Sul, Porto Alegre 91501-970, RS, Brazil
| | - Leticia de Almeida Brondani
- Endocrine Division, Hospital de Clínicas de Porto Alegre, Porto Alegre 90035-903, RS, Brazil
- Experimental Research Center, Hospital de Clínicas de Porto Alegre, Porto Alegre 90035-903, RS, Brazil
| | - Natália Emerim Lemos
- Endocrine Division, Hospital de Clínicas de Porto Alegre, Porto Alegre 90035-903, RS, Brazil
| | - Ariell Freires Schaeffer
- Post-Graduate Program in Medical Sciences, Endocrinology, Department of Internal Medicine, Faculty of Medicine, Universidade Federal do Rio Grande do Sul, Porto Alegre 91501-970, RS, Brazil
| | - Caroline Zanotto
- Endocrine Division, Hospital de Clínicas de Porto Alegre, Porto Alegre 90035-903, RS, Brazil
| | - Denise Taurino Ramos
- Endocrine Division, Hospital de Clínicas de Porto Alegre, Porto Alegre 90035-903, RS, Brazil
| | - Eliandra Girardi
- Endocrine Division, Hospital de Clínicas de Porto Alegre, Porto Alegre 90035-903, RS, Brazil
| | - Felipe Mateus Pellenz
- Endocrine Division, Hospital de Clínicas de Porto Alegre, Porto Alegre 90035-903, RS, Brazil
- Post-Graduate Program in Medical Sciences, Endocrinology, Department of Internal Medicine, Faculty of Medicine, Universidade Federal do Rio Grande do Sul, Porto Alegre 91501-970, RS, Brazil
| | - Joiza Lins Camargo
- Post-Graduate Program in Medical Sciences, Endocrinology, Department of Internal Medicine, Faculty of Medicine, Universidade Federal do Rio Grande do Sul, Porto Alegre 91501-970, RS, Brazil
- Experimental Research Center, Hospital de Clínicas de Porto Alegre, Porto Alegre 90035-903, RS, Brazil
- Diabetes and Metabolism Group, Centro de Pesquisa Clínica, Hospital de Clínicas de Porto Alegre, Porto Alegre 90035-903, RS, Brazil
| | - Karla Suzana Moresco
- Campus Realeza, Universidade Federal da Fronteira Sul, Realeza 85770-000, PR, Brazil
| | - Lucas Lima da Silva
- Endocrine Division, Hospital de Clínicas de Porto Alegre, Porto Alegre 90035-903, RS, Brazil
| | - Mariana Rauback Aubin
- Endocrine Division, Hospital de Clínicas de Porto Alegre, Porto Alegre 90035-903, RS, Brazil
| | - Mayara Souza de Oliveira
- Endocrine Division, Hospital de Clínicas de Porto Alegre, Porto Alegre 90035-903, RS, Brazil
- Post-Graduate Program in Medical Sciences, Endocrinology, Department of Internal Medicine, Faculty of Medicine, Universidade Federal do Rio Grande do Sul, Porto Alegre 91501-970, RS, Brazil
| | - Tatiana Helena Rech
- Post-Graduate Program in Medical Sciences, Endocrinology, Department of Internal Medicine, Faculty of Medicine, Universidade Federal do Rio Grande do Sul, Porto Alegre 91501-970, RS, Brazil
- Diabetes and Metabolism Group, Centro de Pesquisa Clínica, Hospital de Clínicas de Porto Alegre, Porto Alegre 90035-903, RS, Brazil
| | - Luís Henrique Canani
- Endocrine Division, Hospital de Clínicas de Porto Alegre, Porto Alegre 90035-903, RS, Brazil
- Post-Graduate Program in Medical Sciences, Endocrinology, Department of Internal Medicine, Faculty of Medicine, Universidade Federal do Rio Grande do Sul, Porto Alegre 91501-970, RS, Brazil
- Diabetes and Metabolism Group, Centro de Pesquisa Clínica, Hospital de Clínicas de Porto Alegre, Porto Alegre 90035-903, RS, Brazil
| | - Fernando Gerchman
- Endocrine Division, Hospital de Clínicas de Porto Alegre, Porto Alegre 90035-903, RS, Brazil
- Post-Graduate Program in Medical Sciences, Endocrinology, Department of Internal Medicine, Faculty of Medicine, Universidade Federal do Rio Grande do Sul, Porto Alegre 91501-970, RS, Brazil
- Diabetes and Metabolism Group, Centro de Pesquisa Clínica, Hospital de Clínicas de Porto Alegre, Porto Alegre 90035-903, RS, Brazil
| | - Cristiane Bauermann Leitão
- Endocrine Division, Hospital de Clínicas de Porto Alegre, Porto Alegre 90035-903, RS, Brazil
- Post-Graduate Program in Medical Sciences, Endocrinology, Department of Internal Medicine, Faculty of Medicine, Universidade Federal do Rio Grande do Sul, Porto Alegre 91501-970, RS, Brazil
- Diabetes and Metabolism Group, Centro de Pesquisa Clínica, Hospital de Clínicas de Porto Alegre, Porto Alegre 90035-903, RS, Brazil
| | - Daisy Crispim
- Endocrine Division, Hospital de Clínicas de Porto Alegre, Porto Alegre 90035-903, RS, Brazil
- Post-Graduate Program in Medical Sciences, Endocrinology, Department of Internal Medicine, Faculty of Medicine, Universidade Federal do Rio Grande do Sul, Porto Alegre 91501-970, RS, Brazil
- Diabetes and Metabolism Group, Centro de Pesquisa Clínica, Hospital de Clínicas de Porto Alegre, Porto Alegre 90035-903, RS, Brazil
- Correspondence:
| |
Collapse
|
13
|
D'Ardes D, Tana C, Salzmann A, Ricci F, Guagnano MT, Giamberardino MA, Cipollone F. Ultrasound assessment of SARS-CoV-2 pneumonia: a literature review for the primary care physician. Ann Med 2022; 54:1140-1149. [PMID: 35465821 PMCID: PMC9045761 DOI: 10.1080/07853890.2022.2067896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
The SARS-CoV-2 pandemic is considered one of the most critical global health emergencies in the last century. The diagnostic approach to the novel coronavirus disease (COVID-19) and its possible complications through a point-of-care-ultrasound (POCUS) evaluation could represent a good solution in the primary care setting. POCUS is a non-invasive technique that can be used outside hospitals to screen COVID-19 patients and their complications safely. Moreover, it offers several applications of diagnostic evaluation not only on lung parenchyma but also to search disease complications, such as the cardiovascular system, even at the patients' home. This narrative review aims to analyse the literature and provide data to primary care physicians engaged in monitoring and treating patients with SARS-CoV-2 infection. Key MessagesPOCUS is an important tool for the diagnostic approach in the primary care setting already before the start of the SARS-CoV-2 pandemic.Portable devices are useful in monitoring the clinical evolution of patients with infection from SARS-CoV-2 at home.The ultrasonographic features can help the general practice physicians to evaluate the presence of lung involvement and to diagnose complications from the SARS-CoV-2 infection involving districts such as the cardiovascular system.
Collapse
Affiliation(s)
- Damiano D'Ardes
- "Clinica Medica" Institute, "SS. Annunziata" Hospital of Chieti, Department of Medicine and Aging Sciences, "G. D'Annunzio", University of Chieti, Pescara, Italy
| | - Claudio Tana
- "Geriatric and COVID-19 Unit", "SS. Annunziata" Hospital of Chieti, "G. D'Annunzio" University of Chieti, Pescara, Italy
| | - Alessandro Salzmann
- "Clinica Medica" Institute, "SS. Annunziata" Hospital of Chieti, Department of Medicine and Aging Sciences, "G. D'Annunzio", University of Chieti, Pescara, Italy
| | - Fabrizio Ricci
- Department of Neuroscience, Imaging and Clinical Sciences, "G. d'Annunzio" University, Chieti, Italy
| | - Maria Teresa Guagnano
- "Clinica Medica" Institute, "SS. Annunziata" Hospital of Chieti, Department of Medicine and Aging Sciences, "G. D'Annunzio", University of Chieti, Pescara, Italy
| | - Maria Adele Giamberardino
- "Geriatric and COVID-19 Unit", "SS. Annunziata" Hospital of Chieti, "G. D'Annunzio" University of Chieti, Pescara, Italy
| | - Francesco Cipollone
- "Clinica Medica" Institute, "SS. Annunziata" Hospital of Chieti, Department of Medicine and Aging Sciences, "G. D'Annunzio", University of Chieti, Pescara, Italy
| |
Collapse
|
14
|
Braga-Paz I, Ferreira de Araújo JL, Alves HJ, de Ávila RE, Resende GG, Teixeira MM, de Aguiar RS, de Souza RP, Bahia D. Negative correlation between ACE2 gene expression levels and loss of taste in a cohort of COVID-19 hospitalized patients: New clues to long-term cognitive disorders. Front Cell Infect Microbiol 2022; 12:905757. [PMID: 36250059 PMCID: PMC9556632 DOI: 10.3389/fcimb.2022.905757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Accepted: 08/17/2022] [Indexed: 11/13/2022] Open
Abstract
In early 2020, one of the most prevalent symptoms of SARS-CoV-2 infection was the loss of smell (anosmia), found in 60-70% of all cases. Anosmia used to occur early, concomitantly with other symptoms, and often persisted after recovery for an extended period, sometimes for months. In addition to smell disturbance, COVID-19 has also been associated with loss of taste (ageusia). The latest research suggests that SARS-CoV-2 could spread from the respiratory system to the brain through receptors in sustentacular cells localized to the olfactory epithelium. The virus invades human cells via the obligatory receptor, angiotensin-converting enzyme II (ACE2), and a priming protease, TMPRSS2, facilitating viral penetration. There is an abundant expression of both ACE2 and TMPRSS2 in sustentacular cells. In this study, we evaluated 102 COVID-19 hospitalized patients, of which 17.60% presented anosmia and 9.80% ageusia. ACE1, ACE2, and TMPRSS2 gene expression levels in nasopharyngeal tissue were obtained by RT-qPCR and measured using ΔCT analysis. ACE1 Alu287bp association was also evaluated. Logistic regression models were generated to estimate the effects of variables on ageusia and anosmia Association of ACE2 expression levels with ageusia. was observed (OR: 1.35; 95% CI: 1.098-1.775); however, no association was observed between TMPRSS2 and ACE1 expression levels and ageusia. No association was observed among the three genes and anosmia, and the Alu287bp polymorphism was not associated with any of the outcomes. Lastly, we discuss whetherthere is a bridge linking these initial symptoms, including molecular factors, to long-term COVID-19 health consequences such as cognitive dysfunctions.
Collapse
Affiliation(s)
- Isabela Braga-Paz
- Departamento de Genética, Ecologia e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - João Locke Ferreira de Araújo
- Departamento de Genética, Ecologia e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Hugo José Alves
- Departamento de Genética, Ecologia e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | | | - Gustavo Gomes Resende
- Hospital das Clínicas, Universidade Federal de Minas Gerais (HC-UFMG/EBSERH), Belo Horizonte, Brazil
| | - Mauro Martins Teixeira
- Departamento de Bioquimica e imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Renato Santana de Aguiar
- Departamento de Genética, Ecologia e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
- Instituto D’Or de Pesquisa e Ensino, Instituto D'OR (IDOR), Rio de Janeiro, Brazil
| | - Renan Pedra de Souza
- Departamento de Genética, Ecologia e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
- *Correspondence: Renan Pedra de Souza, ; Diana Bahia,
| | - Diana Bahia
- Departamento de Genética, Ecologia e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
- *Correspondence: Renan Pedra de Souza, ; Diana Bahia,
| |
Collapse
|
15
|
Faridzadeh A, Mahmoudi M, Ghaffarpour S, Zamani MS, Hoseinzadeh A, Naghizadeh MM, Ghazanfari T. The role of ACE1 I/D and ACE2 polymorphism in the outcome of Iranian COVID-19 patients: A case-control study. Front Genet 2022; 13:955965. [PMID: 36134024 PMCID: PMC9483011 DOI: 10.3389/fgene.2022.955965] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Accepted: 08/08/2022] [Indexed: 11/13/2022] Open
Abstract
Background: Since the beginning of the pandemic of coronavirus disease 2019 (COVID-19), many countries have experienced a considerable number of COVID-19 cases and deaths. The etiology of a broad spectrum of symptoms is still debated. Host genetic variants might also significantly influence the outcome of the disease. This study aimed to evaluate the association of angiotensin-converting enzyme (ACE1) gene Insertion/Deletion (I/D) polymorphism (rs1799752) and ACE2 gene rs1978124 single nucleotide polymorphism with the COVID-19 severity. Methods: This study was conducted on 470 COVID-19 patients and a control group of 56 healthy individuals across several major cities in Iran. The blood sample and clinical data were collected from the participants, and their ACE1 I/D and ACE2 rs1978124 polymorphisms were determined using polymerase chain reaction and PCR-RFLP, respectively. Serum levels of C-reactive protein (CRP), interleukin 6 (IL-6), and ACE1 were measured in the blood samples. Results: We found that the ACE1 DD genotype frequency was inversely correlated with the risk of intubation (p = 0.017) and mortality in COVID-19 patients (p = 0.049). Even after adjustment, logistic regression demonstrated that this significant inverse association remained constant for the above variables at odds ratios of (OR) = 0.35 and Odds Ratio = 0.49, respectively. Also, in the expired (p = 0.042) and intubated (p = 0.048) groups with II + ID genotypes, the mean level of CRP was significantly higher than in the DD genotype group. Furthermore, in both intubated and expired groups, the mean serum level of ACE1 was higher compared with non-intubated and survived groups with II or II + ID genotypes. The results also indicated that ACE2 rs1978124 TT + CT genotypes in females have a significant positive role in susceptibility to COVID-19; however, in females, the TT + CT genotypes had a protective effect (OR = 0.098) against the severity of COVID-19. Conclusion: These findings suggest that ACE1 I/D and ACE2 rs1978124 polymorphism could potentially influence the outcome of COVID-19 in the Iranian population.
Collapse
Affiliation(s)
- Arezoo Faridzadeh
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Immunology and Allergy, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahmoud Mahmoudi
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Immunology and Allergy, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- *Correspondence: Mahmoud Mahmoudi, ; Tooba Ghazanfari,
| | - Sara Ghaffarpour
- Immunoregulation Research Center, Shahed University, Tehran, Iran
| | | | - Akram Hoseinzadeh
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Immunology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Tooba Ghazanfari
- Immunoregulation Research Center, Shahed University, Tehran, Iran
- Department of Immunology, Shahed University, Tehran, Iran
- *Correspondence: Mahmoud Mahmoudi, ; Tooba Ghazanfari,
| |
Collapse
|
16
|
Dambha-Miller H, Hinton W, Wilcox CR, Lemanska A, Joy M, Feher M, Stuart B, de Lusignan S, Hippisley-Cox J, Griffin S. Mortality from angiotensin-converting enzyme-inhibitors and angiotensin receptor blockers in people infected with COVID-19: a cohort study of 3.7 million people. Fam Pract 2022; 40:330-337. [PMID: 36003039 PMCID: PMC9452130 DOI: 10.1093/fampra/cmac094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Concerns have been raised that angiotensin-converting enzyme-inhibitors (ACE-I) and angiotensin receptor blockers (ARBs) might facilitate transmission of severe acute respiratory syndrome coronavirus 2 leading to more severe coronavirus disease (COVID-19) disease and an increased risk of mortality. We aimed to investigate the association between ACE-I/ARB treatment and risk of death amongst people with COVID-19 in the first 6 months of the pandemic. METHODS We identified a cohort of adults diagnosed with either confirmed or probable COVID-19 (from 1 January to 21 June 2020) using computerized medical records from the Oxford-Royal College of General Practitioners (RCGP) Research and Surveillance Centre (RSC) primary care database. This comprised 465 general practices in England, United Kingdom with a nationally representative population of 3.7 million people. We constructed mixed-effects logistic regression models to quantify the association between ACE-I/ARBs and all-cause mortality among people with COVID-19, adjusted for sociodemographic factors, comorbidities, concurrent medication, smoking status, practice clustering, and household number. RESULTS There were 9,586 COVID-19 cases in the sample and 1,463 (15.3%) died during the study period between 1 January 2020 and 21 June 2020. In adjusted analysis ACE-I and ARBs were not associated with all-cause mortality (adjusted odds ratio [OR] 1.02, 95% confidence interval [CI] 0.85-1.21 and OR 0.84, 95% CI 0.67-1.07, respectively). CONCLUSION Use of ACE-I/ARB, which are commonly used drugs, did not alter the odds of all-cause mortality amongst people diagnosed with COVID-19. Our findings should inform patient and prescriber decisions concerning continued use of these medications during the pandemic.
Collapse
Affiliation(s)
- Hajira Dambha-Miller
- Division of Primary Care and Population Health, University of Southampton, Southampton, United Kingdom
| | - William Hinton
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, United Kingdom
| | - Christopher R Wilcox
- Division of Primary Care and Population Health, University of Southampton, Southampton, United Kingdom
| | - Agnieszka Lemanska
- Department of Clinical and Experimental Medicine, School of Health Sciences, University of Surrey, Surrey, United Kingdom
| | - Mark Joy
- Department of Clinical and Experimental Medicine, School of Health Sciences, University of Surrey, Surrey, United Kingdom
| | - Michael Feher
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, United Kingdom
| | - Beth Stuart
- Division of Primary Care and Population Health, University of Southampton, Southampton, United Kingdom
| | - Simon de Lusignan
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, United Kingdom
| | - Julia Hippisley-Cox
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, United Kingdom
| | - Simon Griffin
- Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom.,MRC Epidemiology Unit, School of Clinical Medicine, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
17
|
Hussain A, Singh L, McAlister III J, Jo Y, Makaryan TT, Hussain S, Trenschel RW, Kesselman MM. Serum Procalcitonin as a Predictive Biomarker in COVID-19: A Retrospective Cohort Analysis. Cureus 2022; 14:e27816. [PMID: 36106293 PMCID: PMC9452059 DOI: 10.7759/cureus.27816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 08/09/2022] [Indexed: 01/08/2023] Open
|
18
|
Govender N, Khaliq O, Moodley J, Naicker T. Unravelling the Mechanistic Role of ACE2 and TMPRSS2 in Hypertension: A Risk Factor for COVID-19. Curr Hypertens Rev 2022; 18:130-137. [PMID: 36508271 DOI: 10.2174/1573402118666220816090809] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 05/25/2022] [Accepted: 05/30/2022] [Indexed: 01/27/2023]
Abstract
BACKGROUND This review explores the mechanistic action of angiotensin-converting enzyme- 2 (ACE2) and transmembrane protease serine 2 (TMPRSS2) in the renin-angiotensinaldosterone system (RAAS) that predisposes hypertensive patients to the adverse outcome of severe COVID-19. METHODS AND RESULTS Entry of SARS-CoV-2 into the host cell via ACE2 disrupts the RAAS system, creating an imbalance between ACE and ACE2, with an increased inflammatory response, leading to hypertension (HTN), pulmonary vasoconstriction and acute respiratory distress. SARSCoV- 2 may also predispose infected individuals with existing HTN to a greater risk of severe COVID-19 complications. In the duality of COVID-19 and HTN, the imbalance of ACE and ACE2 results in an elevation of AngII and a decrease in Ang (1-7), a hyperinflammatory response and endothelial dysfunction. Endothelial dysfunction is the main factor predisposing hypertensive patients to severe COVID-19 and vice-versa. CONCLUSION Despite the increase in ACE2 expression in hypertensive SARS-CoV-2 infected patients, ARBs/ACE inhibitors do not influence their severity and clinical outcomes, implicating continued usage. Future large-scale clinical trials are warranted to further elucidate the association between HTN and SARS-CoV-2 infection and the use of ARBs/ACEIs in SARS-CoV-2 hypertensive patients.
Collapse
Affiliation(s)
- Nalini Govender
- Department of Basic Medical Sciences, Faculty of Health Sciences, Durban University of Technology, Durban 4001, South Africa
| | - Olive Khaliq
- The Department of Paediatrics and Child Health, Faculty of Health Sciences, The University of the Free State, Bloemfontein 9300, South Africa
| | - Jagidesa Moodley
- Women's Health and HIV Research Group, Department of Obstetrics and Gynaecology, School of Clinical Medicine, College of Health Sciences, University of KwaZulu-Natal, Durban 4001, South Africa
| | - Thajasvarie Naicker
- Optics & Imaging Centre, Doris Duke Medical Research Institute, College of Health Sciences, University of KwaZulu-Natal, Durban 4001, South Africa
| |
Collapse
|
19
|
Boccatonda A, D’Ardes D, Rossi I, Grignaschi A, Lanotte A, Cipollone F, Guagnano MT, Giostra F. Platelet Count in Patients with SARS-CoV-2 Infection: A Prognostic Factor in COVID-19. J Clin Med 2022; 11:4112. [PMID: 35887877 PMCID: PMC9325150 DOI: 10.3390/jcm11144112] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 07/13/2022] [Accepted: 07/14/2022] [Indexed: 12/18/2022] Open
Abstract
COVID-19 patients may manifest thrombocytopenia and some of these patients succumb to infection due to coagulopathy. The aim of our study was to examine platelet count values in patients infected with SARS-CoV-2, comparing them to a control group consisting of non-COVID-19 patients. Moreover, we evaluated the correlation between the platelet value and the respiratory alteration parameters and the outcome (hospitalization and mortality) in COVID-19 patients. The mean platelet values (×109/L) differed between patients with positive or negative SARS-CoV-2 swabs (242.1 ± 92.1 in SARS-CoV-2 negative vs. 215.2 ± 82.8 in COVID-19 patients, p < 0.001). In COVID-19 patients, the platelet count correlated with the A-aO2 gradient (p = 0.001, rho = −0.149), with its increase over the expected (p = 0.013; rho = −0.115), with the PaO2 values (p = 0.036; rho = 0.093), with the PCO2 values (p = 0.003; rho = 0.134) and with the pH values (p = 0.016; rho = −0.108). In COVID-19 negative patients, the platelet values correlated only with the A-aO2 gradient: (p = 0.028; rho = −0.101). Patients discharged from emergency department had a mean platelet value of 234.3 ± 68.7, those hospitalized in ordinary wards had a mean value of 204.3 ± 82.5 and in patients admitted to sub-intensive/intensive care, the mean value was 201.7 ± 75.1. In COVID-19 patients, the survivors had an average platelet value at entry to the emergency department of 220.1 ± 81.4, while that of those who died was 206.4 ± 87.7. Our data confirm that SARS-CoV-2 infection may induce thrombocytopenia, and that the reduction in platelet counts could be correlated with the main blood gas parameters and with clinical outcome; as a consequence, platelet count could be an important prognostic factor to evaluate and stratify COVID-19 patients.
Collapse
Affiliation(s)
- Andrea Boccatonda
- Emergency Department, IRCCS Azienda Ospedaliero, Universitaria di Bologna, 40138 Bologna, Italy; (A.B.); (A.G.); (A.L.); (F.G.)
| | - Damiano D’Ardes
- Institute of “Clinica Medica”, Department of Medicine and Aging Science, “G. D’Annunzio” University of Chieti, Vestini Road, 66100 Chieti, Italy; (I.R.); (F.C.); (M.T.G.)
| | - Ilaria Rossi
- Institute of “Clinica Medica”, Department of Medicine and Aging Science, “G. D’Annunzio” University of Chieti, Vestini Road, 66100 Chieti, Italy; (I.R.); (F.C.); (M.T.G.)
| | - Alice Grignaschi
- Emergency Department, IRCCS Azienda Ospedaliero, Universitaria di Bologna, 40138 Bologna, Italy; (A.B.); (A.G.); (A.L.); (F.G.)
| | - Antonella Lanotte
- Emergency Department, IRCCS Azienda Ospedaliero, Universitaria di Bologna, 40138 Bologna, Italy; (A.B.); (A.G.); (A.L.); (F.G.)
| | - Francesco Cipollone
- Institute of “Clinica Medica”, Department of Medicine and Aging Science, “G. D’Annunzio” University of Chieti, Vestini Road, 66100 Chieti, Italy; (I.R.); (F.C.); (M.T.G.)
| | - Maria Teresa Guagnano
- Institute of “Clinica Medica”, Department of Medicine and Aging Science, “G. D’Annunzio” University of Chieti, Vestini Road, 66100 Chieti, Italy; (I.R.); (F.C.); (M.T.G.)
| | - Fabrizio Giostra
- Emergency Department, IRCCS Azienda Ospedaliero, Universitaria di Bologna, 40138 Bologna, Italy; (A.B.); (A.G.); (A.L.); (F.G.)
| |
Collapse
|
20
|
Khazaal S, Harb J, Rima M, Annweiler C, Wu Y, Cao Z, Abi Khattar Z, Legros C, Kovacic H, Fajloun Z, Sabatier JM. The Pathophysiology of Long COVID throughout the Renin-Angiotensin System. Molecules 2022; 27:2903. [PMID: 35566253 PMCID: PMC9101946 DOI: 10.3390/molecules27092903] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 04/28/2022] [Accepted: 04/29/2022] [Indexed: 01/08/2023] Open
Abstract
COVID-19 has expanded across the world since its discovery in Wuhan (China) and has had a significant impact on people's lives and health. Long COVID is a term coined by the World Health Organization (WHO) to describe a variety of persistent symptoms after acute SARS-CoV-2 infection. Long COVID has been demonstrated to affect various SARS-CoV-2-infected persons, independently of the acute disease severity. The symptoms of long COVID, like acute COVID-19, consist in the set of damage to various organs and systems such as the respiratory, cardiovascular, neurological, endocrine, urinary, and immune systems. Fatigue, dyspnea, cardiac abnormalities, cognitive and attention impairments, sleep disturbances, post-traumatic stress disorder, muscle pain, concentration problems, and headache were all reported as symptoms of long COVID. At the molecular level, the renin-angiotensin system (RAS) is heavily involved in the pathogenesis of this illness, much as it is in the acute phase of the viral infection. In this review, we summarize the impact of long COVID on several organs and tissues, with a special focus on the significance of the RAS in the disease pathogenesis. Long COVID risk factors and potential therapy approaches are also explored.
Collapse
Affiliation(s)
- Shaymaa Khazaal
- Faculty of Sciences 3, Department of Biology, Lebanese University, Campus Michel Slayman Ras Maska, Tripoli P.O. Box 45061, Lebanon;
| | - Julien Harb
- Faculty of Medicine and Medical Sciences, University of Balamand, Dekouene Campus, Sin El Fil P.O. Box 55251, Lebanon;
| | - Mohamad Rima
- Laboratory of Applied Biotechnology (LBA3B), Azm Center for Research in Biotechnology and Its Applications, EDST, Lebanese University, Tripoli P.O. Box 45061, Lebanon;
| | - Cédric Annweiler
- Department of Geriatric Medicine and Memory Clinic, Research Center on Autonomy and Longevity, University Hospital & Laboratoire de Psychologie des Pays de la Loire, LPPL EA 4638, SFR Confluences, University of Angers, 44312 Angers, France;
| | - Yingliang Wu
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan 430072, China; (Y.W.); (Z.C.)
| | - Zhijian Cao
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan 430072, China; (Y.W.); (Z.C.)
| | - Ziad Abi Khattar
- Laboratory of Georesources, Geosciences and Environment (L2GE), Microbiology/Tox-Ecotoxicology Team, Faculty of Sciences 2, Lebanese University, Campus Fanar, Jdeidet El-Matn, Beirut P.O. Box 90656, Lebanon;
| | - Christian Legros
- INSERM, CNRS, MITOVASC, Team 2 CarMe, SFR ICAT, University of Angers, 49000, France;
| | - Hervé Kovacic
- Institut de Neurophysiopathologie (INP), Aix-Marseille Université CNRS, 13385 Marseille, France;
| | - Ziad Fajloun
- Faculty of Sciences 3, Department of Biology, Lebanese University, Campus Michel Slayman Ras Maska, Tripoli P.O. Box 45061, Lebanon;
- Laboratory of Applied Biotechnology (LBA3B), Azm Center for Research in Biotechnology and Its Applications, EDST, Lebanese University, Tripoli P.O. Box 45061, Lebanon;
| | - Jean-Marc Sabatier
- Institut de Neurophysiopathologie (INP), Aix-Marseille Université CNRS, 13385 Marseille, France;
| |
Collapse
|
21
|
The potential role of renin-angiotensin system in mild traumatic brain injury. Neurol Sci 2022; 43:3353-3359. [DOI: 10.1007/s10072-021-05856-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 12/24/2021] [Indexed: 11/26/2022]
|
22
|
Khalid H, Khalid S, Sufyan M, Ashfaq UA. In-silico elucidation reveals potential phytochemicals against angiotensin-converting enzyme 2 (ACE-2) receptor to fight coronavirus disease 2019 (COVID-19). Z NATURFORSCH C 2022; 77:473-482. [PMID: 35470645 DOI: 10.1515/znc-2021-0325] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2021] [Accepted: 03/30/2022] [Indexed: 12/18/2022]
Abstract
The coronavirus (SARS-CoV-2) pandemic is rapidly advancing and spreading worldwide, which poses an urgent need to develop anti-SARS-CoV-2 agents. A human receptor, namely, angiotensin-converting enzyme 2 (ACE-2), supports the SARS-CoV-2 entry, therefore, serves as a target for intervention via drug. In the current study, bioinformatic approaches were employed to screen potent bioactive compounds that might be ACE-2 receptor inhibitors. The employment of a docking study using ACE receptor protein with a ready-to-dock database of phytochemicals via MOE software revealed five compounds as potent molecules. Among them, astragaloside exhibited the highest binding affinity -21.8 kcal/mol and stable interactions within the active site of the ACE-2 receptor. Similarly, the phytochemicals such as pterocaryanin B, isoastragaloside II, and astraisoflavan glucoside followed by oleuropein showed a stronger binding affinity. We hypothesize these compounds as potential lead candidates for the development of anti- COVID-19 target-specific drugs.
Collapse
Affiliation(s)
- Hina Khalid
- Department of Bioinformatics and Biotechnology, Government College University, Faisalabad, Pakistan
| | - Sana Khalid
- Department of Agricultural Engineering and Food Science, Shandong University of Technology, Zibo, China
| | - Muhammad Sufyan
- Department of Bioinformatics and Biotechnology, Government College University, Faisalabad, Pakistan
| | - Usman Ali Ashfaq
- Department of Bioinformatics and Biotechnology, Government College University, Faisalabad, Pakistan
| |
Collapse
|
23
|
Boccatonda A, Grignaschi A, Lanotte AMG, Cocco G, Vidili G, Giostra F, Schiavone C. Role of Lung Ultrasound in the Management of Patients with Suspected SARS-CoV-2 Infection in the Emergency Department. J Clin Med 2022; 11:jcm11082067. [PMID: 35456160 PMCID: PMC9025104 DOI: 10.3390/jcm11082067] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Revised: 03/21/2022] [Accepted: 04/02/2022] [Indexed: 01/27/2023] Open
Abstract
Background: The lung ultrasound (LUS) score has been proposed as an optimal scheme for the ultrasound study of patients with suspected/confirmed COVID-19 pneumonia. The aims of our study were to evaluate the use of lung ultrasound as a diagnostic tool for diagnosing SARS-CoV-2 pneumonia, to examine the validity of the LUS score for the diagnosis of COVID-19 pneumonia, and to correlate this score with hospitalization rate and 30-day mortality. Materials and Methods: A retrospective analysis was performed on 1460 patients who were referred to the General Emergency Department of the S. Orsola-Malpighi Hospital from April 2020 to May 2020 for symptoms suspected to indicate SARS-CoV-2 infection. The ultrasound examination was based on a common execution scheme called the LUS score, as previously described. Results and Conclusions: The LUS score was found to correlate with the degree of clinical severity and respiratory failure (paO2/FiO2 ratio and the alveolar−arterial gradient increase than expected for age). It was shown that COVID-19 patients with an LUS score of >7 require the use of oxygen support, and a value of >10 is associated with an increased risk of oro-tracheal intubation. The LUS score was found to present higher values in hospitalized patients, increasing according to the degree of care intensity. Patients who died from COVID-19 were characterized by a mean LUS score of 11 at presentation to the emergency department. An LUS score of >7.5 was found to indicate a sensitivity of 83% and a specificity of 89% for 30-day mortality in COVID-19 patients. The use of LUS seems to be an optimal first level method for pneumonia detection and risk stratification in patients with suspected SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Andrea Boccatonda
- Emergency Department, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Azienda Ospedaliero Universitaria di Bologna, 40138 Bologna, Italy; (A.G.); (A.M.G.L.); (F.G.)
- Correspondence: ; Tel.: +39-051-214-3324; Fax: +39-051-214-3349
| | - Alice Grignaschi
- Emergency Department, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Azienda Ospedaliero Universitaria di Bologna, 40138 Bologna, Italy; (A.G.); (A.M.G.L.); (F.G.)
| | - Antonella Maria Grazia Lanotte
- Emergency Department, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Azienda Ospedaliero Universitaria di Bologna, 40138 Bologna, Italy; (A.G.); (A.M.G.L.); (F.G.)
| | - Giulio Cocco
- Internal Medicine, G. d’Annunzio University, 66100 Chieti, Italy; (G.C.); (C.S.)
| | - Gianpaolo Vidili
- Department of Medical, Surgical and Experimental Sciences, University of Sassari, 07100 Sassari, Italy;
| | - Fabrizio Giostra
- Emergency Department, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Azienda Ospedaliero Universitaria di Bologna, 40138 Bologna, Italy; (A.G.); (A.M.G.L.); (F.G.)
| | - Cosima Schiavone
- Internal Medicine, G. d’Annunzio University, 66100 Chieti, Italy; (G.C.); (C.S.)
| |
Collapse
|
24
|
Does the Serum Concentration of Angiotensin II Type 1 Receptor Have an Effect on the Severity of COVID-19? A Prospective Preliminary Observational Study among Healthcare Professionals. J Clin Med 2022; 11:jcm11071769. [PMID: 35407377 PMCID: PMC8999741 DOI: 10.3390/jcm11071769] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 03/19/2022] [Accepted: 03/21/2022] [Indexed: 01/09/2023] Open
Abstract
SARS-CoV-2 is a virus that causes severe respiratory distress syndrome. The pathophysiology of COVID-19 is related to the renin-angiotensin system (RAS). SARS-CoV-2, a vector of COVID-19, uses angiotensin-converting enzyme 2 (ACE-2), which is highly expressed in human lung tissue, nasal cavity, and oral mucosa, to gain access into human cells. After entering the cell, SARS-CoV-2 inhibits ACE-2, thus favouring the ACE/Ang II/angiotensin II type 1 receptor (AT1R) axis, which plays a role in the development of acute lung injury (ALI). This study aimed to analyse the influence of angiotensin 1 receptor (AT1R) levels in the serum on the course of the severity of symptoms in healthcare professionals who had a SARS-CoV-2 infection. This prospective observational study was conducted on a group of 82 participants. The study group included physicians and nurses who had a COVID-19 infection confirmed by real-time reverse transcription-polymerase chain reaction (RT-PCR) test for SARS-CoV-2. The control group consisted of healthy medical professionals who had not had a SARS-CoV-2 infection or who had no symptoms of COVID-19 and who tested negative for SARS-CoV-2 on the day of examination. We analysed the correlation between AT1R concentration and the severity of COVID-19, as well as with sex, age, blood group, and comorbidities. There were no statistically significant differences in the mean values of AT1R concentration in the recovered individuals and the non-COVID-19 subjects (3.29 vs. 3.76 ng/mL; p = 0.32). The ROC curve for the AT1R assay showed an optimal cut-off point of 1.33 (AUC = 0.44; 95% CI = 0.32-0.57; p = 0.37). There was also no correlation between AT1R concentration and the severity of symptoms associated with COVID-19. Blood type analysis showed statistically significantly lower levels of AT1R in COVID-19-recovered participants with blood group A than in those with blood group O. In conclusion, AT1R concentration does not affect the severity of symptoms associated with COVID-19 among healthcare professionals.
Collapse
|
25
|
Angiotensin II Type I Receptor (AT1R): The Gate towards COVID-19-Associated Diseases. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27072048. [PMID: 35408447 PMCID: PMC9000463 DOI: 10.3390/molecules27072048] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 03/14/2022] [Accepted: 03/21/2022] [Indexed: 01/08/2023]
Abstract
The binding of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike glycoprotein to its cellular receptor, the angiotensin-converting enzyme 2 (ACE2), causes its downregulation, which subsequently leads to the dysregulation of the renin-angiotensin system (RAS) in favor of the ACE-angiotensin II (Ang II)-angiotensin II type I receptor (AT1R) axis. AT1R has a major role in RAS by being involved in several physiological events including blood pressure control and electrolyte balance. Following SARS-CoV-2 infection, pathogenic episodes generated by the vasoconstriction, proinflammatory, profibrotic, and prooxidative consequences of the Ang II-AT1R axis activation are accompanied by a hyperinflammatory state (cytokine storm) and an acute respiratory distress syndrome (ARDS). AT1R, a member of the G protein-coupled receptor (GPCR) family, modulates Ang II deleterious effects through the activation of multiple downstream signaling pathways, among which are MAP kinases (ERK 1/2, JNK, p38MAPK), receptor tyrosine kinases (PDGF, EGFR, insulin receptor), and nonreceptor tyrosine kinases (Src, JAK/STAT, focal adhesion kinase (FAK)), and nicotinamide adenine dinucleotide phosphate (NADPH) oxidase. COVID-19 is well known for generating respiratory symptoms, but because ACE2 is expressed in various body tissues, several extrapulmonary pathologies are also manifested, including neurologic disorders, vasculature and myocardial complications, kidney injury, gastrointestinal symptoms, hepatic injury, hyperglycemia, and dermatologic complications. Therefore, the development of drugs based on RAS blockers, such as angiotensin II receptor blockers (ARBs), that inhibit the damaging axis of the RAS cascade may become one of the most promising approaches for the treatment of COVID-19 in the near future. We herein review the general features of AT1R, with a special focus on the receptor-mediated activation of the different downstream signaling pathways leading to specific cellular responses. In addition, we provide the latest insights into the roles of AT1R in COVID-19 outcomes in different systems of the human body, as well as the role of ARBs as tentative pharmacological agents to treat COVID-19.
Collapse
|
26
|
D’Ardes D, Boccatonda A, Cocco G, Fabiani S, Rossi I, Bucci M, Guagnano MT, Schiavone C, Cipollone F. Impaired coagulation, liver dysfunction and COVID-19: Discovering an intriguing relationship. World J Gastroenterol 2022; 28:1102-1112. [PMID: 35431501 PMCID: PMC8985482 DOI: 10.3748/wjg.v28.i11.1102] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Revised: 08/09/2021] [Accepted: 02/15/2022] [Indexed: 02/06/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19) is, at present, one of the most relevant global health problems. In the literature hepatic alterations have been described in COVID-19 patients, and they are mainly represented by worsening of underlying chronic liver disease leading to hepatic decompensation and liver failure with higher mortality. Several potential mechanisms used by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) to cause liver damage have been hypothesized. COVID-19 primary liver injury is less common than secondary liver injury. Most of the available data demonstrate how liver damage in SARS-CoV-2 infection is likely due to systemic inflammation, and it is less likely mediated by a cytopathic effect directed on liver cells. Moreover, liver alterations could be caused by hypoxic injury and drugs (antibiotics and non-steroidal anti-inflammatory drugs, remdesivir, tocilizumab, tofacitinib and dexamethasone). SARS-CoV-2 infection can induce multiple vascular district atherothrombosis by affecting simultaneously cerebral, coronary and peripheral vascular beds. Data in the literature highlight how the virus triggers an exaggerated immune response, which added to the cytopathic effect of the virus can induce endothelial damage and a prothrombotic dysregulation of hemostasis. This leads to a higher incidence of symptomatic and confirmed venous thrombosis and of pulmonary embolisms, especially in central, lobar or segmental pulmonary arteries, in COVID-19. There are currently fewer data for arterial thrombosis, while myocardial injury was identified in 7%-17% of patients hospitalized with SARS-CoV-2 infection and 22%-31% in the intensive care unit setting. Available data also revealed a higher occurrence of stroke and more serious forms of peripheral arterial disease in COVID-19 patients. Hemostasis dysregulation is observed during the COVID-19 course. Lower platelet count, mildly increased prothrombin time and increased D-dimer are typical laboratory features of patients with severe SARS-CoV-2 infection, described as "COVID-19 associated coagulopathy." These alterations are correlated to poor outcomes. Moreover, patients with severe SARS-CoV-2 infection are characterized by high levels of von Willebrand factor with subsequent ADAMTS13 deficiency and impaired fibrinolysis. Platelet hyperreactivity, hypercoagulability and hypofibrinolysis during SARS-CoV-2 infection induce a pathological state named as "immuno-thromboinflammation." Finally, liver dysfunction and coagulopathy are often observed at the same time in patients with COVID-19. The hypothesis that liver dysfunction could be mediated by microvascular thrombosis has been supported by post-mortem findings and extensive vascular portal and sinusoidal thrombosis observation. Other evidence has shown a correlation between coagulation and liver damage in COVID-19, underlined by the transaminase association with coagulopathy, identified through laboratory markers such as prothrombin time, international normalized ratio, fibrinogen, D-dimer, fibrin/fibrinogen degradation products and platelet count. Other possible mechanisms like immunogenesis of COVID-19 damage or massive pericyte activation with consequent vessel wall fibrosis have been suggested.
Collapse
Affiliation(s)
- Damiano D’Ardes
- “Clinica Medica” Institute, Department of Medicine and Aging Sciences, “G. D’Annunzio” University of Chieti-Pescara, Chieti 66100, Italy
| | - Andrea Boccatonda
- Unit of Ultrasound, “G. D’Annunzio” University of Chieti-Pescara, Chieti 66100, Italy
| | - Giulio Cocco
- Unit of Ultrasound, “G. D’Annunzio” University of Chieti-Pescara, Chieti 66100, Italy
| | - Stefano Fabiani
- Unit of Ultrasound, “G. D’Annunzio” University of Chieti-Pescara, Chieti 66100, Italy
| | - Ilaria Rossi
- “Clinica Medica” Institute, Department of Medicine and Aging Sciences, “G. D’Annunzio” University of Chieti-Pescara, Chieti 66100, Italy
| | - Marco Bucci
- “Clinica Medica” Institute, Department of Medicine and Aging Sciences, “G. D’Annunzio” University of Chieti-Pescara, Chieti 66100, Italy
| | - Maria Teresa Guagnano
- “Clinica Medica” Institute, Department of Medicine and Aging Sciences, “G. D’Annunzio” University of Chieti-Pescara, Chieti 66100, Italy
| | - Cosima Schiavone
- Unit of Ultrasound, “G. D’Annunzio” University of Chieti-Pescara, Chieti 66100, Italy
| | - Francesco Cipollone
- “Clinica Medica” Institute, Department of Medicine and Aging Sciences, “G. D’Annunzio” University of Chieti-Pescara, Chieti 66100, Italy
| |
Collapse
|
27
|
Liu A, Raja xavier J, Singh Y, Brucker SY, Salker MS. Molecular and Physiological Aspects of SARS-CoV-2 Infection in Women and Pregnancy. Front Glob Womens Health 2022; 3:756362. [PMID: 35284910 PMCID: PMC8908006 DOI: 10.3389/fgwh.2022.756362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 02/01/2022] [Indexed: 01/08/2023] Open
Abstract
Whilst scientific knowledge about SARS-CoV-2 and COVID-19 is rapidly increasing, much of the effects on pregnant women is still unknown. To accommodate pregnancy, the human endometrium must undergo a physiological transformation called decidualization. These changes encompass the remodeling of endometrial immune cells leading to immunotolerance of the semi-allogenic conceptus as well as defense against pathogens. The angiotensin converting enzyme 2 (ACE2) plays an important regulatory role in the renin-angiotensin-system (RAS) and has been shown to be protective against comorbidities known to worsen COVID-19 outcomes. Furthermore, ACE2 is also crucial for decidualization and thus for early gestation. An astounding gender difference has been found in COVID-19 with male patients presenting with more severe cases and higher mortality rates. This could be attributed to differences in sex chromosomes, hormone levels and behavior patterns. Despite profound changes in the female body during pregnancy, expectant mothers do not face worse outcomes compared with non-pregnant women. Whereas mother-to-child transmission through respiratory droplets during labor or in the postnatal period is known, another question of in utero transmission remains unanswered. Evidence of placental SARS-CoV-2 infection and expression of viral entry receptors at the maternal-fetal interface suggests the possibility of in utero transmission. SARS-CoV-2 can cause further harm through placental damage, maternal systemic inflammation, and hindered access to health care during the pandemic. More research on the effects of COVID-19 during early pregnancy as well as vaccination and treatment options for gravid patients is urgently needed.
Collapse
Affiliation(s)
- Anna Liu
- Research Institute of Women's Health, Eberhard Karls University, Tübingen, Germany
| | - Janet Raja xavier
- Research Institute of Women's Health, Eberhard Karls University, Tübingen, Germany
| | - Yogesh Singh
- Research Institute of Women's Health, Eberhard Karls University, Tübingen, Germany
- Institute of Medical Genetics and Applied Genomics, Eberhard Karls University, Tübingen, Germany
| | - Sara Y. Brucker
- Research Institute of Women's Health, Eberhard Karls University, Tübingen, Germany
| | - Madhuri S. Salker
- Research Institute of Women's Health, Eberhard Karls University, Tübingen, Germany
| |
Collapse
|
28
|
Chen CH, Lin SW, Shen CF, Hsieh KS, Cheng CM. Biomarkers during COVID-19: Mechanisms of Change and Implications for Patient Outcomes. Diagnostics (Basel) 2022; 12:509. [PMID: 35204599 PMCID: PMC8870804 DOI: 10.3390/diagnostics12020509] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 02/07/2022] [Accepted: 02/14/2022] [Indexed: 01/08/2023] Open
Abstract
As the COVID-19 (Coronavirus disease 19) pandemic spreads worldwide, the massive numbers of COVID-19 patients have created a considerable healthcare burden for every country. The clinical spectrum of SARS-CoV-2 infection is broad, ranging from asymptomatic to mild, moderate, severe, and critical. Most COVID-19 patients present with no or mild symptoms, but nearly one-fifth of all patients develop severe or life-threatening complications. In addition to localized respiratory manifestations, severe COVID-19 cases also show extra-pulmonary complications or induce multiorgan failure. Identifying, triaging, and treating patients at risk early is essential and urgent. This article reviews the potential prognostic value of various biomarkers at different clinical spectrum stages of COVID-19 infection and includes information on fundamental prognostic mechanisms as well as potential clinical implications. Biomarkers are measurable biochemical substances used to recognize and indicate disease severity or response to therapeutic interventions. The information they provide is objective and suitable for delivering healthcare providers with a means of stratifying disease state in COVID-19 patients. This, in turn, can be used to help select and guide intervention efforts as well as gauge the efficacy of therapeutic approaches. Here, we review a number of potential biomarkers that may be used to guide treatment, monitor treatment efficacy, and form individualized therapeutic guidance based on patient response. Implementation of the COVID-19 biomarkers discussed here may lead to significantly improved quality of care and patient outcomes for those infected with SARS-CoV-2 worldwide.
Collapse
Affiliation(s)
- Cheng-Han Chen
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu 30013, Taiwan; (C.-H.C.); (S.-W.L.)
- Department of Emergency Medicine, Taipei Veterans General Hospital, Taipei 11217, Taiwan
- School of Medicine, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan
| | - Sheng-Wen Lin
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu 30013, Taiwan; (C.-H.C.); (S.-W.L.)
| | - Ching-Fen Shen
- Department of Pediatrics, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan;
| | - Kai-Sheng Hsieh
- Department of Pediatrics, Shuang Ho Hospital, Taipei Medical University, Taipei 23561, Taiwan
| | - Chao-Min Cheng
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu 30013, Taiwan; (C.-H.C.); (S.-W.L.)
| |
Collapse
|
29
|
Saad H, Jabotian K, Sakr C, Mahfouz R, Akl IB, Zgheib NK. The Role of Angiotensin Converting Enzyme 1 Insertion/Deletion Genetic Polymorphism in the Risk and Severity of COVID-19 Infection. Front Med (Lausanne) 2022; 8:798571. [PMID: 35004773 PMCID: PMC8733297 DOI: 10.3389/fmed.2021.798571] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 11/29/2021] [Indexed: 12/17/2022] Open
Abstract
Background: Individuals infected with the COVID-19 virus present with different symptoms of varying severity. In addition, not all individuals are infected despite exposure. Risk factors such as age, sex, and comorbidities play a major role in this variability; however, genetics may also be important in driving the differences in the incidence and prognosis of the disease. An Insertion/Deletion (I/D) polymorphism in the ACE1 gene (rs1799752) may explain these genetic differences. The aims of this study were to determine the potential role of ACE1 I/D genetic polymorphism in the risk of contracting COVID-19 as well as predicting the severity of COVID-19 infection. Methods: Three-hundred and eighty-seven non-related Lebanese subjects, 155 controls and 232 cases, who presented to the American University of Beirut Medical Center (AUBMC) for COVID-19 PCR testing were recruited. Clinical data were collected via filling a questionnaire and accessing the medical records. Peripheral blood was withdrawn for DNA isolation, and genotyping performed with standard PCR followed by band visualization on agarose gel. Results: In our study population, previously described risk factors such as gender, age, and comorbidities were associated with increase in disease susceptibility and severity. ACE1 I was the least common allele, and there was a positive association between ACE1 I and the risk of contracting the COVID-19 disease. More specifically, the frequency of II genotype was significantly higher among cases when compared to controls (P = 0.035) with individuals with the II genotype having greater risk for contracting the COVID-19 disease: OR = 2.074, P = 0.048 in the multivariate analysis. As for disease severity, the DD genotype and D allele were associated with increased risk for developing severe symptoms (OR = 2.845, P = 0.026 and OR = 2.359, P = 0.014, respectively), and the DD genotype with necessitating hospitalization (OR = 2.307, P = 0.042). In parallel, D allele carriers showed a significantly increased risk for developing hypoxia: OR = 4.374, P = 0.045. Conclusion: We found a positive association between ACE1 I and the risk of contracting the COVID-19 disease, and between ACE1 D and a worse outcome of the COVID-19 infection. Therefore, genotyping for ACE1 I/D polymorphism could be used to assess risk and predict severity for better prognosis and management of the disease.
Collapse
Affiliation(s)
- Halim Saad
- Department of Pharmacology and Toxicology, American University of Beirut Faculty of Medicine, Beirut, Lebanon
| | - Karna Jabotian
- Department of Pharmacology and Toxicology, American University of Beirut Faculty of Medicine, Beirut, Lebanon
| | - Carine Sakr
- Employee Health Unit, Department of Family Medicine, American University of Beirut Faculty of Medicine, Beirut, Lebanon
| | - Rami Mahfouz
- Department of Pathology and Laboratory Medicine, American University of Beirut Faculty of Medicine, Beirut, Lebanon
| | - Imad Bou Akl
- Division of Pulmonary, Department of Internal Medicine, American University of Beirut Faculty of Medicine, Beirut, Lebanon
| | - Nathalie K Zgheib
- Department of Pharmacology and Toxicology, American University of Beirut Faculty of Medicine, Beirut, Lebanon
| |
Collapse
|
30
|
Kandasamy S, Gopalakrishnan S, Krishnan B, Krishnan M, Sahul Hameed P, Karunakaran V. The prognostic role of inflammatory markers in COVID-19 patients: A retrospective analysis in a tertiary care hospital of southern India. JOURNAL OF CURRENT RESEARCH IN SCIENTIFIC MEDICINE 2022. [DOI: 10.4103/jcrsm.jcrsm_4_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
|
31
|
COVID-19, the Pandemic of the Century and Its Impact on Cardiovascular Diseases. CARDIOLOGY DISCOVERY 2021; 1:233-258. [PMID: 34888547 PMCID: PMC8638821 DOI: 10.1097/cd9.0000000000000038] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Accepted: 09/19/2021] [Indexed: 01/08/2023]
Abstract
COVID-19 caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection likely ranks among the deadliest diseases in human history. As with other coronaviruses, SARS-CoV-2 infection damages not only the lungs but also the heart and many other organs that express angiotensin-converting enzyme 2 (ACE2), a receptor for SARS-CoV-2. COVID-19 has upended lives worldwide. Dietary behaviors have been altered such that they favor metabolic and cardiovascular complications, while patients have avoided hospital visits because of limited resources and the fear of infection, thereby increasing out-hospital mortality due to delayed diagnosis and treatment. Clinical observations show that sex, age, and race all influence the risk for SARS-CoV-2 infection, as do hypertension, obesity, and pre-existing cardiovascular conditions. Many hospitalized COVID-19 patients suffer cardiac injury, acute coronary syndromes, or cardiac arrhythmia. SARS-CoV-2 infection may lead to cardiomyocyte apoptosis and necrosis, endothelial cell damage and dysfunction, oxidative stress and reactive oxygen species production, vasoconstriction, fibrotic and thrombotic protein expression, vascular permeability and microvascular dysfunction, heart inflammatory cell accumulation and activation, and a cytokine storm. Current data indicate that COVID-19 patients with cardiovascular diseases should not discontinue many existing cardiovascular therapies such as ACE inhibitors, angiotensin receptor blockers, steroids, aspirin, statins, and PCSK9 inhibitors. This review aims to furnish a framework relating to COVID-19 and cardiovascular pathophysiology.
Collapse
|
32
|
Papola F, Biancofiore V, Angeletti C, Grimaldi A, Carucci AC, Cofini V, Necozione S, Rosciano A, Marinangeli F, Cervelli C. Anti-AT1R autoantibodies and prediction of the severity of Covid-19. Hum Immunol 2021; 83:130-133. [PMID: 34772541 PMCID: PMC8577881 DOI: 10.1016/j.humimm.2021.10.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 10/26/2021] [Accepted: 10/27/2021] [Indexed: 12/28/2022]
Abstract
The stimulation of AT1R (Angiotensin II Receptor Type 1) by Angiotensin II has, in addition to the effects on the renin-angiotensin system, also pro-inflammatory effects through stimulation of ADAM17 and subsequent production of INF-gamma and Interleukin-6. This pro-inflammatory action stimulate the cytokine storm that characterizes the most severe forms of SARS-CoV-2 infection. We studied the effect of AT1Rab on the AT1R on 74 subjects with SARS-CoV-2 infection with respiratory symptoms requiring hospitalization. We divided the patients into 2 groups: 34 with moderate and 40 with severe symptoms that required ICU admission. Hospitalized subjects showed a 50% reduction in the frequency of AT1Rab compared to healthy reference population. Of the ICU patients, 33/40 (82.5%) were AT1Rab negative and 16/33 of them (48.5%) died. All 7 patients positive for AT1Rab survived. These preliminary data seem to indicate a protective role played by AT1R autoantibodies on inflammatory activation in SARS-CoV-2 infection pathology.
Collapse
Affiliation(s)
- Franco Papola
- Regional Centre of Immunohaematology and Tissue Typing, S.Salvatore Hospital, L'Aquila, Italy.
| | - Veronica Biancofiore
- Anesthesiology, Intensive Care and Pain Medicine, Emergency Department, S.Salvatore Hospital L'Aquila, Italy
| | - Chiara Angeletti
- Anesthesiology, Intensive Care and Pain Medicine, Emergency Department Civil Hospital G. Mazzini of Teramo, Teramo, Italy
| | | | | | - Vincenza Cofini
- Department of Life, Health and Environmental Science, University of L'Aquila, 67100 L'Aquila, Italy
| | - Stefano Necozione
- Department of Life, Health and Environmental Science, University of L'Aquila, 67100 L'Aquila, Italy
| | - Alessia Rosciano
- Regional Centre of Immunohaematology and Tissue Typing, S.Salvatore Hospital, L'Aquila, Italy
| | - Franco Marinangeli
- Anesthesiology, Intensive Care and Pain Medicine, Emergency Department Civil Hospital G. Mazzini of Teramo, Teramo, Italy
| | - Carla Cervelli
- Regional Centre of Immunohaematology and Tissue Typing, S.Salvatore Hospital, L'Aquila, Italy
| |
Collapse
|
33
|
Chamata Y, Jackson KG, Watson KA, Jauregi P. Whey-Derived Peptides at the Heart of the COVID-19 Pandemic. Int J Mol Sci 2021; 22:11662. [PMID: 34769093 PMCID: PMC8584039 DOI: 10.3390/ijms222111662] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 10/21/2021] [Accepted: 10/26/2021] [Indexed: 12/14/2022] Open
Abstract
The renin-angiotensin system (RAS) is a key regulator of blood pressure and hypertension. Angiotensin-converting enzyme 2 (ACE2) and angiotensin-converting enzyme I (ACE) are two main components of the RAS that play a major role in blood pressure homeostasis. The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) uses ACE2 as a receptor to enter cells. Despite some controversies, numerous studies have reported a significant association between the use of ACE inhibitors and reduced risk of COVID-19. In our previous studies, we produced and identified peptide sequences present in whey hydrolysates exhibiting high ACE inhibitory activity. Therefore, the aim of this work is to obtain an improved understanding of the function of these natural peptides as RAS inhibitors and investigate their potential therapeutic role in the COVID-19 pandemic. The molecular interactions between peptides IPP, LIVTQ, IIAE, LVYPFP, and human ACE2 were assessed by employing a molecular docking approach. The results show that natural whey-derived peptides have a dual inhibitory action against both ACE and ACE2. This dual activity distinguishes these ACE inhibitory peptides from synthetic drugs, such as Captopril and Lisinopril which were not shown to inhibit ACE2 activity, and may represent a potential strategy in the treatment of COVID-19.
Collapse
Affiliation(s)
- Yara Chamata
- Harry Nursten Building, Department of Food and Nutritional Sciences, University of Reading, Reading RG6 6DZ, UK; (K.G.J.); (P.J.)
| | - Kim G. Jackson
- Harry Nursten Building, Department of Food and Nutritional Sciences, University of Reading, Reading RG6 6DZ, UK; (K.G.J.); (P.J.)
| | - Kimberly A. Watson
- Health and Life Sciences Building, School of Biological Sciences, University of Reading, Reading RG6 6EX, UK;
| | - Paula Jauregi
- Harry Nursten Building, Department of Food and Nutritional Sciences, University of Reading, Reading RG6 6DZ, UK; (K.G.J.); (P.J.)
| |
Collapse
|
34
|
Abdurrahman L, Fang X, Zhang Y. Molecular Insights of SARS-CoV-2 Infection and Molecular Treatments. Curr Mol Med 2021; 22:621-639. [PMID: 34645374 DOI: 10.2174/1566524021666211013121831] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 06/15/2021] [Accepted: 07/23/2021] [Indexed: 01/18/2023]
Abstract
The coronavirus disease emerged in December 2019 (COVID-19) caused by Severe Acute Respiratory Syndrome-related coronavirus 2 (SARS-CoV-2) and its rapid global spread has brought an international health emergency and urgent responses for seeking efficient prevention and therapeutic treatment. This has led to imperative needs for illustration of the molecular pathogenesis of SARS-CoV-2, identification of molecular targets or receptors, and development of antiviral drugs, antibodies, and vaccines. In this study, we investigated the current research progress in combating SARS-CoV-2 infection. Based on the published research findings, we first elucidated, at the molecular level, SARS-CoV-2 viral structures, potential viral host-cell-invasion and pathogenic mechanisms, main virus-induced immune responses, and emerging SARS-CoV-2 variants. We then focused on the main virus- and host-based potential targets, summarized and categorized effective inhibitory molecules based on drug development strategies for COVID-19, that can guide efforts for the identification of new drugs and treatment for this problematic disease. Current research and development of antibodies and vaccines were also introduced and discussed. We concluded that the main virus entry route- SARS-CoV-2 spike protein interaction with ACE2 receptors has played a key role in guiding the development of therapeutic treatments against COVID-19, four main therapeutic strategies may be considered in developing molecular therapeutics, and drug repurposing is likely to be an easy, fast and low-cost approach in such a short period of time with urgent need of antiviral drugs. Additionally, the quick development of antibody and vaccine candidates has yielded promising results, but the wide-scale deployment of safe and effective COVID-19 vaccines remains paramount in solving the pandemic crisis. As new variants of the virus begun to emerge, the efficacy of these vaccines and treatments must be closely evaluated. Finally, we discussed the possible challenges of developing molecular therapeutics for COVID-19 and suggested some potential future efforts. Despite the limited availability of literatures, our attempt in this work to provide a relatively comprehensive overview of current SARS-CoV-2 studies can be helpful for quickly acquiring the key information of COVID-19 and further promoting this important research to control and diminish the pandemic.
Collapse
Affiliation(s)
- Lama Abdurrahman
- Department of Chemistry, The University of Texas Rio Grande Valley, Edinburg, Texas 78539. United States
| | - Xiaoqian Fang
- Department of Molecular Science, School of Medicine, The University of Texas Rio Grande Valley, Edinburg, Texas 78539. United States
| | - Yonghong Zhang
- Department of Chemistry, The University of Texas Rio Grande Valley, Edinburg, Texas 78539. United States
| |
Collapse
|
35
|
D’Ardes D, Rossi I, Bucciarelli B, Allegra M, Bianco F, Sinjari B, Marchioni M, Di Nicola M, Santilli F, Guagnano MT, Cipollone F, Bucci M. Metabolic Changes in SARS-CoV-2 Infection: Clinical Data and Molecular Hypothesis to Explain Alterations of Lipid Profile and Thyroid Function Observed in COVID-19 Patients. Life (Basel) 2021; 11:life11080860. [PMID: 34440605 PMCID: PMC8400261 DOI: 10.3390/life11080860] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 08/08/2021] [Accepted: 08/20/2021] [Indexed: 12/11/2022] Open
Abstract
It seems that during SARS-CoV-2 infection, total cholesterol, LDL-C, and HDL-C values decrease and lipids could play a fundamental role in viral replication. Moreover, it has been shown that SARS-CoV-2 infection could influence thyroid function. We performed a retrospective analysis of 118 hospitalized patients with COVID-19, comparing pre-infection lipid profile (53 patients) and thyroid-stimulating hormone (TSH) values (45 patients) to those measured on admission. Our aim was to evaluate whether SARS-CoV-2 infection could be involved in thyroid and lipid profile alterations and study possible correlations with disease severity and clinical outcome. Median baseline values at the admission time were: total cholesterol at 136.89 ± 42.73 mg/dL, LDL-C 81.53 ± 30.35 mg/dL, and HDL-C 32.36 ± 15.13 mg/dL; and triglycerides at 115.00 ± 40.45 mg/dL, non-HDL-C 104.53 ± 32.63 md/dL, and TSH 1.15 ± 1.08 μUI/mL. Median values of pre-infection total cholesterol, HDL-C, and TSH were significantly higher than those measured at the admission time (p value < 0.05). The C-reactive protein (CRP) negatively correlated with LDL-C (p = 0.013) and HDL-C (p = 0.05). Our data underline a possible impact of SARS-CoV-2 infection on thyroid function. Moreover it suggests a possible relation between COVID-19 and the lipid profile with a negative correlation between CRP, LDL-C, and HDL-C values, proposing the hypothesis that lipid lowering could follow the rising of the COVID-19 inflammatory state.
Collapse
Affiliation(s)
- Damiano D’Ardes
- “Clinica Medica” Institute, Department of “Medicine and Science of Aging”, “G. d’Annunzio” University, 66100 Chieti, Italy; (B.B.); (M.A.); (F.S.); (M.T.G.); (F.C.); (M.B.)
- Azienda Sanitaria Locale n° 2 Abruzzo Lanciano-Vasto-Chieti, 66100 Chieti, Italy
- Correspondence: (D.D.); (I.R.)
| | - Ilaria Rossi
- “Clinica Medica” Institute, Department of “Medicine and Science of Aging”, “G. d’Annunzio” University, 66100 Chieti, Italy; (B.B.); (M.A.); (F.S.); (M.T.G.); (F.C.); (M.B.)
- Correspondence: (D.D.); (I.R.)
| | - Benedetta Bucciarelli
- “Clinica Medica” Institute, Department of “Medicine and Science of Aging”, “G. d’Annunzio” University, 66100 Chieti, Italy; (B.B.); (M.A.); (F.S.); (M.T.G.); (F.C.); (M.B.)
| | - Marco Allegra
- “Clinica Medica” Institute, Department of “Medicine and Science of Aging”, “G. d’Annunzio” University, 66100 Chieti, Italy; (B.B.); (M.A.); (F.S.); (M.T.G.); (F.C.); (M.B.)
| | - Francesco Bianco
- Pediatric Cardiology and Adult Congenital Heart Disease, Azienda Ospedaliero-Universitaria “Ospedali Riuniti” of Ancona, 60126 Ancona, Italy;
| | - Bruna Sinjari
- Department of Innovative Technologies in Medicine and Dentistry, “G. d’Annunzio” University, Chieti-Pescara, 66100 Chieti, Italy;
| | - Michele Marchioni
- Laboratory of Biostatistics, Department of “Medical, Oral and Biotechnological Sciences”, “G. d’Annunzio” University, Chieti-Pescara, 66100 Chieti, Italy; (M.M.); (M.D.N.)
| | - Marta Di Nicola
- Laboratory of Biostatistics, Department of “Medical, Oral and Biotechnological Sciences”, “G. d’Annunzio” University, Chieti-Pescara, 66100 Chieti, Italy; (M.M.); (M.D.N.)
| | - Francesca Santilli
- “Clinica Medica” Institute, Department of “Medicine and Science of Aging”, “G. d’Annunzio” University, 66100 Chieti, Italy; (B.B.); (M.A.); (F.S.); (M.T.G.); (F.C.); (M.B.)
- Azienda Sanitaria Locale n° 2 Abruzzo Lanciano-Vasto-Chieti, 66100 Chieti, Italy
| | - Maria Teresa Guagnano
- “Clinica Medica” Institute, Department of “Medicine and Science of Aging”, “G. d’Annunzio” University, 66100 Chieti, Italy; (B.B.); (M.A.); (F.S.); (M.T.G.); (F.C.); (M.B.)
- Azienda Sanitaria Locale n° 2 Abruzzo Lanciano-Vasto-Chieti, 66100 Chieti, Italy
| | - Francesco Cipollone
- “Clinica Medica” Institute, Department of “Medicine and Science of Aging”, “G. d’Annunzio” University, 66100 Chieti, Italy; (B.B.); (M.A.); (F.S.); (M.T.G.); (F.C.); (M.B.)
- Azienda Sanitaria Locale n° 2 Abruzzo Lanciano-Vasto-Chieti, 66100 Chieti, Italy
| | - Marco Bucci
- “Clinica Medica” Institute, Department of “Medicine and Science of Aging”, “G. d’Annunzio” University, 66100 Chieti, Italy; (B.B.); (M.A.); (F.S.); (M.T.G.); (F.C.); (M.B.)
- Azienda Sanitaria Locale n° 2 Abruzzo Lanciano-Vasto-Chieti, 66100 Chieti, Italy
| |
Collapse
|
36
|
Alabsi W, Acosta MF, Al-Obeidi FA, Hay M, Polt R, Mansour HM. Synthesis, Physicochemical Characterization, In Vitro 2D/3D Human Cell Culture, and In Vitro Aerosol Dispersion Performance of Advanced Spray Dried and Co-Spray Dried Angiotensin (1-7) Peptide and PNA5 with Trehalose as Microparticles/Nanoparticles for Targeted Respiratory Delivery as Dry Powder Inhalers. Pharmaceutics 2021; 13:1278. [PMID: 34452239 PMCID: PMC8398878 DOI: 10.3390/pharmaceutics13081278] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 08/03/2021] [Accepted: 08/09/2021] [Indexed: 12/17/2022] Open
Abstract
The peptide hormone Angiotensin (1-7), Ang (1-7) or (Asp-Arg-Val-Tyr-Ile-His-Pro), is an essential component of the renin-angiotensin system (RAS) peripherally and is an agonist of the Mas receptor centrally. Activation of this receptor in the CNS stimulates various biological activities that make the Ang (1-7)/MAS axis a novel therapeutic approach for the treatment of many diseases. The related O-linked glycopeptide, Asp-Arg-Val-Tyr-Ile-His-Ser-(O-β-D-Glc)-amide (PNA5), is a biousian revision of the native peptide hormone Ang (1-7) and shows enhanced stability in vivo and greater levels of brain penetration. We have synthesized the native Ang (1-7) peptide and the glycopeptide, PNA5, and have formulated them for targeted respiratory delivery as inhalable dry powders. Solid phase peptide synthesis (SPPS) successfully produced Ang (1-7) and PNA5. Measurements of solubility and lipophilicity of raw Ang (1-7) and raw PNA5 using experimental and computational approaches confirmed that both the peptide and glycopeptide have high-water solubility and are amphipathic. Advanced organic solution spray drying was used to engineer the particles and produce spray-dried powders (SD) of both the peptide and the glycopeptide, as well as co-spray-dried powders (co-SD) with the non-reducing sugar and pharmaceutical excipient, trehalose. The native peptide, glycopeptide, SD, and co-SD powders were comprehensively characterized, and exhibited distinct glass transitions (Tg) consistent with the amorphous glassy state formation with Tgs that are compatible with use in vivo. The homogeneous particles displayed small sizes in the nanometer size range and low residual water content in the solid-state. Excellent aerosol dispersion performance with a human DPI device was demonstrated. In vitro human cell viability assays showed that Ang (1-7) and PNA5 are biocompatible and safe for different human respiratory and brain cells.
Collapse
Affiliation(s)
- Wafaa Alabsi
- Department of Chemistry & Biochemistry, The University of Arizona, Tucson, AZ 85721, USA; (W.A.); (F.A.A.-O.); (R.P.)
- Skaggs Pharmaceutical Sciences Center, College of Pharmacy, The University of Arizona, Tucson, AZ 85721, USA;
| | - Maria F. Acosta
- Skaggs Pharmaceutical Sciences Center, College of Pharmacy, The University of Arizona, Tucson, AZ 85721, USA;
| | - Fahad A. Al-Obeidi
- Department of Chemistry & Biochemistry, The University of Arizona, Tucson, AZ 85721, USA; (W.A.); (F.A.A.-O.); (R.P.)
| | - Meredith Hay
- The BIO5 Institute, The University of Arizona, Tucson, AZ 85721, USA;
- Department of Physiology, The University of Arizona, Tucson, AZ 85721, USA
- Evelyn F. McKnight Brain Institute, The University of Arizona, Tucson, AZ 85721, USA
| | - Robin Polt
- Department of Chemistry & Biochemistry, The University of Arizona, Tucson, AZ 85721, USA; (W.A.); (F.A.A.-O.); (R.P.)
- Skaggs Pharmaceutical Sciences Center, College of Pharmacy, The University of Arizona, Tucson, AZ 85721, USA;
- The BIO5 Institute, The University of Arizona, Tucson, AZ 85721, USA;
| | - Heidi M. Mansour
- Skaggs Pharmaceutical Sciences Center, College of Pharmacy, The University of Arizona, Tucson, AZ 85721, USA;
- The BIO5 Institute, The University of Arizona, Tucson, AZ 85721, USA;
- Division of Translational & Regenerative Medicine, College of Medicine, The University of Arizona, Tucson, AZ 85721, USA
| |
Collapse
|
37
|
Effects of Statins on Renin-Angiotensin System. J Cardiovasc Dev Dis 2021; 8:jcdd8070080. [PMID: 34357323 PMCID: PMC8305238 DOI: 10.3390/jcdd8070080] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 07/02/2021] [Accepted: 07/05/2021] [Indexed: 12/16/2022] Open
Abstract
Statins, a class of drugs for lowering serum LDL-cholesterol, have attracted attention because of their wide range of pleiotropic effects. An important but often neglected effect of statins is their role in the renin–angiotensin system (RAS) pathway. This pathway plays an integral role in the progression of several diseases including hypertension, heart failure, and renal disease. In this paper, the role of statins in the blockade of different components of this pathway and the underlying mechanisms are reviewed and new therapeutic possibilities of statins are suggested.
Collapse
|
38
|
Rattis BAC, Ramos SG, Celes MRN. Curcumin as a Potential Treatment for COVID-19. Front Pharmacol 2021; 12:675287. [PMID: 34025433 PMCID: PMC8138567 DOI: 10.3389/fphar.2021.675287] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 04/21/2021] [Indexed: 01/08/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19) is an infectious disease that rapidly spread throughout the world leading to high mortality rates. Despite the knowledge of previous diseases caused by viruses of the same family, such as MERS and SARS-CoV, management and treatment of patients with COVID-19 is a challenge. One of the best strategies around the world to help combat the COVID-19 has been directed to drug repositioning; however, these drugs are not specific to this new virus. Additionally, the pathophysiology of COVID-19 is highly heterogeneous, and the way of SARS-CoV-2 modulates the different systems in the host remains unidentified, despite recent discoveries. This complex and multifactorial response requires a comprehensive therapeutic approach, enabling the integration and refinement of therapeutic responses of a given single compound that has several action potentials. In this context, natural compounds, such as Curcumin, have shown beneficial effects on the progression of inflammatory diseases due to its numerous action mechanisms: antiviral, anti-inflammatory, anticoagulant, antiplatelet, and cytoprotective. These and many other effects of curcumin make it a promising target in the adjuvant treatment of COVID-19. Hence, the purpose of this review is to specifically point out how curcumin could interfere at different times/points during the infection caused by SARS-CoV-2, providing a substantial contribution of curcumin as a new adjuvant therapy for the treatment of COVID-19.
Collapse
Affiliation(s)
- Bruna A. C. Rattis
- Department of Pathology, Faculty of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
- Department of Bioscience and Technology, Institute of Tropical Pathology and Public Health, Federal University of Goias, Goiania, Brazil
| | - Simone G. Ramos
- Department of Pathology, Faculty of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Mara R. N. Celes
- Department of Pathology, Faculty of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
- Department of Bioscience and Technology, Institute of Tropical Pathology and Public Health, Federal University of Goias, Goiania, Brazil
| |
Collapse
|
39
|
Kanimozhi G, Pradhapsingh B, Singh Pawar C, Khan HA, Alrokayan SH, Prasad NR. SARS-CoV-2: Pathogenesis, Molecular Targets and Experimental Models. Front Pharmacol 2021; 12:638334. [PMID: 33967772 PMCID: PMC8100521 DOI: 10.3389/fphar.2021.638334] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Accepted: 03/26/2021] [Indexed: 02/05/2023] Open
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a recent pandemic outbreak threatening human beings worldwide. This novel coronavirus disease-19 (COVID-19) infection causes severe morbidity and mortality and rapidly spreading across the countries. Therefore, there is an urgent need for basic fundamental research to understand the pathogenesis and druggable molecular targets of SARS-CoV-2. Recent sequencing data of the viral genome and X-ray crystallographic data of the viral proteins illustrate potential molecular targets that need to be investigated for structure-based drug design. Further, the SARS-CoV-2 viral pathogen isolated from clinical samples needs to be cultivated and titrated. All of these scenarios demand suitable laboratory experimental models. The experimental models should mimic the viral life cycle as it happens in the human lung epithelial cells. Recently, researchers employing primary human lung epithelial cells, intestinal epithelial cells, experimental cell lines like Vero cells, CaCo-2 cells, HEK-293, H1299, Calu-3 for understanding viral titer values. The human iPSC-derived lung organoids, small intestinal organoids, and blood vessel organoids increase interest among researchers to understand SARS-CoV-2 biology and treatment outcome. The SARS-CoV-2 enters the human lung epithelial cells using viral Spike (S1) protein and human angiotensin-converting enzyme 2 (ACE-2) receptor. The laboratory mouse show poor ACE-2 expression and thereby inefficient SARS-CoV-2 infection. Therefore, there was an urgent need to develop transgenic hACE-2 mouse models to understand antiviral agents' therapeutic outcomes. This review highlighted the viral pathogenesis, potential druggable molecular targets, and suitable experimental models for basic fundamental research.
Collapse
Affiliation(s)
- G. Kanimozhi
- Department of Biochemistry, Dharmapuram Gnanambigai Government Arts College for Women, Mayiladuthurai, India
| | - B. Pradhapsingh
- Department of Biochemistry and Biotechnology, Annamalai University, Annamalainagar, India
| | - Charan Singh Pawar
- Department of Biochemistry and Biotechnology, Annamalai University, Annamalainagar, India
| | - Haseeb A. Khan
- Department of Biochemistry, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Salman H. Alrokayan
- Department of Biochemistry, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - N. Rajendra Prasad
- Department of Biochemistry and Biotechnology, Annamalai University, Annamalainagar, India
| |
Collapse
|
40
|
Boccatonda A, Cocco G, Ianniello E, Montanari M, D'Ardes D, Borghi C, Giostra F, Copetti R, Schiavone C. One year of SARS-CoV-2 and lung ultrasound: what has been learned and future perspectives. J Ultrasound 2021; 24:115-123. [PMID: 33851369 PMCID: PMC8043441 DOI: 10.1007/s40477-021-00575-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 03/11/2021] [Indexed: 12/25/2022] Open
Abstract
A first screening by ultrasound can be relevant to set a specific diagnostic and therapeutic route for a patient with a COVID-19 infection. The finding of bilateral B-lines and white lung areas with patchy peripheral distribution and sparing areas is the most suggestive ultrasound picture of COVID-19 pneumonia. Failure to detect bilateral interstitial syndrome (A pattern) on ultrasound excludes COVID-19 pneumonia with good diagnostic accuracy, but does not exclude current infection. The use of shared semiotic and reporting schemes allows the comparison and monitoring of the COVID-19 pulmonary involvement over time. This review aims to summarise the main data on pulmonary ultrasound and COVID-19 to provide accurate and relevant information for clinical practice.
Collapse
Affiliation(s)
- Andrea Boccatonda
- Division of Emergency Medicine, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy. .,Medicina d'Urgenza e Pronto Soccorso, Policlinico Sant'Orsola-Malpighi, Azienda Ospedaliero-Universitaria di Bologna, Via Pietro Albertoni, 15, 40138, Bologna, BO, Italy.
| | - Giulio Cocco
- Internal Medicine, Department of Medicine and Science of Aging, 'G. D'Annunzio' University, Chieti, Italy
| | - Eugenia Ianniello
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | | | - Damiano D'Ardes
- Internal Medicine, Department of Medicine and Science of Aging, 'G. D'Annunzio' University, Chieti, Italy
| | - Claudio Borghi
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Fabrizio Giostra
- Division of Emergency Medicine, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Roberto Copetti
- Emergency Department, Azienda Sanitaria Universitaria Friuli Centrale, Latisana General Hospital, Latisana, Italy
| | - Cosima Schiavone
- Internal Medicine, Department of Medicine and Science of Aging, 'G. D'Annunzio' University, Chieti, Italy
| |
Collapse
|
41
|
Pacheco-Herrero M, Soto-Rojas LO, Harrington CR, Flores-Martinez YM, Villegas-Rojas MM, León-Aguilar AM, Martínez-Gómez PA, Campa-Córdoba BB, Apátiga-Pérez R, Corniel-Taveras CN, Dominguez-García JDJ, Blanco-Alvarez VM, Luna-Muñoz J. Elucidating the Neuropathologic Mechanisms of SARS-CoV-2 Infection. Front Neurol 2021; 12:660087. [PMID: 33912129 PMCID: PMC8072392 DOI: 10.3389/fneur.2021.660087] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 03/09/2021] [Indexed: 01/08/2023] Open
Abstract
The current pandemic caused by the new severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has become a public health emergency. To date, March 1, 2021, coronavirus disease 2019 (COVID-19) has caused about 114 million accumulated cases and 2.53 million deaths worldwide. Previous pieces of evidence suggest that SARS-CoV-2 may affect the central nervous system (CNS) and cause neurological symptoms in COVID-19 patients. It is also known that angiotensin-converting enzyme-2 (ACE2), the primary receptor for SARS-CoV-2 infection, is expressed in different brain areas and cell types. Thus, it is hypothesized that infection by this virus could generate or exacerbate neuropathological alterations. However, the molecular mechanisms that link COVID-19 disease and nerve damage are unclear. In this review, we describe the routes of SARS-CoV-2 invasion into the central nervous system. We also analyze the neuropathologic mechanisms underlying this viral infection, and their potential relationship with the neurological manifestations described in patients with COVID-19, and the appearance or exacerbation of some neurodegenerative diseases.
Collapse
Affiliation(s)
- Mar Pacheco-Herrero
- Neuroscience Research Laboratory, Faculty of Health Sciences, Pontificia Universidad Católica Madre y Maestra, Santiago de los Caballeros, Dominican Republic
| | - Luis O. Soto-Rojas
- Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Charles R. Harrington
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, United Kingdom
| | - Yazmin M. Flores-Martinez
- Programa Institucional de Biomedicina Molecular, Escuela Nacional de Medicina y Homeopatía, Instituto Politécnico Nacional, Mexico City, Mexico
| | - Marcos M. Villegas-Rojas
- Unidad Profesional Interdisciplinaria de Biotecnología del Instituto Politécnico Nacional (UPIBI- IPN), Mexico City, Mexico
| | - Alfredo M. León-Aguilar
- Unidad Profesional Interdisciplinaria de Biotecnología del Instituto Politécnico Nacional (UPIBI- IPN), Mexico City, Mexico
| | - Paola A. Martínez-Gómez
- Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - B. Berenice Campa-Córdoba
- Departamento de Fisiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City, Mexico
- National Dementia BioBank, Ciencias Biológicas, Facultad de Estudios Superiores, Cuautitlán, Mexico
| | - Ricardo Apátiga-Pérez
- Departamento de Fisiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City, Mexico
- National Dementia BioBank, Ciencias Biológicas, Facultad de Estudios Superiores, Cuautitlán, Mexico
| | - Carolin N. Corniel-Taveras
- Neuroscience Research Laboratory, Faculty of Health Sciences, Pontificia Universidad Católica Madre y Maestra, Santiago de los Caballeros, Dominican Republic
| | - Jesabelle de J. Dominguez-García
- Neuroscience Research Laboratory, Faculty of Health Sciences, Pontificia Universidad Católica Madre y Maestra, Santiago de los Caballeros, Dominican Republic
| | | | - José Luna-Muñoz
- National Dementia BioBank, Ciencias Biológicas, Facultad de Estudios Superiores, Cuautitlán, Mexico
- Banco Estado de Cerebros-UNPHU, Universidad Nacional Pedro Henriquez Ureña, Santo Domingo, Dominican Republic
| |
Collapse
|
42
|
Samprathi M, Jayashree M. Biomarkers in COVID-19: An Up-To-Date Review. Front Pediatr 2021; 8:607647. [PMID: 33859967 PMCID: PMC8042162 DOI: 10.3389/fped.2020.607647] [Citation(s) in RCA: 142] [Impact Index Per Article: 47.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 12/23/2020] [Indexed: 01/08/2023] Open
Abstract
The ongoing pandemic of coronavirus disease 2019 (COVID-19) poses several challenges to clinicians. Timely diagnosis and hospitalization, risk stratification, effective utilization of intensive care services, selection of appropriate therapies, monitoring and timely discharge are essential to save the maximum number of lives. Clinical assessment is indispensable, but laboratory markers, or biomarkers, can provide additional, objective information which can significantly impact these components of patient care. COVID-19 is not a localized respiratory infection but a multisystem disease caused by a diffuse systemic process involving a complex interplay of the immunological, inflammatory and coagulative cascades. The understanding of what the virus does to the body and how the body reacts to it has uncovered a gamut of potential biomarkers. This review discusses the different classes of biomarkers - immunological, inflammatory, coagulation, hematological, cardiac, biochemical and miscellaneous - in terms of their pathophysiological basis followed by the current evidence. Differences between children and adults are highlighted. The role of biomarkers in the diagnosis and management of Multisystem Inflammatory Syndrome in Children (MIS-C) is reviewed. The correlation of biomarkers with clinical and radiological features and the viral load, temporal evolution and the effect of treatment remain to be studied in detail. Which biomarker needs to be evaluated when and in whom, and how best this information can contribute to patient care are questions which currently lack convincing answers. With the evidence currently available broad guidelines on the rational use of available biomarkers are presented. Integrating clinical and laboratory data, monitoring trends rather than a single value, correlating with the natural course of the disease and tailoring guidelines to the individual patient and healthcare setting are essential.
Collapse
|
43
|
Ziaja M, Urbanek KA, Kowalska K, Piastowska-Ciesielska AW. Angiotensin II and Angiotensin Receptors 1 and 2-Multifunctional System in Cells Biology, What Do We Know? Cells 2021; 10:cells10020381. [PMID: 33673178 PMCID: PMC7917773 DOI: 10.3390/cells10020381] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 02/05/2021] [Accepted: 02/09/2021] [Indexed: 12/13/2022] Open
Abstract
For years, the renin-angiotensin system (RAS) has been perceived as a system whose role is to primarily modulate the functioning of the cardiovascular system. Years of research into the role of RAS have provided the necessary data to confirm that the role of RAS is very complex and not limited to the cardiovascular system. The presence of individual elements of the renin-angiotensin (RA) system allows to control many processes, ranging from the memorization to pro-cancer processes. Maintaining the proportions between the individual axes of the RA system allows for achieving a balance, often called homeostasis. Thus, any disturbance in the expression or activity of individual RAS elements leads to pathophysiological processes.
Collapse
|
44
|
Hassib M, Hamilton S, Elkhouly A, Li Y, Kaplan AC. Renin-Angiotensin-Aldosterone System Inhibitors and COVID-19: A Meta-Analysis and Systematic Review. Cureus 2021; 13:e13124. [PMID: 33728141 PMCID: PMC7936537 DOI: 10.7759/cureus.13124] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Introduction Increased virulence, the severity of illness, and mortality have all been hypothesized with respect to angiotensin-converting enzyme inhibitor (ACEi)/angiotensin receptor blocker (ARB) use in coronavirus disease 2019 (COVID-19) infection. Our study aims to assess whether ACEi/ARB use in patients with COVID-19 conferred worsened severity of illness or increased mortality. Additionally, we explore the possibility of an unearthed protective benefit due to their interruption of the RAS signaling pathway as observed in cardiovascular diseases. Methods The Cochrane Library, MEDLINE, and EMBASE were searched for studies relevant to COVID-19 severity, mortality, and inflammation in the context of ACEi/ARB use. Eight studies were included with a total of 17,943 patients, 4,292 (23.9%) of which were taking an ACEi or an ARB. The study population was 47.9% female and the average age across all studies was 65. The studies chosen had a sample size of at least 100 patients. Results Mortality outcomes were assessed in six studies and showed no significant difference in mortality among the ACEi/ARB and control groups (odds ratio [OR]: 0.99, 95%CI: 0.48-2.04). Seven studies assessed the severity of COVID-19 and showed no statistically significant difference in disease severity when comparing the ACEi/ARB group to the control group (odds ratio [OR]: 1.30, 95% CI 0.87-1.94). Four studies reported the length of stay with no significant difference between the ACEi/ARB groups as compared to non-users. Four studies included inflammatory markers C-reactive protein (CRP) and D-Dimer, which were noted to be consistently lower in the ACEi/ARB groups when compared to control groups, however, this was not statistically significant. Conclusion Our study found no significant difference in mortality, severity of illness, or length of stay between ACEi/ARB users and non-users with COVID-19 infection. These results support the continuation of ACEi and ARBs in the setting of COVID-19 as advised by the American College of Cardiology (ACC)/American Heart Association (AHA). The decrease in CRP and D-dimer suggests a possible protective effect related to ACEi/ARB use in COVID-19, however, more studies with larger sample sizes are needed to establish this effect.
Collapse
Affiliation(s)
- Mohab Hassib
- Internal Medicine, Saint Francis Medical Center, Trenton, USA
| | - Steven Hamilton
- Internal Medicine, Saint Francis Medical Center, Trenton, USA
| | - Ahmed Elkhouly
- Internal Medicine, Saint Francis Medical Center, Trenton, USA
| | - Yiting Li
- Internal Medicine, Saint Francis Medical Center, Trenton, USA
| | - Adam C Kaplan
- Internal Medicine, Saint Francis Medical Center, Trenton, USA
| |
Collapse
|
45
|
Identification of novel inhibitors of angiotensin-converting enzyme 2 (ACE-2) receptor from Urtica dioica to combat coronavirus disease 2019 (COVID-19). Mol Divers 2021; 25:1795-1809. [PMID: 33398633 PMCID: PMC7781418 DOI: 10.1007/s11030-020-10159-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Accepted: 11/17/2020] [Indexed: 11/08/2022]
Abstract
Abstract The pandemic outbreak of coronavirus (SARS-CoV-2) is rapidly spreading across the globe, so the development of anti-SARS-CoV-2 agents is urgently needed. Angiotensin-converting enzyme 2 (ACE-2), a human receptor that facilitates entry of SARS-CoV-2, serves as a prominent target for drug discovery. In the present study, we have applied the bioinformatics approach for screening of a series of bioactive chemical compounds from Himalayan stinging nettle (Urtica dioica) as potent inhibitors of ACE-2 receptor (PDB ID: 1R4L). The molecular docking was applied to dock a set of representative compounds within the active site region of target receptor protein using 0.8 version of the PyRx virtual screen tool and analyzed by using discovery studio visualizer. Based on the highest binding affinity, 23 compounds were shortlisted as a lead molecule using molecular docking analysis. Among them, β-sitosterol was found with the highest binding affinity − 12.2 kcal/mol and stable interactions with the amino acid residues present on the active site of the ACE-2 receptor. Similarly, luteoxanthin and violaxanthin followed by rutin also displayed stronger binding efficiency. We propose these compounds as potential lead candidates for the development of target-specific therapeutic drugs against COVID-19. Graphic abstract ![]()
Electronic supplementary material The online version of this article (10.1007/s11030-020-10159-2) contains supplementary material, which is available to authorized users.
Collapse
|
46
|
Abstract
PURPOSE OF REVIEW This review focuses on the associations between the renin-angiotensin system, hypertension, and severe acute respiratory syndrome (SARS-COV-2) infection. A brief prelude on the current state of affairs with COVID-19 is given. In addition to an overview of ACE2, Ang II, and Ang (1-7), this review presents a brief statement on hypertension, including the function of enzymes involved in the control of hypertension, cardiovascular disease, diabetes mellitus, and other malignancies. RECENT FINDINGS There is currently no data in support of the concerns raised with the use of ACEIs/ARBs. Many researchers have voiced concerns that the use of ACEIs and ARBs may increase tissue ACE2 levels. These researchers therefore recommend that individuals on ACEIs/ARB's medications withhold such antihypertensive drugs, unless advised by their physicians to do so. SARS-CoV-2 uses ACE2 receptors as the port of entry to human hosts. ACE2 and ACE are different enzymes and ACE inhibitors do not inhibit ACE2. Therefore, the use of ARB's or ACEIs should not be discontinued if an individual is infected by SARS-CoV-2. Further studies are required to investigate the effect of ACEIs and ARBs on ACE2 expression and COVID-19.
Collapse
|
47
|
Íñiguez M, Pérez-Matute P, Villoslada-Blanco P, Recio-Fernandez E, Ezquerro-Pérez D, Alba J, Ferreira-Laso ML, Oteo JA. ACE Gene Variants Rise the Risk of Severe COVID-19 in Patients With Hypertension, Dyslipidemia or Diabetes: A Spanish Pilot Study. Front Endocrinol (Lausanne) 2021; 12:688071. [PMID: 34489863 PMCID: PMC8417306 DOI: 10.3389/fendo.2021.688071] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Accepted: 08/05/2021] [Indexed: 01/28/2023] Open
Abstract
Coronavirus disease 19 (COVID-19) caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection continues to scale and threaten human health and public safety. It is essential to identify those risk factors that lead to a poor prognosis of the disease. A predisposing host genetic background could be one of these factors that explain the interindividual variability to COVID-19 severity. Thus, we have studied whether the rs4341 and rs4343 polymorphisms of the angiotensin converting enzyme (ACE) gene, key regulator of the renin-aldosterone-angiotensin system (RAAS), could explain the different outcomes of 128 COVID-19 patients with diverse degree of severity (33 asymptomatic or mildly symptomatic, 66 hospitalized in the general ward, and 29 admitted to the ICU). We found that G allele of rs4341 and rs4343 was associated with severe COVID-19 in hypertensive patients, independently of gender (p<0.05). G-carrier genotypes of both polymorphisms were also associated with higher mortality (p< 0.05) and higher severity of COVID-19 in dyslipidemic (p<0.05) and type 2 diabetic patients (p< 0.01). The association of G alleles with disease severity was adjusted for age, sex, BMI and number of comorbidities, suggesting that both the metabolic comorbidities and the G allele act synergistically on COVID-19 outcome. Although we did not find a direct association between serum ACE levels and COVID-19 severity, we found higher levels of ACE in the serum of patients with the GG genotype of rs4341 and rs4343 (p<0.05), what could explain the higher susceptibility to develop severe forms of the disease in patients with the GG genotype, in addition to hypertension and dyslipidemia. In conclusion, our preliminary study suggests that the G-containing genotypes of rs4341 and rs4343 confer an additional risk of adverse COVID-19 prognosis. Thus, rs4341 and rs4343 polymorphisms of ACE could be predictive markers of severity of COVID-19 in those patients with hypertension, dyslipidemia or diabetes. The knowledge of these genetic data could contribute to precision management of SARS-CoV-2 infected patients when admitted to hospital.
Collapse
Affiliation(s)
- María Íñiguez
- Infectious Diseases, Microbiota and Metabolism Unit, Infectious Diseases Department, Center for Biomedical Research of La Rioja (CIBIR), Logroño, Spain
| | - Patricia Pérez-Matute
- Infectious Diseases, Microbiota and Metabolism Unit, Infectious Diseases Department, Center for Biomedical Research of La Rioja (CIBIR), Logroño, Spain
| | - Pablo Villoslada-Blanco
- Infectious Diseases, Microbiota and Metabolism Unit, Infectious Diseases Department, Center for Biomedical Research of La Rioja (CIBIR), Logroño, Spain
| | - Emma Recio-Fernandez
- Infectious Diseases, Microbiota and Metabolism Unit, Infectious Diseases Department, Center for Biomedical Research of La Rioja (CIBIR), Logroño, Spain
| | - Diana Ezquerro-Pérez
- Infectious Diseases Department, Hospital Universitario San Pedro, Logroño, Spain
| | - Jorge Alba
- Infectious Diseases Department, Hospital Universitario San Pedro, Logroño, Spain
| | - M. Lourdes Ferreira-Laso
- Department of Anesthesiology and Postoperative Care, Hospital Universitario San Pedro, Logroño, Spain
| | - José A. Oteo
- Infectious Diseases, Microbiota and Metabolism Unit, Infectious Diseases Department, Center for Biomedical Research of La Rioja (CIBIR), Logroño, Spain
- Infectious Diseases Department, Hospital Universitario San Pedro, Logroño, Spain
- *Correspondence: José A. Oteo,
| |
Collapse
|
48
|
Lim JH, Jung HY, Choi JY, Park SH, Kim CD, Kim YL, Cho JH. Hypertension and Electrolyte Disorders in Patients with COVID-19. Electrolyte Blood Press 2020; 18:23-30. [PMID: 33408744 PMCID: PMC7781764 DOI: 10.5049/ebp.2020.18.2.23] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2020] [Revised: 11/21/2020] [Accepted: 11/22/2020] [Indexed: 02/07/2023] Open
Abstract
The worldwide coronavirus disease 2019 (COVID-19) pandemic is still in progress, but much remains unknown about the disease. In this article, we review the association of hypertension or the renin-angiotensin system (RAS) with COVID-19 and the correlation between electrolyte disorders and disease severity. Underlying hypertension is likely to be associated with severe or critical COVID-19, but the relationship is not clear owing to confounding factors. Angiotensin-converting enzyme 2 (ACE2) plays an important role in the non-classical RAS pathway and binds to a receptor binding domain of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). The RAS blockade is known to increase ACE2 levels, but controversy remains regarding the effect of RAS blockade therapy in the course of COVID-19. Some reports have indicated a protective effect of RAS blockade on COVID-19, whereas others have reported an association of RAS blockade therapy with the occurrence of severe complications such as acute kidney injury and admission to the intensive care unit. Electrolyte disorders are not uncommon in patients with COVID-19, and severe COVID-19 has frequently shown hypokalemia, hyponatremia, and hypocalcemia. Electrolyte imbalances are caused by alteration of RAS, gastrointestinal loss, effects of proinflammatory cytokines, and renal tubular dysfunction by the invasion of SARS-CoV-2.
Collapse
Affiliation(s)
- Jeong-Hoon Lim
- Division of Nephrology, Department of Internal Medicine, Kyungpook National University Hospital, Kyungpook National University School of Medicine, Daegu, Korea
| | - Hee-Yeon Jung
- Division of Nephrology, Department of Internal Medicine, Kyungpook National University Hospital, Kyungpook National University School of Medicine, Daegu, Korea
| | - Ji-Young Choi
- Division of Nephrology, Department of Internal Medicine, Kyungpook National University Hospital, Kyungpook National University School of Medicine, Daegu, Korea
| | - Sun-Hee Park
- Division of Nephrology, Department of Internal Medicine, Kyungpook National University Hospital, Kyungpook National University School of Medicine, Daegu, Korea
| | - Chan-Duck Kim
- Division of Nephrology, Department of Internal Medicine, Kyungpook National University Hospital, Kyungpook National University School of Medicine, Daegu, Korea
| | - Yong-Lim Kim
- Division of Nephrology, Department of Internal Medicine, Kyungpook National University Hospital, Kyungpook National University School of Medicine, Daegu, Korea
| | - Jang-Hee Cho
- Division of Nephrology, Department of Internal Medicine, Kyungpook National University Hospital, Kyungpook National University School of Medicine, Daegu, Korea
| |
Collapse
|
49
|
Montanari M, De Ciantis P, Boccatonda A, Venturi M, D'Antuono G, Gangitano G, Cocco G, D'Ardes D, Schiavone C, Giostra F, Perin T. Lung ultrasound monitoring of CPAP effectiveness on SARS-CoV-2 pneumonia: A case report. EMERGENCY CARE JOURNAL 2020. [DOI: 10.4081/ecj.2020.9333] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
SARS-CoV-2 infection is characterized by extremely heterogeneous features, going from cases with few symptoms to severe respiratory failures. Chest Computed Tomography (CT) is currently the gold-standard imaging method, although burdened by the risk of exposure to ionizing radiation and management / organizational concerns. In particular, the critical patient undergoing ventilation (invasive or not) seems to be difficult to monitor by repeated CT scan over time. We report the case of a 55-year-old male patient subjected to Continuous Positive Airway Pressure (CPAP) and prone positioning, in which the use of ultrasound monitoring allowed to verify the effectiveness of the pressure support used in recruiting previously atelectasis lung areas. Lung ultrasound can guide pulmonary recruitment and pronation maneuvers in patients undergoing non-invasive ventilation. Ultrasound can identify atelectatic lung areas, which demonstrate an alveolar re-expansion following the setting of high PEEP values, as underlined by the reappearance of pleural/air interface.
Collapse
|
50
|
Muralidar S, Ambi SV, Sekaran S, Krishnan UM. The emergence of COVID-19 as a global pandemic: Understanding the epidemiology, immune response and potential therapeutic targets of SARS-CoV-2. Biochimie 2020; 179:85-100. [PMID: 32971147 PMCID: PMC7505773 DOI: 10.1016/j.biochi.2020.09.018] [Citation(s) in RCA: 142] [Impact Index Per Article: 35.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 09/14/2020] [Accepted: 09/15/2020] [Indexed: 02/07/2023]
Abstract
An acute respiratory disease caused by a severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) that surfaced in China in late 2019, continues to spread rapidly across the globe causing serious concerns. The coronavirus disease 2019 (COVID-19) is declared as a public health emergency worldwide by the World Health Organization (WHO). Increasing evidences have demonstrated human-to-human transmission that primarily affects the upper respiratory tract followed by lower respiratory tract damage leading to severe pneumonia. Based on the current status, the elderly population and people with prior co-morbidities are highly susceptible to serious health effects including cytokine up-regulation and acute respiratory distress syndrome (ARDS). Currently, COVID-19 research is still in the preliminary stage necessitating rigorous studies. There is no specific drug or vaccine targeting SARS-CoV-2 currently and only symptomatic treatment is being administered, but several antivirals are under active investigation. In this review, we have summarized the epidemiology, entry mechanism, immune response, and therapeutic implications, possible drug targets, their ongoing clinical trials, and put forward vital questions to offer new directions to the COVID-19 research.
Collapse
Affiliation(s)
- Shibi Muralidar
- School of Chemical and Biotechnology, SASTRA Deemed-to-be-University, Thanjavur, 613401, Tamil Nadu, India
| | - Senthil Visaga Ambi
- School of Chemical and Biotechnology, SASTRA Deemed-to-be-University, Thanjavur, 613401, Tamil Nadu, India.
| | - Saravanan Sekaran
- School of Chemical and Biotechnology, SASTRA Deemed-to-be-University, Thanjavur, 613401, Tamil Nadu, India; Centre for Nanotechnology & Advanced Biomaterials (CeNTAB), School of Chemical and Biotechnology, SASTRA Deemed-to-be-University, Thanjavur, 613401, Tamil Nadu, India
| | - Uma Maheswari Krishnan
- School of Chemical and Biotechnology, SASTRA Deemed-to-be-University, Thanjavur, 613401, Tamil Nadu, India; Centre for Nanotechnology & Advanced Biomaterials (CeNTAB), School of Chemical and Biotechnology, SASTRA Deemed-to-be-University, Thanjavur, 613401, Tamil Nadu, India; School of Arts, Science & Technology (SASH), SASTRA Deemed-to-be-University, Thanjavur, 613401, Tamil Nadu, India
| |
Collapse
|