1
|
Iwata A, Chelvanambi S, Asano T, Whelan M, Nakamura Y, Aikawa E, Sasaki Y, Aikawa M. Gene expression profiles of precursor cells identify compounds that reduce NRP1 surface expression in macrophages: Implication for drug repositioning for COVID-19. Front Cardiovasc Med 2024; 11:1438396. [PMID: 39512370 PMCID: PMC11541348 DOI: 10.3389/fcvm.2024.1438396] [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: 05/25/2024] [Accepted: 09/23/2024] [Indexed: 11/15/2024] Open
Abstract
Coronavirus disease 2019 (COVID-19) is transitioning from a pandemic to an endemic phase through recurring mutations. Initial efforts focused on developing strategies to mitigate infection of lung epithelial cells which are the primary targets of the SARS-CoV-2 virus using the affinity of the spike protein to human ACE2 receptor. SARS-CoV-2, however, infects additional cell types present in the lung such as macrophages through the alternate entry receptor Neuropilin 1 (NRP1). Developing novel therapeutic strategies to prevent SARS-CoV-2 infection of cells crucial for immunosurveillance could thus be integral to treat post-acute sequelae of COVID-19 (PASC). Since traditional drug development process takes a long time, it is imperative to establish new strategies that can be rapidly deployed to combat the dynamic nature of COVID-19 evolution and to contribute to prevention of future pandemics. We obtained the gene expression profiles of THP-1 monocytes from L1000-based Connectivity Map using CLUE, cloud- based software platform for the analysis of perturbational datasets to identify compounds that could reduce the expression level of NRP1. Out of 33,590 compounds, we analyzed the profiles of 45 compounds for their ability to reduce NRP1 expression. We selected the top five small molecule inhibitors predicted to decrease the expression of NRP1 for validation studies. All five selected compounds showed low cytotoxicity at tested doses and their ability to reduce NRP1 surface expression was evaluated in THP-1 monocytes, THP-1-derived macrophage like cells and human peripheral blood mononuclear cell (PBMC)-derived primary macrophages. Five compounds with the largest predicted reduction of NRP1 expression decreased macrophage NRP1 surface expression measured using flow cytometry and fluorescent microscopy assays in both cell line and primary macrophages. Using our computational approach, we identified 45 compounds that could potentially decrease NRP1 surface expression in macrophages based on their effect on THP-1 monocytes. Validation studies showed that such an approach can help to identify compounds for drug repositioning in target cells that are absent in the L1000 database. Our proposed approach can be applicable for the rapid compound exploration to combat novel cell types that SARS-CoV-2 targets for infection and could provide molecular bases for the development of new drugs.
Collapse
Affiliation(s)
- Akira Iwata
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Sarvesh Chelvanambi
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Takaharu Asano
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Mary Whelan
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Yuto Nakamura
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Elena Aikawa
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
- Center for Excellence in Vascular Biology, Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Yusuke Sasaki
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Masanori Aikawa
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
- Center for Excellence in Vascular Biology, Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
2
|
Mukherjee S, Bayry J. The Yin and Yang of TLR4 in COVID-19. Cytokine Growth Factor Rev 2024:S1359-6101(24)00080-7. [PMID: 39490235 DOI: 10.1016/j.cytogfr.2024.10.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Revised: 10/03/2024] [Accepted: 10/03/2024] [Indexed: 11/05/2024]
Abstract
Various pattern recognition receptors (PRRs), including toll-like receptors (TLRs), play a crucial role in recognizing invading pathogens as well as damage-associated molecular patterns (DAMPs) released in response to infection. The resulting signaling cascades initiate appropriate immune responses to eliminate these pathogens. Current evidence suggests that SARS-CoV-2-driven activation of TLR4, whether through direct recognition of the spike glycoprotein (alone or in combination with endotoxin) or by sensing various TLR4-activating DAMPs or alarmins released during viral infection, acts as a critical mediator of antiviral immunity. However, TLR4 exerts a dual role in COVID-19, demonstrating both beneficial and deleterious effects. Dysregulated TLR4 signaling is implicated in the proinflammatory consequences linked to the immunopathogenesis of COVID-19. Additionally, TLR4 polymorphisms contribute to severity of the disease. Given its significant immunoregulatory impact on COVID-19 immunopathology and host immunity, TLR4 has emerged as a key target for developing inhibitors and immunotherapeutic strategies to mitigate the adverse effects associated with SARS-CoV-2 and related infections. Furthermore, TLR4 agonists are also being explored as adjuvants to enhance immune responses to SARS-CoV-2 vaccines.
Collapse
Affiliation(s)
- Suprabhat Mukherjee
- Integrative Biochemistry & Immunology Laboratory (IBIL), Department of Animal Science, Kazi Nazrul University, Asansol, West Bengal 713 340, India.
| | - Jagadeesh Bayry
- Institut National de la Santé et de la Recherche Médicale, Centre de Recherche des Cordeliers, Sorbonne Université, Université Paris Cité, Paris 75006, France; Department of Biological Sciences & Engineering, Indian Institute of Technology Palakkad, Palakkad 678 623, India.
| |
Collapse
|
3
|
Utay NS, Güerri-Fernández R, Gharakhanian S, Asmuth DM, Contreras M, Kunkler C, Detzel CJ, Warner CD. Serum-derived bovine immunoglobulin treatment in COVID-19 is associated with faster resolution of symptoms: A randomized pilot clinical trial. J Med Virol 2024; 96:e70005. [PMID: 39390688 DOI: 10.1002/jmv.70005] [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: 05/23/2024] [Revised: 09/30/2024] [Accepted: 10/02/2024] [Indexed: 10/12/2024]
Abstract
Effective treatment to prevent hospitalization and death in people with COVID-19 exists, but people still need interventions that alleviate symptoms without drug interactions. Oral serum-derived bovine immunoglobulins (SBI) may reduce symptoms and time-to-improvement in people with mild-to-moderate COVID-19. In this randomized, open-label, single-site study, participants with mild-to-moderate COVID-19 received SBI 5.0 g bis in die (BID) + Standard of Care (SOC) or SOC alone (2:1) for 2 weeks. After 2 weeks, 78.8% of hospitalized participants on SBI + SOC improved by World Health Organization (WHO) scale of ≥3 compared to 61.1% on SOC alone (odds ratio: OR = 2.4; p = 0.0663), with older participants (>57 years) showing more significant differences between the arms (OR = 6.1; p = 0.0109). Further, more participants on SBI + SOC reported absence of COVID-19 symptoms at Week 2 (74.2%) compared to SOC alone (43.6%; OR = 3.7; p = 0.0031), most notably the absence of dyspnea on exertion (OR = 4.4; p = 0.0047), with women exhibiting the most significant eradication of all symptoms (OR = 5.8; p = 0.0080). No difference in change of IL-6 between arms was observed. Overall, participants with mild-to-moderate COVID-19 on SBI + SOC had a shorter time-to-recovery than on SOC alone, with a significantly higher rate of complete resolution of symptoms. Dyspnea on exertion was the symptom most significantly impacted. For people with mild-to-moderate COVID-19, oral SBI could be a safe and effective intervention, devoid of drug interactions.
Collapse
Affiliation(s)
- Netanya S Utay
- Department of Internal Medicine, Division of Infectious Diseases and Geographic Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | | | - Shahin Gharakhanian
- CIC: Cambridge Innovation Center, Pharmaceutical Medicine & Infectious Diseases, Shahin Gharakhanian MD Consulting LLC, One Broadway, Cambridge, Massachusetts, USA
| | - David M Asmuth
- Department of Internal Medicine, University of California, Davis, Sacramento, California, USA
| | | | | | | | | |
Collapse
|
4
|
Niu C, Liang T, Chen Y, Zhu S, Zhou L, Chen N, Qian L, Wang Y, Li M, Zhou X, Cui J. SARS-CoV-2 spike protein induces the cytokine release syndrome by stimulating T cells to produce more IL-2. Front Immunol 2024; 15:1444643. [PMID: 39359733 PMCID: PMC11445618 DOI: 10.3389/fimmu.2024.1444643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 08/13/2024] [Indexed: 10/04/2024] Open
Abstract
Introduction Cytokine release syndrome (CRS) is one of the leading causes of mortality in patients with COVID-19 caused by the SARS-CoV-2 coronavirus. However, the mechanism of CRS induced by SARS-CoV-2 is vague. Methods Using spike protein combined with IL-2, IFN-γ, and TNF-α to stimulate human peripheral blood mononuclear cells (PBMCs) to secrete CRS-related cytokines, the content of cytokines in the supernatant was detected, and the effects of NK, T, and monocytes were analyzed. Results This study shows that dendritic cells loaded with spike protein of SARS-CoV-2 stimulate T cells to release much more interleukin-2 (IL-2,) which subsequently cooperates with spike protein to facilitate PBMCs to release IL-1β, IL-6, and IL-8. These effects are achieved via IL-2 stimulation of NK cells to release tumor necrosis factor-α (TNF-α) and interferon-γ (IFN-γ), as well as T cells to release IFN-γ Mechanistically, IFN-γ and TNF-α enhance the transcription of CD40, and the interaction of CD40 and its ligand stabilizes the membrane expression of toll-like receptor 4 (TLR4) that serves as a receptor of spike protein on the surface of monocytes. As a result, there is a constant interaction between spike protein and TLR4, leading to continuous activation of nuclear factor-κ-gene binding (NF-κB). Furthermore, TNF-α also activates NF-κB signaling in monocytes, which further cooperates with IFN-γ and spike protein to modulate NF-κB-dependent transcription of CRS-related inflammatory cytokines. Discussion Targeting TNF-α/IFN-γ in combination with TLR4 may represent a promising therapeutic approach for alleviating CRS in individuals with COVID-19.
Collapse
Affiliation(s)
- Chao Niu
- Cancer Center, The First Hospital of Jilin University, Changchun, China
- International Center of Future Science, Jilin University, Changchun, China
| | - Tingting Liang
- Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Yongchong Chen
- Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Shan Zhu
- Cancer Center, The First Hospital of Jilin University, Changchun, China
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, China
| | - Lei Zhou
- Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Naifei Chen
- Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Lei Qian
- Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Yufeng Wang
- Cancer Center, The First Hospital of Jilin University, Changchun, China
- Cancer Research Institute of Jilin University, The First Hospital of Jilin University, Changchun, China
| | - Min Li
- Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Xin Zhou
- Cancer Center, The First Hospital of Jilin University, Changchun, China
- International Center of Future Science, Jilin University, Changchun, China
- Cancer Research Institute of Jilin University, The First Hospital of Jilin University, Changchun, China
| | - Jiuwei Cui
- Cancer Center, The First Hospital of Jilin University, Changchun, China
- Cancer Research Institute of Jilin University, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
5
|
Matamalas JT, Chelvanambi S, Decano JL, França RF, Halu A, Santinelli-Pestana DV, Aikawa E, Malhotra R, Aikawa M. Obesity and age are transmission risk factors for SARS-CoV-2 infection among exposed individuals. PNAS NEXUS 2024; 3:pgae294. [PMID: 39192848 PMCID: PMC11348562 DOI: 10.1093/pnasnexus/pgae294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Accepted: 05/07/2024] [Indexed: 08/29/2024]
Abstract
The coronavirus disease (COVID-19) pandemic has occurred in Massachusetts in multiple waves led by a series of emerging variants. While the evidence has linked obesity with severe symptoms of COVID-19, the effect of obesity on susceptibility to SARS-CoV-2 infection remains unclear. Identification of intrinsic factors, which increase the likelihood of exposed individuals succumbing to productive SARS-CoV-2 infection could help plan mitigation efforts to curb the illness. We aim to investigate whether obese individuals have a higher susceptibility to developing productive SARS-CoV-2 infection given comparable exposure to nonobese individuals. This case-control study leveraged data from the Mass General Brigham's (MGB) electronic medical records (EMR), containing 687,813 patients, to determine whether obesity at any age increases the proportion of infections. We used PCR results of 72,613 subjects who tested positive to SARS-CoV-2 or declared exposure to the virus independently of the result of the test. For this study, we defined susceptibility as the likelihood of testing positive upon suspected exposure. We demonstrate evidence that SARS-CoV-2 exposed obese individuals were more prone to become COVID positive than nonobese individuals [adjusted odds ratio = 1.34 (95% CI: 1.29-1.39)]. Temporal analysis showed significantly increased susceptibility in obese individuals across the duration of the pandemic in Massachusetts. Obese exposed individuals are at a higher risk of getting infected with SARS-CoV-2. This indicates that obesity is not only a risk factor for worsened outcomes but also increases the risk for infection upon exposure. Identifying such populations early will be crucial for curbing the spread of this infectious disease.
Collapse
Affiliation(s)
- Joan T Matamalas
- Department of Medicine, Cardiovascular Division, Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA
| | - Sarvesh Chelvanambi
- Department of Medicine, Cardiovascular Division, Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA
| | - Julius L Decano
- Department of Medicine, Cardiovascular Division, Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA
| | - Raony F França
- Faculty of Medicine, University of São Paulo, Av. Dr. Arnaldo, 455 - Cerqueira César, São Paulo, SP 01246-903, Brazil
| | - Arda Halu
- Department of Medicine, Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, 181 Longwood Avenue, Boston, MA 02115, USA
| | - Diego V Santinelli-Pestana
- Department of Medicine, Cardiovascular Division, Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA
| | - Elena Aikawa
- Department of Medicine, Cardiovascular Division, Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA
- Department of Medicine, Center for Excellence in Vascular Biology, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Rajeev Malhotra
- Cardiology Division, Massachusetts General Hospital, Harvard Medical School, 55 Fruit Street, Boston, MA 02114, USA
| | - Masanori Aikawa
- Department of Medicine, Cardiovascular Division, Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA
- Department of Medicine, Center for Excellence in Vascular Biology, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| |
Collapse
|
6
|
Landolina N, Ricci B, Veneziani I, Alicata C, Mariotti FR, Pelosi A, Quatrini L, Mortari EP, Carsetti R, Vacca P, Tumino N, Azzarone B, Moretta L, Maggi E. TLR2/4 are novel activating receptors for SARS-CoV-2 spike protein on NK cells. Front Immunol 2024; 15:1368946. [PMID: 38881905 PMCID: PMC11176535 DOI: 10.3389/fimmu.2024.1368946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 05/03/2024] [Indexed: 06/18/2024] Open
Abstract
Background In early infected or severe coronavirus disease 2019 (COVID-19) patients, circulating NK cells are consistently reduced, despite being highly activated or exhausted. The aim of this paper was to establish whether severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike glycoprotein (SP) may directly trigger NK cells and through which receptor(s). Methods SP-stimulated human NK cells have been evaluated for the expression of activation markers, cytokine release, and cytotoxic activity, as well as for gene expression profiles and NF-kB phosphorylation, and they have been silenced with specific small interfering RNAs. Results SPs from the Wuhan strain and other variants of concern (VOCs) directly bind and stimulate purified NK cells by increasing activation marker expression, cytokine release, and cytolytic activity, prevalently in the CD56brightNK cell subset. VOC-SPs differ in their ability to activate NK cells, G614, and Delta-Plus strains providing the strongest activity in the majority of donors. While VOC-SPs do not trigger ACE2, which is not expressed on NK cells, or other activating receptors, they directly and variably bind to both Toll-like receptor 2 (TLR2) and TLR4. Moreover, SP-driven NK cell functions are inhibited upon masking such receptors or silencing the relative genes. Lastly, VOC-SPs upregulate CD56dimNK cell functions in COVID-19 recovered, but not in non-infected, individuals. Conclusions TLR2 and TLR4 are novel activating receptors for SP in NK cells, suggesting a new role of these cells in orchestrating the pathophysiology of SARS-CoV-2 infection. The pathogenic relevance of this finding is highlighted by the fact that free SP providing NK cell activation is frequently detected in a SARS-CoV-2 inflamed environment and in plasma of infected and long-COVID-19 subjects.
Collapse
Affiliation(s)
- Nadine Landolina
- Tumor Immunology Unit, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Biancamaria Ricci
- Tumor Immunology Unit, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Irene Veneziani
- Tumor Immunology Unit, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Claudia Alicata
- Tumor Immunology Unit, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | | | - Andrea Pelosi
- Tumor Immunology Unit, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Linda Quatrini
- Innate Lymphoid Cells Unit, Immunology Research Area, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Eva Piano Mortari
- B cell Unit, Immunology Research Area, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Rita Carsetti
- B cell Unit, Immunology Research Area, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Paola Vacca
- Innate Lymphoid Cells Unit, Immunology Research Area, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Nicola Tumino
- Innate Lymphoid Cells Unit, Immunology Research Area, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Bruno Azzarone
- Tumor Immunology Unit, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Lorenzo Moretta
- Tumor Immunology Unit, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Enrico Maggi
- Tumor Immunology Unit, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| |
Collapse
|
7
|
Zhang J, Cruz-Cosme R, Zhang C, Liu D, Tang Q, Zhao RY. Endoplasmic reticulum-associated SARS-CoV-2 ORF3a elicits heightened cytopathic effects despite robust ER-associated degradation. mBio 2024; 15:e0303023. [PMID: 38078754 PMCID: PMC10790703 DOI: 10.1128/mbio.03030-23] [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: 11/09/2023] [Accepted: 11/10/2023] [Indexed: 01/17/2024] Open
Abstract
IMPORTANCE The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic has tragically claimed millions of lives through coronavirus disease 2019 (COVID-19), and there remains a critical gap in our understanding of the precise molecular mechanisms responsible for the associated fatality. One key viral factor of interest is the SARS-CoV-2 ORF3a protein, which has been identified as a potent inducer of host cellular proinflammatory responses capable of triggering the catastrophic cytokine storm, a primary contributor to COVID-19-related deaths. Moreover, ORF3a, much like the spike protein, exhibits a propensity for frequent mutations, with certain variants linked to the severity of COVID-19. Our previous research unveiled two distinct types of ORF3a mutant proteins, categorized by their subcellular localizations, setting the stage for a comparative investigation into the functional and mechanistic disparities between these two types of ORF3a variants. Given the clinical significance and functional implications of the natural ORF3a mutations, the findings of this study promise to provide invaluable insights into the potential roles undertaken by these mutant ORF3a proteins in the pathogenesis of COVID-19.
Collapse
Affiliation(s)
- Jiantao Zhang
- Department of Pathology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Ruth Cruz-Cosme
- Department of Microbiology, Howard University College of Medicine, Washington, DC, USA
| | - Chenyu Zhang
- Department of Pathology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Dongxiao Liu
- Department of Microbiology, Howard University College of Medicine, Washington, DC, USA
| | - Qiyi Tang
- Department of Microbiology, Howard University College of Medicine, Washington, DC, USA
| | - Richard Y. Zhao
- Department of Pathology, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Institute of Global Health, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Research & Development Service, VA Maryland Health Care System, Baltimore, Maryland, USA
| |
Collapse
|
8
|
Rasmi Y, Shokati A, Hatamkhani S, Farnamian Y, Naderi R, Jalali L. Assessment of the relationship between the dopaminergic pathway and severe acute respiratory syndrome coronavirus 2 infection, with related neuropathological features, and potential therapeutic approaches in COVID-19 infection. Rev Med Virol 2024; 34:e2506. [PMID: 38282395 DOI: 10.1002/rmv.2506] [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: 07/23/2022] [Revised: 07/06/2023] [Accepted: 12/17/2023] [Indexed: 01/30/2024]
Abstract
Dopamine is a known catecholamine neurotransmitter involved in several physiological processes, including motor control, motivation, reward, cognition, and immune function. Dopamine receptors are widely distributed throughout the nervous system and in immune cells. Several viruses, including human immunodeficiency virus and Japanese encephalitis virus, can use dopaminergic receptors to replicate in the nervous system and are involved in viral neuropathogenesis. In addition, studies suggest that dopaminergic receptors may play a role in the progression and pathogenesis of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. When SARS-CoV-2 binds to angiotensin-converting enzyme 2 receptors on the surface of neuronal cells, the spike protein of the virus can bind to dopaminergic receptors on neighbouring cells to accelerate its life cycle and exacerbate neurological symptoms. In addition, recent research has shown that dopamine is an important regulator of the immune-neuroendocrine system. Most immune cells express dopamine receptors and other dopamine-related proteins, indicating the importance of dopaminergic immune regulation. The increase in dopamine concentration during SARS-CoV2 infection may reduce immunity (innate and adaptive) that promotes viral spread, which could lead to neuronal damage. In addition, dopaminergic signalling in the nervous system may be affected by SARS-CoV-2 infection. COVID -19 can cause various neurological symptoms as it interacts with the immune system. One possible treatment strategy for COVID -19 patients could be the use of dopamine antagonists. To fully understand how to protect the neurological system and immune cells from the virus, we need to study the pathophysiology of the dopamine system in SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Yousef Rasmi
- Cellular and Molecular Research Center, Cellular and Molecular Medicine Research Institute, Urmia University of Medical Sciences, Urmia, Iran
- Department of Biochemistry, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Ameneh Shokati
- Department of Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Multiple Sclerosis Research Center, Neuroscience Institute, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Shima Hatamkhani
- Experimental and Applied Pharmaceutical Sciences Research Center, Urmia University of Medical Sciences, Urmia, Iran
- Department of Clinical Pharmacy, School of Pharmacy, Urmia University of Medical Sciences, Urmia, Iran
| | - Yeganeh Farnamian
- Student Research Committee, Urmia University of Medical Sciences, Urmia, Iran
| | - Roya Naderi
- Nephrology and Kidney Transplant Research Center, Clinical Research Institute, Urmia University of Medical Sciences, Urmia, Iran
- Department of Physiology, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Ladan Jalali
- Cellular and Molecular Research Center, Cellular and Molecular Medicine Research Institute, Urmia University of Medical Sciences, Urmia, Iran
- Department of Biochemistry, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| |
Collapse
|
9
|
Tosta BR, de Almeida IM, da Cruz Pena L, Dos Santos Silva H, Reis-Goes FS, Silva NN, Cruz JVA, Dos Anjos Silva M, de Araújo JF, Rodrigues JL, Oliveira G, Figueiredo RG, Vaz SN, Montaño-Castellón I, Santana D, de Lima Beltrão FE, Carneiro VL, Campos GS, Brites C, Fortuna V, Figueiredo CA, Trindade SC, Ramos HE, Costa RDS. MTOR gene variants are associated with severe COVID-19 outcomes: A multicenter study. Int Immunopharmacol 2023; 125:111155. [PMID: 37951192 DOI: 10.1016/j.intimp.2023.111155] [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: 08/25/2023] [Revised: 10/25/2023] [Accepted: 10/30/2023] [Indexed: 11/13/2023]
Abstract
BACKGROUND The worst outcomes linked to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection have been attributed to the cytokine storm, which contributes significantly to the immunopathogenesis of the disease. The mammalian target of rapamycin (mTOR) pathway is essential for orchestrating innate immune cell defense including cytokine production and is dysregulated in severe Coronavirus Disease 2019 (COVID-19) individuals. The individual genetic background might play a role in the exacerbated immune response. OBJECTIVE In this study, we aimed to investigate the association between MTOR genetic variants and COVID-19 outcomes. METHODS This study enrolled groups of individuals with severe (n = 285) and mild (n = 207) COVID-19 from Brazilian states. The MTOR variants, rs1057079 and rs2536, were genotyped. A logistic regression analysis and Kaplan-Meier survival curves were performed. We applied a genotyping risk score to estimate the cumulative contribution of the risk alleles. Tumor necrosis factor (TNF) and interleukin-6 (IL-6) plasma levels were also measured. RESULTS The T allele of the MTOR rs1057079 variant was associated with a higher likelihood of developing the most severe form of COVID-19. In addition, higher levels of IL-6 and COVID-19 death was linked to the T allele of the rs2536 variant. These variants exhibited a cumulative risk when inherited collectively. CONCLUSIONS These results show a potential pathogenetic role of MTOR gene variants and may be useful for predicting severe outcomes following COVID-19 infection, resulting in a more effective allocation of health resources.
Collapse
Affiliation(s)
- Bruna Ramos Tosta
- Laboratório de Imunofarmacologia e Biologia Molecular, Instituto de Ciências da Saúde, Universidade Federal da Bahia, Brazil
| | - Ingrid Marins de Almeida
- Laboratório de Imunofarmacologia e Biologia Molecular, Instituto de Ciências da Saúde, Universidade Federal da Bahia, Brazil
| | - Laiane da Cruz Pena
- Laboratório de Imunofarmacologia e Biologia Molecular, Instituto de Ciências da Saúde, Universidade Federal da Bahia, Brazil
| | - Hatilla Dos Santos Silva
- Laboratório de Imunofarmacologia e Biologia Molecular, Instituto de Ciências da Saúde, Universidade Federal da Bahia, Brazil
| | - Fabiane S Reis-Goes
- Laboratório de Imunologia e Biologia Molecular, Instituto de Ciências da Saúde, Universidade Federal da Bahia, Brazil
| | - Nívia N Silva
- Laboratório de Imunologia e Biologia Molecular, Instituto de Ciências da Saúde, Universidade Federal da Bahia, Brazil
| | - João Victor Andrade Cruz
- Laboratório de Imunofarmacologia e Biologia Molecular, Instituto de Ciências da Saúde, Universidade Federal da Bahia, Brazil
| | - Mailane Dos Anjos Silva
- Laboratório de Imunofarmacologia e Biologia Molecular, Instituto de Ciências da Saúde, Universidade Federal da Bahia, Brazil
| | - Jéssica Francisco de Araújo
- Laboratório de Imunofarmacologia e Biologia Molecular, Instituto de Ciências da Saúde, Universidade Federal da Bahia, Brazil
| | - Juliana Lopes Rodrigues
- Laboratório de Imunofarmacologia e Biologia Molecular, Instituto de Ciências da Saúde, Universidade Federal da Bahia, Brazil
| | | | | | - Sara Nunes Vaz
- Hospital Universitário Professor Edgard Santos, Universidade Federal da Bahia, Bahia, Brazil
| | - Iris Montaño-Castellón
- Hospital Universitário Professor Edgard Santos, Universidade Federal da Bahia, Bahia, Brazil
| | - Daniele Santana
- Hospital Universitário Professor Edgard Santos, Universidade Federal da Bahia, Bahia, Brazil
| | | | | | - Gubio Soares Campos
- Laboratório de Virologia, Instituto de Ciências da Saúde, Universidade Federal da Bahia, Brazil
| | - Carlos Brites
- Hospital Universitário Professor Edgard Santos, Universidade Federal da Bahia, Bahia, Brazil
| | - Vitor Fortuna
- Laboratório de Imunologia e Biologia Molecular, Instituto de Ciências da Saúde, Universidade Federal da Bahia, Brazil
| | - Camila Alexandrina Figueiredo
- Laboratório de Imunofarmacologia e Biologia Molecular, Instituto de Ciências da Saúde, Universidade Federal da Bahia, Brazil
| | - Soraya Castro Trindade
- Laboratório de Imunologia e Biologia Molecular, Instituto de Ciências da Saúde, Universidade Federal da Bahia, Brazil; Universidade Estadual de Feira de Santana, Bahia, Brazil
| | - Helton Estrela Ramos
- Programa de Pós-Graduação em Processos Interativos de Órgãos e Sistema, Instituto de Saúde e Ciência, Universidade Federal da Bahia, Salvador, Bahia, Brazil
| | - Ryan Dos Santos Costa
- Laboratório de Imunofarmacologia e Biologia Molecular, Instituto de Ciências da Saúde, Universidade Federal da Bahia, Brazil.
| |
Collapse
|
10
|
Capra AP, Crupi L, Pantò G, Repici A, Calapai F, Squeri R, Ardizzone A, Esposito E. Serum Pentraxin 3 as Promising Biomarker for the Long-Lasting Inflammatory Response of COVID-19. Int J Mol Sci 2023; 24:14195. [PMID: 37762499 PMCID: PMC10531731 DOI: 10.3390/ijms241814195] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 09/06/2023] [Accepted: 09/14/2023] [Indexed: 09/29/2023] Open
Abstract
Currently, biological markers for COVID-19 disease severity still constitute the main goal of enhancing an efficient treatment to reduce critical consequences such as an abnormal systemic inflammatory response. In this regard, the latest research has shown that Pentraxin 3 (PTX3), a highly conserved innate immunity protein, may serve as a valuable biochemical marker. Based on this evidence, we conducted a case-control study to compare the PTX3 serum levels and several immune-inflammatory mediators of 80 healthcare workers who were subdivided into subjects who were previously infected with SARS-CoV-2 (n = 40) and individuals who were never infected (n = 40). Using a commercially available Enzyme-Linked Immunosorbent Assay (ELISA), PTX3 and various immune-inflammatory protein levels were assessed in serum samples, while also considering possible variables (e.g., gender-related differences). We have shown elevated levels of PTX3 and other inflammatory proteins in previously infected COVID-19-positive subjects (p < 0.001). Moreover, the obtained data also indicate a degree of severity influenced by gender, as shown by the subgroup analysis, in which PTX3 expression was more pronounced in previously COVID-19-positive males (p < 0.001) than in females (p < 0.05) compared to the respective controls. In addition, our data further validate, through a direct comparison of previously COVID-19-positive subjects, greater pro-inflammatory levels in males than in females. Overall, our results may support the validity of PTX3 as a systemic biomarker in prolonged systemic inflammatory responses in the context of COVID-19. Thus, PTX3 modulation could constitute an effective therapeutic strategy for improving the recovery from COVID-19 and its systemic long-term consequences.
Collapse
Affiliation(s)
- Anna Paola Capra
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno d’Alcontres, 98166 Messina, Italy; (A.P.C.); (L.C.); (A.R.); (F.C.); (E.E.)
| | - Lelio Crupi
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno d’Alcontres, 98166 Messina, Italy; (A.P.C.); (L.C.); (A.R.); (F.C.); (E.E.)
| | - Giuseppe Pantò
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Via Consolare Valeria 1, 98125 Messina, Italy; (G.P.); (R.S.)
| | - Alberto Repici
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno d’Alcontres, 98166 Messina, Italy; (A.P.C.); (L.C.); (A.R.); (F.C.); (E.E.)
| | - Fabrizio Calapai
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno d’Alcontres, 98166 Messina, Italy; (A.P.C.); (L.C.); (A.R.); (F.C.); (E.E.)
- Department of Clinical and Experimental Medicine, University of Messina, Via Consolare Valeria 1, 98125 Messina, Italy
| | - Raffaele Squeri
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Via Consolare Valeria 1, 98125 Messina, Italy; (G.P.); (R.S.)
| | - Alessio Ardizzone
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno d’Alcontres, 98166 Messina, Italy; (A.P.C.); (L.C.); (A.R.); (F.C.); (E.E.)
| | - Emanuela Esposito
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno d’Alcontres, 98166 Messina, Italy; (A.P.C.); (L.C.); (A.R.); (F.C.); (E.E.)
| |
Collapse
|
11
|
The effects of lipoic acid on respiratory diseases. Int Immunopharmacol 2023; 116. [PMCID: PMC9933494 DOI: 10.1016/j.intimp.2023.109713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/19/2023]
Abstract
Respiratory diseases, including lung cancer, pulmonary fibrosis, asthma, and the recently emerging fatal coronavirus disease-19 (COVID-19), are the leading causes of illness and death worldwide. The increasing incidence and mortality rates have attracted much attention to the prevention and treatment of these conditions. Lipoic acid (LA), a naturally occurring organosulfur compound, is not only essential for mitochondrial aerobic metabolism but also shows therapeutic potential via certain pharmacological effects (e.g., antioxidative and anti-inflammatory effects). In recent years, accumulating evidence (animal experiments and in vitro studies) has suggested a role of LA in ameliorating many respiratory diseases (e.g., lung cancer, fibrosis, asthma, acute lung injury and smoking-induced lung injury). Therefore, this review will provide an overview of the present investigational evidence on the therapeutic effect of LA against respiratory diseases in vitro and in vivo. We also summarize the corresponding mechanisms of action to inspire further basic studies and clinical trials to confirm the health benefits of LA in the context of respiratory diseases.
Collapse
Key Words
- lipoic acid
- respiratory diseases
- antioxidation
- anti-inflammatory effects
- mechanism of action
- akt, protein kinase b;
- aif, apoptosis-inducing factor;
- ampk, adenosine monophosphate-activated protein kinase;
- α-sma, alpha-smooth muscle actin;
- bcl-2, b-cell lymphoma 2;
- cox-2, cyclooxygenase-2;
- dna, deoxyribonucleic acid;
- er, endoplasmic reticulum;
- erk, extracellular-regulated kinase;
- egfr, epidermal growth factor receptor;
- gr, glutathione reductase;
- gpx, glutathione peroxidase;
- grb2, growth factor receptor-bound protein 2;
- gsh, reduced glutathione;
- gssg, oxidized glutathione;
- hif, hypoxia-inducible factor;
- ho-1, heme oxygenase 1;
- keap-1, kelch-like ech-associated protein 1;
- ig-e, immunoglobulin e;
- il, interleukin
- oct-4, octamer-binding transcription factor 4;
- parp-1, poly (adp-ribose) polymerase-1;
- pdk1, phosphoinositide-dependent kinase-1;
- pdh, pyruvate dehydrogenase;
- pi3k, phosphoinositide 3-kinase;
- pge2, prostaglandin e2;
- pgc1α, peroxisome proliferator-activated receptor‑γ co-activator 1α;
- p70s6k, p70 ribosomal protein s6 kinase;
- fak, focal adhesion kinase;
- sod, superoxide dismutase;
- mapk, mitogen-activated protein kinase;
- mtor, mammalian target of rapamycin;
- nf-κb, nuclear factor-kappa b;
- no, nitric oxide;
- nox-4, nicotinamide adenine dinucleotide phosphate (nadph) oxidase-4;
- nqo1, nadph quinone oxidoreductase 1;
- tnf-α, tumor necrosis factor-α;
- tgf-β1, transforming growth factor beta-1;
- vegf, vascular endothelial growth factor;
Collapse
|
12
|
Almazmomi MA, Alsieni M. Targeting TLR-4 Signaling to Treat COVID-19-induced Acute Kidney Injury. J Pharmacol Pharmacother 2023. [DOI: 10.1177/0976500x221147798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
The newly discovered severe acute respiratory syndrome coronavirus 2 (SARS-CoV2) has turned into a potentially fatal pandemic illness. Numerous acute kidney injury (AKI) cases have been reported, although diffuse alveolar destruction and acute respiratory failure are the major symptoms of SARS-CoV-2 infection. The AKI, often known as a sudden loss of kidney function, carries a greater risk of mortality and morbidity. AKI was the second most frequent cause of death after acute respiratory distress syndrome (ARDS) in critically ill patients with coronavirus disease 2019 (COVID-19). While most patients with COVID-19 have moderate symptoms, some have severe symptoms, such as septic shock and ARDS. Also, it has been proven that some patients have severe symptoms, such as the failure of several organs. The kidneys are often affected either directly or indirectly. The major signs of kidney involvement are proteinuria and AKI. It is hypothesized that multiple mechanisms contribute to kidney injury in COVID-19. Direct infection of podocytes and proximal tubular cells in the kidneys may lead to acute tubular necrosis and collapsing glomerulopathy. SARS-CoV2 may also trigger a cascade of immunological responses that lead to AKI, including cytokine storm (CS), macrophage activation syndrome, and Toll-like receptor type-4 activation (TLR-4). Other proposed processes of AKI include interactions between organs, endothelial failure, hypercoagulability, rhabdomyolysis, and sepsis. Furthermore, ischemic damage to the kidney might result from the decreased oxygen supply. This article focuses on kidney injury’s epidemiology, etiology, and pathophysiological processes. Specifically, it focuses on the CS and the role of TLR-4 in this process. To effectively manage and treat acute kidney damage and AKI in COVID-19, it is crucial to understand the underlying molecular pathways and pathophysiology.
Collapse
Affiliation(s)
- Meaad A. Almazmomi
- Pharmaceutical Care Department, Ministry of National Guard—Health Affairs, Jeddah, Saudi Arabia
- Pharmacology Department, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Mohammed Alsieni
- Pharmacology Department, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
13
|
Makarova YA, Ryabkova VA, Salukhov VV, Sagun BV, Korovin AE, Churilov LP. Atherosclerosis, Cardiovascular Disorders and COVID-19: Comorbid Pathogenesis. Diagnostics (Basel) 2023; 13:478. [PMID: 36766583 PMCID: PMC9914751 DOI: 10.3390/diagnostics13030478] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 01/19/2023] [Accepted: 01/26/2023] [Indexed: 02/01/2023] Open
Abstract
The article describes how atherosclerosis and coronavirus disease 19 (COVID-19) may affect each other. The features of this comorbid pathogenesis at various levels (vascular, cellular and molecular) are considered. A bidirectional influence of these conditions is described: the presence of cardiovascular diseases affects different individuals' susceptibility to viral infection. In turn, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) can have a negative effect on the endothelium and cardiomyocytes, causing blood clotting, secretion of pro-inflammatory cytokines, and thus exacerbating the development of atherosclerosis. In addition to the established entry into cells via angiotensin-converting enzyme 2 (ACE2), other mechanisms of SARS-CoV-2 entry are currently under investigation, for example, through CD147. Pathogenesis of comorbidity can be determined by the influence of the virus on various links which are meaningful for atherogenesis: generation of oxidized forms of low-density lipoproteins (LDL), launch of a cytokine storm, damage to the endothelial glycocalyx, and mitochondrial injury. The transformation of a stable plaque into an unstable one plays an important role in the pathogenesis of atherosclerosis complications and can be triggered by COVID-19. The impact of SARS-CoV-2 on large vessels such as the aorta is more complex than previously thought considering its impact on vasa vasorum. Current information on the mutual influence of the medicines used in the treatment of atherosclerosis and acute COVID-19 is briefly summarized.
Collapse
Affiliation(s)
- Yulia A. Makarova
- Laboratory of the Microangiopathic Mechanisms of Atherogenesis, Saint Petersburg State University, 199034 Saint-Petersburg, Russia
| | - Varvara A. Ryabkova
- Laboratory of the Microangiopathic Mechanisms of Atherogenesis, Saint Petersburg State University, 199034 Saint-Petersburg, Russia
- M.V. Chernorutsky Department of Internal Medicine (Hospital Course), Pavlov First Saint Petersburg State Medical University, 197022 Saint-Petersburg, Russia
| | - Vladimir V. Salukhov
- N.S. Molchanov 1st Clinic for the Improvement of Physicians, S.M. Kirov Military Medical Academy, 194044 Saint-Petersburg, Russia
| | - Boris V. Sagun
- N.S. Molchanov 1st Clinic for the Improvement of Physicians, S.M. Kirov Military Medical Academy, 194044 Saint-Petersburg, Russia
| | - Aleksandr E. Korovin
- Laboratory of the Microangiopathic Mechanisms of Atherogenesis, Saint Petersburg State University, 199034 Saint-Petersburg, Russia
- N.S. Molchanov 1st Clinic for the Improvement of Physicians, S.M. Kirov Military Medical Academy, 194044 Saint-Petersburg, Russia
| | - Leonid P. Churilov
- Laboratory of the Microangiopathic Mechanisms of Atherogenesis, Saint Petersburg State University, 199034 Saint-Petersburg, Russia
- Department of Experimental Tuberculosis, Saint Petersburg Research Institute of Phthisiopulmonology, 191036 Saint-Petersburg, Russia
| |
Collapse
|
14
|
Bayani F, Safaei Hashkavaei N, Karamian MR, Uskoković V, Sefidbakht Y. In silico design of a multi-epitope vaccine against the spike and the nucleocapsid proteins of the Omicron variant of SARS-CoV-2. J Biomol Struct Dyn 2023; 41:11748-11762. [PMID: 36703619 DOI: 10.1080/07391102.2023.2170470] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 12/22/2022] [Indexed: 01/28/2023]
Abstract
Computational studies can comprise an effective approach to treating and preventing viral infections. Since 2019, the world has been dealing with the outbreak of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). The most important achievement in this short period of time in the effort to reduce morbidity and mortality was the production of vaccines and effective antiviral drugs. Although the virus has been significantly suppressed, it continues to evolve, spread, and evade the host's immune system. Recently, researchers have turned to immunoinformatics tools to reduce side effects and save the time and cost of traditional vaccine production methods. In the present study, an attempt has been made to design a multi-epitope vaccine with humoral and cellular immune response stimulation against the Omicron variant of SARS-CoV-2 by investigating new mutations in spike (S) and nucleocapsid (N) proteins. The population coverage of the vaccine was evaluated as appropriate compared to other studies. The results of molecular dynamics simulation and molecular mechanics/generalized Born surface area (MM/GBSA) calculations predict the stability and proper interaction of the vaccine with Toll-like receptor 4 (TLR-4) as an innate immune receptor. The results of the immune simulation show a significant increase in the coordinated response of IgM and IgG after the third injection of the vaccine. Also, in the continuation of the research, spike proteins from BA.4 and BA.5 lineages were screened by immunoinformatics filters and effective epitopes were suggested for vaccine design. Despite the high precision of computational studies, in-vivo and in-vitro research is needed for final confirmation.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Fatemeh Bayani
- Protein Research Center, Shahid Beheshti University, Tehran, Iran
| | | | - Mohammad Reza Karamian
- Department of Cell and Molecular Biology, Faculty of Science, Kharazmi University, Tehran, Iran
| | - Vuk Uskoković
- TardigradeNano LLC, Irvine, CA, USA
- Department of Mechanical Engineering, San Diego State University, San Diego, CA, USA
| | - Yahya Sefidbakht
- Protein Research Center, Shahid Beheshti University, Tehran, Iran
| |
Collapse
|
15
|
Gholami M, Adibipour F, Valipour SM, Ulloa L, Motaghinejad M. Potential Regulation of NF-κB by Curcumin in Coronavirus-Induced Cytokine Storm and Lung Injury. Int J Prev Med 2022; 13:156. [PMID: 36911003 PMCID: PMC9999103 DOI: 10.4103/ijpvm.ijpvm_565_20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Accepted: 04/08/2022] [Indexed: 03/14/2023] Open
Abstract
The current pandemic coronavirus disease-19 (COVID-19) is still a global medical and economic emergency with over 244 million confirmed infections and over 4.95 million deaths by October 2021, in less than 2 years. Severe acute respiratory syndrome (SARS), the Middle East respiratory syndrome coronavirus (MERS), and COVID-19 are three recent coronavirus pandemics with major medical and economic implications. Currently, there is no effective treatment for these infections. One major pathological hallmark of these infections is the so-called 'cytokine storm,' which depicts an unregulated production of inflammatory cytokines inducing detrimental inflammation leading to organ injury and multiple organ failure including severe pulmonary, cardiovascular, and kidney failure in COVID-19. Several studies have suggested the potential of curcumin to inhibit the replication of some viruses similar to coronaviruses. Multiple experimental and clinical studies also reported the anti-inflammatory potential of curcumin in multiple infectious and inflammatory disorders. Thus, we hypothesized that curcumin may provide antiviral and anti-inflammatory effects for treating COVID-19. Although these studies suggest that curcumin could serve as an adjuvant treatment for COVID-19, its molecular mechanisms are still debated, especially its potential to modulate the toll-like receptors/TIR-domain-containing adapter-inducing interferon-β/nuclear factor kappa-light-chain-enhancer of activated B cells (TLR/TRIF/NF-κB) pathway. The preliminary results showed that curcumin modulates the nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) pathway, a common pathway controlling cytokine production in multiple infectious and inflammatory disorders. Here, we hypothesize and discuss whether curcumin treatment may provide antiviral and anti-inflammatory clinical advantages for treating COVID-19 by modulating the TLR/TRIF/NF-κB pathway. We also review the current data on curcumin and discuss potential experimental and clinical studies that require defining its potential clinical implications in COVID-19.
Collapse
Affiliation(s)
- Mina Gholami
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Adibipour
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Sanaz M. Valipour
- Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Luis Ulloa
- Center for Perioperative Organ Protection, Department of Anesthesiology, Duke University, Durham, NC, USA
| | - Majid Motaghinejad
- Chronic Respiratory Disease Research Center, National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
16
|
Heine H, Zamyatina A. Therapeutic Targeting of TLR4 for Inflammation, Infection, and Cancer: A Perspective for Disaccharide Lipid A Mimetics. Pharmaceuticals (Basel) 2022; 16:23. [PMID: 36678520 PMCID: PMC9864529 DOI: 10.3390/ph16010023] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 12/13/2022] [Accepted: 12/14/2022] [Indexed: 12/28/2022] Open
Abstract
The Toll-like receptor 4 (TLR4) signaling pathway plays a central role in the prompt defense against infectious challenge and provides immediate response to Gram-negative bacterial infection. The TLR4/MD-2 complex can sense and respond to various pathogen-associated molecular patterns (PAMPs) with bacterial lipopolysaccharide (LPS) being the most potent and the most frequently occurring activator of the TLR4-mediated inflammation. TLR4 is believed to be both a friend and foe since improperly regulated TLR4 signaling can result in the overactivation of immune responses leading to sepsis, acute lung injury, or pathologic chronic inflammation involved in cancer and autoimmune disease. TLR4 is also considered a legitimate target for vaccine adjuvant development since its activation can boost the adaptive immune responses. The dual action of the TLR4 complex justifies the efforts in the development of both TLR4 antagonists as antisepsis drug candidates or remedies for chronic inflammatory diseases and TLR4 agonists as vaccine adjuvants or immunotherapeutics. In this review, we provide a brief overview of the biochemical evidences for possible pharmacologic applications of TLR4 ligands as therapeutics and report our systematic studies on the design, synthesis, and immunobiological evaluation of carbohydrate-based TLR4 antagonists with nanomolar affinity for MD-2 as well as disaccharide-based TLR4 agonists with picomolar affinity for the TLR4/MD-2 complex.
Collapse
Affiliation(s)
- Holger Heine
- Research Group Innate Immunity, Research Center Borstel—Leibniz Lung Center, Airway Research Center North (ARCN), German Center for Lung Research (DZL), Parkallee 22, 23845 Borstel, Germany
| | - Alla Zamyatina
- Department of Chemistry, University of Natural Resources and Life Sciences, Muthgasse 18, 1190 Vienna, Austria
| |
Collapse
|
17
|
Ali HN, Niranji SS, Al-Jaf SMA. Association of Toll-like receptor-4 polymorphism with SARS CoV-2 infection in Kurdish Population. HUMAN GENE (AMSTERDAM, NETHERLANDS) 2022; 34:201115. [PMID: 37521442 PMCID: PMC9529343 DOI: 10.1016/j.humgen.2022.201115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Revised: 09/17/2022] [Accepted: 09/28/2022] [Indexed: 11/07/2022]
Abstract
Genetic variations are critical for understanding clinical outcomes of infections including server acute respiratory syndrome coronavirus 2 (SARS CoV-2). The immunological reactions of human immune genes with SARS CoV-2 have been under investigation. Toll-like receptors (TLRs), a group of proteins, are important for microbial detections including bacteria and viruses. TLR4 can sense both bacterial lipopolysaccharides (LPS) and endogenous oxidized phospholipids triggered by Covid-19 infection. Two TLR4 single nucleotide polymorphisms (SNPs), Asp299Gly and Thr399Ile have been linked to infectious diseases. No studies have focused on these SNPs in association with Covid-19. This study aims to reveal the association between Covid-19 infection with these SNPs by comparing a group of patients and a general population. Restriction fragment length polymorphisms (RFLP) were used to identify the TLR4 SNPs in both the general population (n = 114) and Covid-19 patient groups (n = 125). The results found no association between the TLR4 polymorphisms and Covid-19 infections as the data showed no statistically significant difference between the compared groups. This suggested that these TLR4 SNPs may not be associated with Covid-19 infections.
Collapse
Key Words
- ACE2, Angiotensin converting enzyme 2
- ARSD, Acute respiratory distress syndrome
- Covid-19
- Covid-19, Coronavirus disease 2019
- IL, Interleukin
- LPS, Lipopolysaccharides (LPS)
- MyD88, Myeloid differentiation factor
- PCR-RFLP, Polymerase chain reaction-restriction fragment polymorphism
- SARS CoV-2
- SARS CoV-2, Server acute respiratory syndrome coronavirus 2
- SNP
- SNPs, Single nucleotide polymorphisms
- TLR4
- TLRs, Toll-like receptors
- TNF-α, Tumour necrosis factor alpha;
- TRIF, TIR-domain-containing adapter-inducing interferon-β.
- polymorphism
Collapse
Affiliation(s)
- Hussein N Ali
- College of Medicine, University of Garmian, Kurdistan region, Iraq
| | - Sherko S Niranji
- College of Medicine, University of Garmian, Kurdistan region, Iraq
- Coronavirus Research and Identification Lab., University of Garmian, Kurdistan region, Iraq
- Department of Biology, College of Education, University of Garmian, Kurdistan region, Iraq
| | - Sirwan M A Al-Jaf
- College of Medicine, University of Garmian, Kurdistan region, Iraq
- Coronavirus Research and Identification Lab., University of Garmian, Kurdistan region, Iraq
- Department of Biology, College of Education, University of Garmian, Kurdistan region, Iraq
| |
Collapse
|
18
|
Alkhayyat SS, Al-kuraishy HM, Al-Gareeb AI, El-Bouseary MM, AboKamer AM, Batiha GES, Simal-Gandara J. Fenofibrate for COVID-19 and related complications as an approach to improve treatment outcomes: the missed key for Holy Grail. Inflamm Res 2022; 71:1159-1167. [PMID: 35941297 PMCID: PMC9360649 DOI: 10.1007/s00011-022-01615-w] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 07/05/2022] [Accepted: 07/17/2022] [Indexed: 12/18/2022] Open
Abstract
INTRODUCTION Fenofibrate is an agonist of peroxisome proliferator activated receptor alpha (PPAR-α), that possesses anti-inflammatory, antioxidant, and anti-thrombotic properties. Fenofibrate is effective against a variety of viral infections and different inflammatory disorders. Therefore, the aim of critical review was to overview the potential role of fenofibrate in the pathogenesis of SARS-CoV-2 and related complications. RESULTS By destabilizing SARS-CoV-2 spike protein and preventing it from binding angiotensin-converting enzyme 2 (ACE2), a receptor for SARS-CoV-2 entry, fenofibrate can reduce SARS-CoV-2 entry in human cells Fenofibrate also suppresses inflammatory signaling pathways, which decreases SARS-CoV-2 infection-related inflammatory alterations. In conclusion, fenofibrate anti-inflammatory, antioxidant, and antithrombotic capabilities may help to minimize the inflammatory and thrombotic consequences associated with SARSCoV-2 infection. Through attenuating the interaction between SARS-CoV-2 and ACE2, fenofibrate can directly reduce the risk of SARS-CoV-2 infection. CONCLUSIONS As a result, fenofibrate could be a potential treatment approach for COVID-19 control.
Collapse
Affiliation(s)
- Shadi Salem Alkhayyat
- Department of Internal Medicine, Faculty of Medicine, King Abdulaziz University, Jeddah, 21589 Saudi Arabia
| | - Hayder M. Al-kuraishy
- Department of Clinical Pharmacology and Medicine, College of Medicine, Al-Mustansiriyah University, Baghdad, Iraq
| | - Ali I. Al-Gareeb
- Department of Clinical Pharmacology and Medicine, College of Medicine, Al-Mustansiriyah University, Baghdad, Iraq
| | - Maisra M. El-Bouseary
- Department of Pharmaceutical Microbiology, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| | - Amal M. AboKamer
- Department of Pharmaceutical Microbiology, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour, 22511 Al Beheira Egypt
| | - Jesus Simal-Gandara
- Nutrition and Bromatology Group, Department of Analytical Chemistry and Food Science, Faculty Science, Universidade de Vigo, 32004 Ourense, Spain
| |
Collapse
|
19
|
Khan AA, Farooq F, Jain SK, Golinska P, Rai M. Comparative Host-Pathogen Interaction Analyses of SARS-CoV2 and Aspergillus fumigatus, and Pathogenesis of COVID-19-Associated Aspergillosis. MICROBIAL ECOLOGY 2022; 84:1236-1244. [PMID: 34738157 PMCID: PMC8568490 DOI: 10.1007/s00248-021-01913-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 10/25/2021] [Indexed: 05/03/2023]
Abstract
COVID-19 caused a global catastrophe with a large number of cases making it one of the major pandemics of the human history. The clinical presentations of the disease are continuously challenging healthcare workers with the variation of pandemic waves and viral variants. Recently, SARS-CoV2 patients have shown increased occurrence of invasive pulmonary aspergillosis infection even in the absence of traditional risk factors. The mechanism of COVID-19-associated aspergillosis is not completely understood and therefore, we performed this system biological study in order to identify mechanistic implications of aspergillosis susceptibility in COVID-19 patients and the important targets associated with this disease. We performed host-pathogen interaction (HPI) analysis of SARS-CoV2, and most common COVID-19-associated aspergillosis pathogen, Aspergillus fumigatus, using in silico approaches. The known host-pathogen interactions data of SARS-CoV2 was obtained from BIOGRID database. In addition, A. fumigatus host-pathogen interactions were predicted through homology modeling. The human targets interacting with both pathogens were separately analyzed for their involvement in aspergillosis. The aspergillosis human targets were screened from DisGeNet and GeneCards. The aspergillosis targets involved in both HPI were further analyzed for functional overrepresentation analysis using PANTHER. The results indicate that both pathogens interact with a number of aspergillosis targets and altogether they recruit more aspergillosis targets in host-pathogen interaction than alone. Common aspergillosis targets involved in HPI with both SARS-CoV2 and A. fumigatus can indicate strategies for the management of both conditions by modulating these common disease targets.
Collapse
Affiliation(s)
- Abdul Arif Khan
- Division of Microbiology, Indian Council of Medical Research-National AIDS Research Institute, Pune, Maharashtra, India.
| | - Fozia Farooq
- School of Studies in Microbiology, Vikram University, Ujjain, Madhya Pradesh, India
| | - Sudhir K Jain
- School of Studies in Microbiology, Vikram University, Ujjain, Madhya Pradesh, India
| | - Patrycja Golinska
- Department of Microbiology, Nicolaus Copernicus University, Torun, Poland
| | - Mahendra Rai
- Department of Microbiology, Nicolaus Copernicus University, Torun, Poland
- Department of Biotechnology, Sant Gadge Baba Amravati University, Amravati, Maharashtra, India
| |
Collapse
|
20
|
Villapalos-García G, Zubiaur P, Rivas-Durán R, Campos-Norte P, Arévalo-Román C, Fernández-Rico M, García-Fraile Fraile L, Fernández-Campos P, Soria-Chacartegui P, Fernández de Córdoba-Oñate S, Delgado-Wicke P, Fernández-Ruiz E, González-Álvaro I, Sanz J, Abad-Santos F, de Los Santos I. Transmembrane protease serine 2 ( TMPRSS2) rs75603675, comorbidity, and sex are the primary predictors of COVID-19 severity. Life Sci Alliance 2022; 5:e202201396. [PMID: 35636966 PMCID: PMC9152129 DOI: 10.26508/lsa.202201396] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 05/12/2022] [Accepted: 05/13/2022] [Indexed: 01/08/2023] Open
Abstract
By the end of December 2021, coronavirus disease 2019 (COVID-19) produced more than 271 million cases and 5.3 million deaths. Although vaccination is an effective strategy for pandemic control, it is not yet equally available in all countries. Therefore, identification of prognostic biomarkers remains crucial to manage COVID-19 patients. The aim of this study was to evaluate predictors of COVID-19 severity previously proposed. Clinical and demographic characteristics and 120 single-nucleotide polymorphisms were analyzed from 817 patients with COVID-19, who attended the emergency department of the Hospital Universitario de La Princesa during March and April 2020. The main outcome was a modified version of the 7-point World Health Organization (WHO) COVID-19 severity scale (WHOCS); both in the moment of the first hospital examination (WHOCS-1) and of the severest WHOCS score (WHOCS-2). The TMPRSS2 rs75603675 genotype (OR = 0.586), dyslipidemia (OR = 2.289), sex (OR = 0.586), and the Charlson Comorbidity Index (OR = 1.126) were identified as the main predictors of disease severity. Consequently, these variables might influence COVID-19 severity and could be used as predictors of disease development.
Collapse
Affiliation(s)
- Gonzalo Villapalos-García
- Clinical Pharmacology Department, Hospital Universitario La Princesa, Instituto Teófilo Hernando, Universidad Autónoma de Madrid (UAM), Instituto de Investigación Sanitaria La Princesa (IIS-IP), Madrid, Spain
| | - Pablo Zubiaur
- Clinical Pharmacology Department, Hospital Universitario La Princesa, Instituto Teófilo Hernando, Universidad Autónoma de Madrid (UAM), Instituto de Investigación Sanitaria La Princesa (IIS-IP), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, Spain
| | - Rebeca Rivas-Durán
- Infectious Diseases Unit, Hospital Universitario La Princesa, Instituto de Investigación Sanitaria La Princesa (IIS-IP), Madrid, Spain
| | - Pilar Campos-Norte
- Infectious Diseases Unit, Hospital Universitario La Princesa, Instituto de Investigación Sanitaria La Princesa (IIS-IP), Madrid, Spain
| | - Cristina Arévalo-Román
- Infectious Diseases Unit, Hospital Universitario La Princesa, Instituto de Investigación Sanitaria La Princesa (IIS-IP), Madrid, Spain
| | - Marta Fernández-Rico
- Infectious Diseases Unit, Hospital Universitario La Princesa, Instituto de Investigación Sanitaria La Princesa (IIS-IP), Madrid, Spain
| | - Lucio García-Fraile Fraile
- Infectious Diseases Unit, Hospital Universitario La Princesa, Instituto de Investigación Sanitaria La Princesa (IIS-IP), Madrid, Spain
| | - Paula Fernández-Campos
- Clinical Pharmacology Department, Hospital Universitario La Princesa, Instituto Teófilo Hernando, Universidad Autónoma de Madrid (UAM), Instituto de Investigación Sanitaria La Princesa (IIS-IP), Madrid, Spain
| | - Paula Soria-Chacartegui
- Clinical Pharmacology Department, Hospital Universitario La Princesa, Instituto Teófilo Hernando, Universidad Autónoma de Madrid (UAM), Instituto de Investigación Sanitaria La Princesa (IIS-IP), Madrid, Spain
| | - Sara Fernández de Córdoba-Oñate
- Molecular Biology Unit, Hospital Universitario La Princesa, Instituto de Investigación Sanitaria La Princesa (IIS-IP), Madrid, Spain
| | - Pablo Delgado-Wicke
- Molecular Biology Unit, Hospital Universitario La Princesa, Instituto de Investigación Sanitaria La Princesa (IIS-IP), Madrid, Spain
| | - Elena Fernández-Ruiz
- Molecular Biology Unit, Hospital Universitario La Princesa, Instituto de Investigación Sanitaria La Princesa (IIS-IP), Madrid, Spain
| | - Isidoro González-Álvaro
- Rheumatology Service, Hospital Universitario La Princesa, Instituto de Investigación Sanitaria La Princesa (IIS-IP), Madrid, Spain
| | - Jesús Sanz
- Infectious Diseases Unit, Hospital Universitario La Princesa, Instituto de Investigación Sanitaria La Princesa (IIS-IP), Madrid, Spain
| | - Francisco Abad-Santos
- Clinical Pharmacology Department, Hospital Universitario La Princesa, Instituto Teófilo Hernando, Universidad Autónoma de Madrid (UAM), Instituto de Investigación Sanitaria La Princesa (IIS-IP), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, Spain
| | - Ignacio de Los Santos
- Infectious Diseases Unit, Hospital Universitario La Princesa, Instituto de Investigación Sanitaria La Princesa (IIS-IP), Madrid, Spain
| |
Collapse
|
21
|
Cao H, Baranova A, Wei X, Wang C, Zhang F. Bidirectional causal associations between type 2 diabetes and COVID-19. J Med Virol 2022; 95:e28100. [PMID: 36029131 PMCID: PMC9538258 DOI: 10.1002/jmv.28100] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 08/22/2022] [Accepted: 08/25/2022] [Indexed: 01/11/2023]
Abstract
Observational studies have reported high comorbidity between type 2 diabetes (T2D) and severe COVID-19. However, the causality between T2D and COVID-19 has yet to be validated. We performed genetic correlation and Mendelian randomization (MR) analyses to assess genetic relationships and potential causal associations between T2D and three COVID-19 outcomes (severe acute respiratory syndrome coronavirus 2 [SARS-CoV-2] infection, COVID-19 hospitalization, and critical COVID-19). Molecular pathways connecting SARS-CoV-2 and COVID-19 were reconstructed to extract insights into the potential mechanisms underlying the connection. We identified a high genetic overlap between T2D and each COVID-19 outcome (genetic correlations 0.21-0.28). The MR analyses indicated that genetic liability to T2D confers a causal effect on hospitalized COVID-19 (odds ratio 1.08, 95% confidence interval [CI] 1.04-1.12) and critical COVID-19 (1.09, 1.03-1.16), while genetic liability to SARS-CoV-2 infection exerts a causal effect on T2D (1.25, 1.00-1.56). There was suggestive evidence that T2D was associated with an increased risk for SARS-CoV-2 infection (1.02, 1.00-1.03), while critical COVID-19 (1.06, 1.00-1.13) and hospitalized COVID-19 (1.09, 0.99-1.19) were associated with an increased risk for T2D. Pathway analysis identified a panel of immunity-related genes that may mediate the links between T2D and COVID-19 at the molecular level. Our study provides robust support for the bidirectional causal associations between T2D and COVID-19. T2D may contribute to amplifying the severity of COVID-19, while the liability to COVID-19 may increase the risk for T2D.
Collapse
Affiliation(s)
- Hongbao Cao
- School of Systems BiologyGeorge Mason UniversityManassasVirginiaUSA
| | - Ancha Baranova
- School of Systems BiologyGeorge Mason UniversityManassasVirginiaUSA,Research Centre for Medical GeneticsMoscowRussia
| | - Xuejuan Wei
- Fengtai District Fangzhuang Community Health Service Center in BeijingBeijingChina
| | - Chun Wang
- Department of Medical PsychologyThe Affiliated Brain Hospital of Nanjing Medical UniversityNanjingChina
| | - Fuquan Zhang
- Department of PsychiatryThe Affiliated Brain Hospital of Nanjing Medical UniversityNanjingChina,Institute of NeuropsychiatryThe Affiliated Brain Hospital of Nanjing Medical UniversityNanjingChina
| |
Collapse
|
22
|
Tangos M, Budde H, Kolijn D, Sieme M, Zhazykbayeva S, Lódi M, Herwig M, Gömöri K, Hassoun R, Robinson EL, Meister TL, Jaquet K, Kovács Á, Mustroph J, Evert K, Babel N, Fagyas M, Lindner D, Püschel K, Westermann D, Mannherz HG, Paneni F, Pfaender S, Tóth A, Mügge A, Sossalla S, Hamdani N. SARS-CoV-2 infects human cardiomyocytes promoted by inflammation and oxidative stress. Int J Cardiol 2022; 362:196-205. [PMID: 35643215 PMCID: PMC9132721 DOI: 10.1016/j.ijcard.2022.05.055] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 04/11/2022] [Accepted: 05/23/2022] [Indexed: 12/15/2022]
Abstract
Introduction The respiratory illness triggered by severe acute respiratory syndrome virus-2 (SARS-CoV-2) is often particularly serious or fatal amongst patients with pre-existing heart conditions. Although the mechanisms underlying SARS-CoV-2-related cardiac damage remain elusive, inflammation (i.e. ‘cytokine storm’) and oxidative stress are likely involved. Methods and results Here we sought to determine: 1) if cardiomyocytes are targeted by SARS-CoV-2 and 2) how inflammation and oxidative stress promote the viral entry into cardiac cells. We analysed pro-inflammatory and oxidative stress and its impact on virus entry and virus-associated cardiac damage from SARS-CoV-2 infected patients and compared it to left ventricular myocardial tissues obtained from non-infected transplanted hearts either from end stage heart failure or non-failing hearts (donor group). We found that neuropilin-1 potentiates SARS-CoV-2 entry into human cardiomyocytes, a phenomenon driven by inflammatory and oxidant signals. These changes accounted for increased proteases activity and apoptotic markers thus leading to cell damage and apoptosis. Conclusion This study provides new insights into the mechanisms of SARS-CoV-2 entry into the heart and defines promising targets for antiviral interventions for COVID-19 patients with pre-existing heart conditions or patients with co-morbidities.
Collapse
Affiliation(s)
- Melina Tangos
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany; Department of Cardiology, St. Josef-Hospital, Ruhr University Bochum, Bochum, Germany; Institute of Physiology, Ruhr University Bochum, Bochum, Germany
| | - Heidi Budde
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany; Department of Cardiology, St. Josef-Hospital, Ruhr University Bochum, Bochum, Germany; Institute of Physiology, Ruhr University Bochum, Bochum, Germany
| | - Detmar Kolijn
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany; Department of Cardiology, St. Josef-Hospital, Ruhr University Bochum, Bochum, Germany; Institute of Physiology, Ruhr University Bochum, Bochum, Germany
| | - Marcel Sieme
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany; Department of Cardiology, St. Josef-Hospital, Ruhr University Bochum, Bochum, Germany; Institute of Physiology, Ruhr University Bochum, Bochum, Germany
| | - Saltanat Zhazykbayeva
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany; Department of Cardiology, St. Josef-Hospital, Ruhr University Bochum, Bochum, Germany; Institute of Physiology, Ruhr University Bochum, Bochum, Germany
| | - Mária Lódi
- Department of Neuroanatomy and Molecular Brain Research, Ruhr University Bochum, Medical Faculty, Bochum, Germany
| | - Melissa Herwig
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany; Department of Cardiology, St. Josef-Hospital, Ruhr University Bochum, Bochum, Germany; Institute of Physiology, Ruhr University Bochum, Bochum, Germany
| | - Kamilla Gömöri
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany; Department of Cardiology, St. Josef-Hospital, Ruhr University Bochum, Bochum, Germany; Institute of Physiology, Ruhr University Bochum, Bochum, Germany
| | - Roua Hassoun
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany; Department of Cardiology, St. Josef-Hospital, Ruhr University Bochum, Bochum, Germany; Institute of Physiology, Ruhr University Bochum, Bochum, Germany
| | - Emma Louise Robinson
- School of Medicine, Division of Cardiology, University of Colorado Anschutz Medical Campus, Aurora, United States of America
| | - Toni Luise Meister
- Department of Molecular and Medical Virology, Ruhr University Bochum, Bochum, Germany
| | - Kornelia Jaquet
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany; Department of Cardiology, St. Josef-Hospital, Ruhr University Bochum, Bochum, Germany
| | - Árpád Kovács
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany; Department of Cardiology, St. Josef-Hospital, Ruhr University Bochum, Bochum, Germany; Institute of Physiology, Ruhr University Bochum, Bochum, Germany
| | - Julian Mustroph
- Department of Internal Medicine II, University Medical Center Regensburg, Regensburg, Germany
| | - Katja Evert
- Institute of Pathology, University Hospital Regensburg, Regensburg, Germany
| | - Nina Babel
- Center for Translational Medicine and Immune Diagnostics Laboratory, Medical Department I, Marien Hospital Herne, University Hospital of the Ruhr University Bochum, Bochum, Germany
| | - Miklós Fagyas
- Center for Molecular Cardiology, University of Zürich, University Heart Center, Cardiology, University Hospital Zurich, Zürich, Switzerland
| | - Diana Lindner
- Department of Cardiology and Angiology, University Heart Center Freiburg-Bad Krozingen, Germany
| | - Klaus Püschel
- Department of Legal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Dirk Westermann
- Department of Cardiology and Angiology, University Heart Center Freiburg-Bad Krozingen, Germany
| | - Hans Georg Mannherz
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany; Department of Anatomy and Molecular Embryology, Ruhr University Bochum, Bochum, Germany
| | - Francesco Paneni
- Center for Molecular Cardiology, University of Zürich, University Heart Center, Cardiology, University Hospital Zurich, Zürich, Switzerland; University Heart Center, Cardiology, Department of Research and Education, University Hospital Zurich, Zürich, Switzerland
| | - Stephanie Pfaender
- Department of Molecular and Medical Virology, Ruhr University Bochum, Bochum, Germany
| | - Attila Tóth
- Division of Clinical Physiology, Department of Cardiology, Faculty of Medicine, Debrecen, Hungary
| | - Andreas Mügge
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany; Department of Cardiology, St. Josef-Hospital, Ruhr University Bochum, Bochum, Germany
| | - Samuel Sossalla
- Department of Internal Medicine II, University Medical Center Regensburg, Regensburg, Germany; Clinic for Cardiology & Pneumology, Georg-August University Goettingen, DZHK (German Centre for Cardiovascular Research), partner site Goettingen, Germany
| | - Nazha Hamdani
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany; Department of Cardiology, St. Josef-Hospital, Ruhr University Bochum, Bochum, Germany; Institute of Physiology, Ruhr University Bochum, Bochum, Germany.
| |
Collapse
|
23
|
Dopamine Reduces SARS-CoV-2 Replication In Vitro through Downregulation of D2 Receptors and Upregulation of Type-I Interferons. Cells 2022; 11:cells11101691. [PMID: 35626728 PMCID: PMC9139638 DOI: 10.3390/cells11101691] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 05/13/2022] [Accepted: 05/18/2022] [Indexed: 02/06/2023] Open
Abstract
Recent evidence suggests that SARS-CoV-2 hinders immune responses via dopamine (DA)-related mechanisms. Nonetheless, studies addressing the specific role of DA in the frame of SARS-CoV-2 infection are still missing. In the present study, we investigate the role of DA in SARS-CoV-2 replication along with potential links with innate immune pathways in CaLu-3 human epithelial lung cells. We document here for the first time that, besides DA synthetic pathways, SARS-CoV-2 alters the expression of D1 and D2 DA receptors (D1DR, D2DR), while DA administration reduces viral replication. Such an effect occurs at non-toxic, micromolar-range DA doses, which are known to induce receptor desensitization and downregulation. Indeed, the antiviral effects of DA were associated with a robust downregulation of D2DRs both at mRNA and protein levels, while the amount of D1DRs was not significantly affected. While halting SARS-CoV-2 replication, DA, similar to the D2DR agonist quinpirole, upregulates the expression of ISGs and Type-I IFNs, which goes along with the downregulation of various pro-inflammatory mediators. In turn, administration of Type-I IFNs, while dramatically reducing SARS-CoV-2 replication, converges in downregulating D2DRs expression. Besides configuring the CaLu-3 cell line as a suitable model to study SARS-CoV-2-induced alterations at the level of the DA system in the periphery, our findings disclose a previously unappreciated correlation between DA pathways and Type-I IFN response, which may be disrupted by SARS-CoV-2 for host cell invasion and replication.
Collapse
|
24
|
Jiménez D, Torres Arias M. Immunouniverse of SARS-CoV-2. Immunol Med 2022; 45:186-224. [PMID: 35502127 DOI: 10.1080/25785826.2022.2066251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
Abstract
SARS-CoV-2 virus has become a global health problem that has caused millions of deaths worldwide. The infection can present with multiple clinical features ranging from asymptomatic or mildly symptomatic patients to patients with severe or critical illness that can even lead to death. Although the immune system plays an important role in pathogen control, SARS-CoV-2 can drive dysregulation of this response and trigger severe immunopathology. Exploring the mechanisms of the immune response involved in host defense against SARS-CoV-2 allows us to understand its immunopathogenesis and possibly detect features that can be used as potential therapies to eliminate the virus. The main objective of this review on SARS-CoV-2 is to highlight the interaction between the virus and the immune response. We explore the function and action of the immune system, the expression of molecules at the site of infection that cause hyperinflammation and hypercoagulation disorders, the factors leading to the development of pneumonia and subsequent severe acute respiratory distress syndrome which is the leading cause of death in patients with COVID-19.
Collapse
Affiliation(s)
- Dennis Jiménez
- Departamento de Ciencias de la Vida y Agricultura, Carrera de Ingeniería en Biotecnología, Universidad de las Fuerzas Armadas ESPE, Sangolquí, Pichincha, Ecuador
| | - Marbel Torres Arias
- Departamento de Ciencias de la Vida y Agricultura, Carrera de Ingeniería en Biotecnología, Universidad de las Fuerzas Armadas ESPE, Sangolquí, Pichincha, Ecuador.,Laboratorio de Inmunología y Virología, CENCINAT, GISAH, Universidad de las Fuerzas Armadas, Sangolquí, Pichincha, Ecuador
| |
Collapse
|
25
|
Low-grade inflammation, CoVID-19, and obesity: clinical aspect and molecular insights in childhood and adulthood. Int J Obes (Lond) 2022; 46:1254-1261. [PMID: 35393519 PMCID: PMC8988546 DOI: 10.1038/s41366-022-01111-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 02/21/2022] [Accepted: 03/08/2022] [Indexed: 12/12/2022]
Abstract
The new 2019 coronavirus 19 disease (CoVID-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) poses a serious threat to health systems. As a global health problem, this pandemic poses a huge threat to people and is responsible for significant morbidity and mortality worldwide. On the other hand, obesity has also reached epidemic proportions and poses another challenge to the healthcare system. There is increasing evidence of a strong association between obesity and CoVID-19 disease, but the mechanisms underlying the link between the two remain unclear and the role of obesity also remains to be elucidated. In particular obesity-related low-grade inflammation has been hypothesized as the Achille's heel that could predispose subjects with obesity to a more severe CoVID-19 compared to subjects with normal weight. Hence, we summarized recent evidence on the role of low-grade inflammation in clinical aspects of CoVID-19 in subjects with obesity in both childhood and adulthood. Further, we provide molecular insights to explain this link.
Collapse
|
26
|
Designing AbhiSCoVac - A single potential vaccine for all ‘corona culprits’: Immunoinformatics and immune simulation approaches. J Mol Liq 2022; 351:118633. [PMID: 35125571 PMCID: PMC8801591 DOI: 10.1016/j.molliq.2022.118633] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 01/21/2022] [Accepted: 01/26/2022] [Indexed: 12/12/2022]
|
27
|
Asano T, Chelvanambi S, Decano JL, Whelan MC, Aikawa E, Aikawa M. In silico Drug Screening Approach Using L1000-Based Connectivity Map and Its Application to COVID-19. Front Cardiovasc Med 2022; 9:842641. [PMID: 35402570 PMCID: PMC8989014 DOI: 10.3389/fcvm.2022.842641] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 02/14/2022] [Indexed: 01/25/2023] Open
Abstract
Conventional drug screening methods search for a limited number of small molecules that directly interact with the target protein. This process can be slow, cumbersome and has driven the need for developing new drug screening approaches to counter rapidly emerging diseases such as COVID-19. We propose a pipeline for drug repurposing combining in silico drug candidate identification followed by in vitro characterization of these candidates. We first identified a gene target of interest, the entry receptor for the SARS-CoV-2 virus, angiotensin converting enzyme 2 (ACE2). Next, we employed a gene expression profile database, L1000-based Connectivity Map to query gene expression patterns in lung epithelial cells, which act as the primary site of SARS-CoV-2 infection. Using gene expression profiles from 5 different lung epithelial cell lines, we computationally identified 17 small molecules that were predicted to decrease ACE2 expression. We further performed a streamlined validation in the normal human epithelial cell line BEAS-2B to demonstrate that these compounds can indeed decrease ACE2 surface expression and to profile cell health and viability upon drug treatment. This proposed pipeline combining in silico drug compound identification and in vitro expression and viability characterization in relevant cell types can aid in the repurposing of FDA-approved drugs to combat rapidly emerging diseases.
Collapse
Affiliation(s)
- Takaharu Asano
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States
| | - Sarvesh Chelvanambi
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States
| | - Julius L. Decano
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States
| | - Mary C. Whelan
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States
| | - Elena Aikawa
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States
- Center for Excellence in Vascular Biology, Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States
- Department of Human Pathology, I.M. Sechenov First Moscow State Medical University of the Ministry of Health, Moscow, Russia
| | - Masanori Aikawa
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States
- Center for Excellence in Vascular Biology, Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States
- Department of Human Pathology, I.M. Sechenov First Moscow State Medical University of the Ministry of Health, Moscow, Russia
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States
| |
Collapse
|
28
|
Zhang J, Ejikemeuwa A, Gerzanich V, Nasr M, Tang Q, Simard JM, Zhao RY. Understanding the Role of SARS-CoV-2 ORF3a in Viral Pathogenesis and COVID-19. Front Microbiol 2022; 13:854567. [PMID: 35356515 PMCID: PMC8959714 DOI: 10.3389/fmicb.2022.854567] [Citation(s) in RCA: 43] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 02/09/2022] [Indexed: 12/11/2022] Open
Abstract
The ongoing SARS-CoV-2 pandemic has shocked the world due to its persistence, COVID-19-related morbidity and mortality, and the high mutability of the virus. One of the major concerns is the emergence of new viral variants that may increase viral transmission and disease severity. In addition to mutations of spike protein, mutations of viral proteins that affect virulence, such as ORF3a, also must be considered. The purpose of this article is to review the current literature on ORF3a, to summarize the molecular actions of SARS-CoV-2 ORF3a, and its role in viral pathogenesis and COVID-19. ORF3a is a polymorphic, multifunctional viral protein that is specific to SARS-CoV/SARS-CoV-2. It was acquired from β-CoV lineage and likely originated from bats through viral evolution. SARS-CoV-2 ORF3a is a viroporin that interferes with ion channel activities in host plasma and endomembranes. It is likely a virion-associated protein that exerts its effect on the viral life cycle during viral entry through endocytosis, endomembrane-associated viral transcription and replication, and viral release through exocytosis. ORF3a induces cellular innate and pro-inflammatory immune responses that can trigger a cytokine storm, especially under hypoxic conditions, by activating NLRP3 inflammasomes, HMGB1, and HIF-1α to promote the production of pro-inflammatory cytokines and chemokines. ORF3a induces cell death through apoptosis, necrosis, and pyroptosis, which leads to tissue damage that affects the severity of COVID-19. ORF3a continues to evolve along with spike and other viral proteins to adapt in the human cellular environment. How the emerging ORF3a mutations alter the function of SARS-CoV-2 ORF3a and its role in viral pathogenesis and COVID-19 is largely unknown. This review provides an in-depth analysis of ORF3a protein's structure, origin, evolution, and mutant variants, and how these characteristics affect its functional role in viral pathogenesis and COVID-19.
Collapse
Affiliation(s)
- Jiantao Zhang
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD, United States
- Research and Development Service, VA Maryland Health Care System, Baltimore, MD, United States
| | - Amara Ejikemeuwa
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Volodymyr Gerzanich
- Research and Development Service, VA Maryland Health Care System, Baltimore, MD, United States
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Mohamed Nasr
- Drug Development and Clinical Sciences Branch, Division of AIDS, NIAID, NIH, Bethesda, MD, United States
| | - Qiyi Tang
- Department of Microbiology, Howard University College of Medicine, Washington, DC, United States
| | - J. Marc Simard
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD, United States
- Research and Development Service, VA Maryland Health Care System, Baltimore, MD, United States
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Richard Y. Zhao
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD, United States
- Research and Development Service, VA Maryland Health Care System, Baltimore, MD, United States
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, United States
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD, United States
- Institute of Global Health, University of Maryland School of Medicine, Baltimore, MD, United States
| |
Collapse
|
29
|
Espinoza C, Alarcón M. The Immune Response to SARS-CoV-2: Mechanisms, Aging, Sequelae and Vaccines. Mini Rev Med Chem 2022; 22:2166-2185. [PMID: 35249484 DOI: 10.2174/1389557522666220304231537] [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: 08/30/2021] [Revised: 10/28/2021] [Accepted: 12/08/2021] [Indexed: 11/22/2022]
Abstract
This review seeks to clarify the factors involved in the various immune responses to SARS-CoV-2 infection and the mechanisms that influence the development of COVID-19 with severe evolution. The innate immune response that evolves against SARS-CoV-2 in a complex way is highlighted, integrating multiple pathways by coronaviruses to evade it, in addition to characterizing the adaptive immune response, which can lead to an effective immune response or can contribute to immunopathological imbalance. In turn, host-dependent biomarkers such as age, gender, ABO blood group, and risk factors that contribute to the critical and varied progress of COVID-19 immunopathogenesis were analyzed. Finally, the potential vaccine candidates are presented, capable of generating immune protection with humoral and/or cellular neutralizing responses, in favor of blocking and destroying both the new human coronavirus and its variants, which cause the current pandemic.
Collapse
Affiliation(s)
- Carolina Espinoza
- Department of Clinical Biochemistry and Immunohaematology, Faculty of Health Sciences, Universidad de Talca, Talca, Chile
- Thrombosis Research Center, Universidad de Talca, Talca, Chile
| | - Marcelo Alarcón
- Department of Clinical Biochemistry and Immunohaematology, Faculty of Health Sciences, Universidad de Talca, Talca, Chile
- Thrombosis Research Center, Universidad de Talca, Talca, Chile
| |
Collapse
|
30
|
Mukherjee S. Toll-like receptor 4 in COVID-19: friend or foe? Future Virol 2022; 17:10.2217/fvl-2021-0249. [PMID: 35462619 PMCID: PMC9017673 DOI: 10.2217/fvl-2021-0249] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Accepted: 03/30/2022] [Indexed: 01/04/2023]
Abstract
Toll-like receptor 4, an innate immune sensor, is one of the key 'fate-deciding' regulators of immunity as well as COVID-19 immunopathogenesis. Suitable targeting of Toll-like receptor 4 appears to be an effective strategy to counteract the pandemic.
Collapse
Affiliation(s)
- Suprabhat Mukherjee
- Integrative Biochemistry & Immunology Laboratory, Department of Animal Science, Kazi Nazrul University, Asansol, West Bengal, 713340, India
| |
Collapse
|
31
|
Hansur L, Louisa M, Wuyung PE, Fadilah F. Daphnoretin from Carthamus tinctorius as a Potential Inflammatory Inhibitor in COVID-19 by Binding to Toll-like Receptor-4: An in silico Molecular Docking Study. Open Access Maced J Med Sci 2022. [DOI: 10.3889/oamjms.2022.7961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND: Cytokine storm in COVID-19 patients has contributed to many morbidities and mortalities in patients. Studies have found that toll-like receptors (TLRs) and some Fc receptors play essential roles in the hyperactivation of the immune system. Up to date, researchers are still in progress to discover effective and safe drugs to alleviate the hyperinflammatory state in COVID-19. The previous studies had shown that Carthamus tinctorius and its bioactive compounds might have anti-inflammatory activities in animal models.
AIM: We aimed to investigate the possible interactions of several flavonoids from C. tinctorius with several immune system components using a biocomputational approach.
METHODS: Molecular docking was done using the AutoDock program based on the Kyoto Encyclopedia of Genes and Genomes (KEGG) COVID-19 pathway. The most suitable receptors found were studied to study the interactions with several flavonoids from C. tinctorius.
RESULTS: TLR4, TLR8, and FcγRIIa were found to bind with SARS CoV2 inflammatory pathway and further selected as macromolecules for potential interactions study with 22 flavonoids from C. tinctorius. Of the 22 flavonoids studied, daphnoretin showed the best binding affinity with TLR4 and Rutin was shown to attach best with FcγRIIa. Unlike its excellent binding to TLR4, daphnoretin showed weak binding to TLR8.
CONCLUSION: Daphnoretin showed an excellent affinity with TLR4 and might be a good candidate as an inhibitor in hyperinflammatory reactions in COVID-19 DTLR8.
Collapse
|
32
|
Feng S, Song F, Guo W, Tan J, Zhang X, Qiao F, Guo J, Zhang L, Jia X. Potential Genes Associated with COVID-19 and Comorbidity. Int J Med Sci 2022; 19:402-415. [PMID: 35165525 PMCID: PMC8795808 DOI: 10.7150/ijms.67815] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 01/05/2022] [Indexed: 11/10/2022] Open
Abstract
Hypertension, diabetes mellitus, and coronary artery disease are common comorbidities and dangerous factors for infection and serious COVID-19. Polymorphisms in genes associated with comorbidities may help observe susceptibility and disease severity variation. However, specific genetic factors and the extent to which they can explain variation in susceptibility of severity are unclear. Therefore, we evaluated candidate genes associated with COVID-19 and hypertension, diabetes mellitus, and coronary artery disease. In particular, we performed searches against OMIM, NCBI, and other databases, protein-protein interaction network construction, and GO and KEGG pathway enrichment analyses. Results showed that the associated overlapping genes were TLR4, NLRP3, MBL2, IL6, IL1RN, IL1B, CX3CR1, CCR5, AGT, ACE, and F2. GO and KEGG analyses yielded 302 GO terms (q < 0.05) and 29 signaling pathways (q < 0.05), respectively, mainly including coronavirus disease-COVID-19 and cytokine-cytokine receptor interaction. IL6 and AGT were central in the PPI, with 8 and 5 connections, respectively. In this study, we identified 11 genes associated with both COVID-19 and three comorbidities that may contribute to infection and disease severity. The key genes IL6 and AGT are involved in regulating immune response, cytokine activity, and viral infection. Therefore, RAAS inhibitors, AGT antisense nucleotides, cytokine inhibitors, vitamin D, fenofibrate, and vaccines regulating non-immune and immune factors could be potential strategies to prevent and cure COVID-19. The study provides a basis for further investigation of genes and pathways with predictive value for the risk of infection and prognosis and could help guide drug and vaccine development to improve treatment efficacy and the development of personalised treatments, especially for COVID-19 individuals with common comorbidities.
Collapse
Affiliation(s)
- Shanshan Feng
- College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
- Non-coding RNA and Drug Discovery Key Laboratory of Sichuan Province, Chengdu Medical College, Chengdu, Sichuan, China
| | - Fuqiang Song
- Department of medical Laboratory, The General Hospital of Western Theater Command, Chengdu, China
| | | | - Jishan Tan
- Department of medical Laboratory, The General Hospital of Western Theater Command, Chengdu, China
| | - Xianqin Zhang
- School of Basic Medical Sciences, Chengdu Medical College, Chengdu, Sichuan, China
| | - Fengling Qiao
- College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Jinlin Guo
- Key Laboratory of Systematic Research of Distinctive Chinese Medicine Resources in Southwest China, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Chongqing Key Laboratory of Sichuan-Chongqing Co-construction for Diagnosis and Treatment of Infectious Diseases Integrated Traditional Chinese and Western Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Lin Zhang
- Department of Pharmacy, Shaoxing People's Hospital, Shaoxing Hospital, Zhejiang University School of Medicine, Shaoxing, China
| | - Xu Jia
- Non-coding RNA and Drug Discovery Key Laboratory of Sichuan Province, Chengdu Medical College, Chengdu, Sichuan, China
- School of Basic Medical Sciences, Chengdu Medical College, Chengdu, Sichuan, China
| |
Collapse
|
33
|
Zepeda-Cervantes J, Martínez-Flores D, Ramírez-Jarquín JO, Tecalco-Cruz ÁC, Alavez-Pérez NS, Vaca L, Sarmiento-Silva RE. Implications of the Immune Polymorphisms of the Host and the Genetic Variability of SARS-CoV-2 in the Development of COVID-19. Viruses 2022; 14:94. [PMID: 35062298 PMCID: PMC8778858 DOI: 10.3390/v14010094] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 12/22/2021] [Accepted: 12/28/2021] [Indexed: 01/08/2023] Open
Abstract
The severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) is responsible for the current pandemic affecting almost all countries in the world. SARS-CoV-2 is the agent responsible for coronavirus disease 19 (COVID-19), which has claimed millions of lives around the world. In most patients, SARS-CoV-2 infection does not cause clinical signs. However, some infected people develop symptoms, which include loss of smell or taste, fever, dry cough, headache, severe pneumonia, as well as coagulation disorders. The aim of this work is to report genetic factors of SARS-CoV-2 and host-associated to severe COVID-19, placing special emphasis on the viral entry and molecules of the immune system involved with viral infection. Besides this, we analyze SARS-CoV-2 variants and their structural characteristics related to the binding to polymorphic angiotensin-converting enzyme type 2 (ACE2). Additionally, we also review other polymorphisms as well as some epigenetic factors involved in the immunopathogenesis of COVID-19. These factors and viral variability could explain the increment of infection rate and/or in the development of severe COVID-19.
Collapse
Affiliation(s)
- Jesús Zepeda-Cervantes
- Departamento de Microbiología e Inmunología, Facultad de Medicina Veterinaria y Zootecnia, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico;
| | - Daniel Martínez-Flores
- Departamento de Biología Celular y del Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico;
| | - Josué Orlando Ramírez-Jarquín
- Departamento de Neuropatología Molecular, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico;
| | - Ángeles C. Tecalco-Cruz
- Posgrado en Ciencias Genómicas, Universidad Autónoma de la Ciudad de México (UACM), Mexico City 06720, Mexico;
| | - Noé Santiago Alavez-Pérez
- Escuela Nacional de Medicina y Homeopatía, Instituto Politécnico Nacional, Mexico City 07340, Mexico;
| | - Luis Vaca
- Departamento de Biología Celular y del Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico;
| | - Rosa Elena Sarmiento-Silva
- Departamento de Microbiología e Inmunología, Facultad de Medicina Veterinaria y Zootecnia, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico;
| |
Collapse
|
34
|
Alves HR, Lomba GSB, Gonçalves-de-Albuquerque CF, Burth P. Irisin, Exercise, and COVID-19. Front Endocrinol (Lausanne) 2022; 13:879066. [PMID: 35784579 PMCID: PMC9248970 DOI: 10.3389/fendo.2022.879066] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 04/28/2022] [Indexed: 12/12/2022] Open
Abstract
Muscle and adipose tissue produce irisin during exercise. Irisin is thermogenic adipomyokine, improves glucose and lipid metabolism, and ameliorates the effects of obesity-driven inflammation, metabolic syndrome, and diabetes. In addition, exercise-induced irisin activates anti-inflammatory pathways and may play an essential role in improving the outcomes of inflammatory conditions, such as coronavirus disease (COVID-19). COVID-19 infection can activate different intracellular receptors and modulate various pathways during the course of the disease. The cytokine release storm (CRS) produced is significant because it promotes the context for systemic inflammation, which increases the risk of mortality in patients with severe acute respiratory syndrome coronavirus 2 (SARS-CoV2). In addition, viral infection and the resulting organ damage may stimulate the mitogen-activated protein kinase(MAPK) and toll-like receptor 4 (TLR4)/toll interleukin receptor (TIR)-domain-containing adaptor (MyD88) pathways while negatively modulating the AMP-activated protein kinase (AMPK) pathway, leading to increased inflammatory cytokine production. Exercise-induced irisin may counteract this inflammatory modulation by decreasing cytokine production. Consequently, increased irisin levels, as found in healthy patients, may favor a better prognosis in patients with SARS-CoV2. This review aims to explore the molecular mechanisms underlying the anti-inflammatory properties of irisin in mitigating CRS and preventing severe outcomes due to infection with SARS-CoV2.
Collapse
Affiliation(s)
- Hugo Rodrigues Alves
- Department of Cell and Molecular Biology, Fluminense Federal University, Niterói, Brazil
| | | | - Cassiano Felippe Gonçalves-de-Albuquerque
- Laboratory of Immunopharmacology, Federal University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
- Postgraduate Program in Biotechnology, Fluminense Federal University, Rio de Janeiro, Brazil
- *Correspondence: Patricia Burth, ; Cassiano Felippe Gonçalves-de-Albuquerque,
| | - Patricia Burth
- Department of Cell and Molecular Biology, Fluminense Federal University, Niterói, Brazil
- Postgraduate Program in Biotechnology, Fluminense Federal University, Rio de Janeiro, Brazil
- *Correspondence: Patricia Burth, ; Cassiano Felippe Gonçalves-de-Albuquerque,
| |
Collapse
|
35
|
Palacios-Rápalo SN, De Jesús-González LA, Cordero-Rivera CD, Farfan-Morales CN, Osuna-Ramos JF, Martínez-Mier G, Quistián-Galván J, Muñoz-Pérez A, Bernal-Dolores V, del Ángel RM, Reyes-Ruiz JM. Cholesterol-Rich Lipid Rafts as Platforms for SARS-CoV-2 Entry. Front Immunol 2021; 12:796855. [PMID: 34975904 PMCID: PMC8719300 DOI: 10.3389/fimmu.2021.796855] [Citation(s) in RCA: 63] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 11/29/2021] [Indexed: 12/13/2022] Open
Abstract
Since its appearance, the Severe Acute Respiratory Syndrome Coronavirus (SARS-CoV-2), the causal agent of Coronavirus Disease 2019 (COVID-19), represents a global problem for human health that involves the host lipid homeostasis. Regarding, lipid rafts are functional membrane microdomains with highly and tightly packed lipid molecules. These regions enriched in sphingolipids and cholesterol recruit and concentrate several receptors and molecules involved in pathogen recognition and cellular signaling. Cholesterol-rich lipid rafts have multiple functions for viral replication; however, their role in SARS-CoV-2 infection remains unclear. In this review, we discussed the novel evidence on the cholesterol-rich lipid rafts as a platform for SARS-CoV-2 entry, where receptors such as the angiotensin-converting enzyme-2 (ACE-2), heparan sulfate proteoglycans (HSPGs), human Toll-like receptors (TLRs), transmembrane serine proteases (TMPRSS), CD-147 and HDL-scavenger receptor B type 1 (SR-B1) are recruited for their interaction with the viral spike protein. FDA-approved drugs such as statins, metformin, hydroxychloroquine, and cyclodextrins (methyl-β-cyclodextrin) can disrupt cholesterol-rich lipid rafts to regulate key molecules in the immune signaling pathways triggered by SARS-CoV-2 infection. Taken together, better knowledge on cholesterol-rich lipid rafts in the SARS-CoV-2-host interactions will provide valuable insights into pathogenesis and the identification of novel therapeutic targets.
Collapse
Affiliation(s)
- Selvin Noé Palacios-Rápalo
- Department of Infectomics and Molecular Pathogenesis, Center for Research and Advanced Studies (CINVESTAV-IPN), Mexico City, Mexico
| | - Luis Adrián De Jesús-González
- Department of Infectomics and Molecular Pathogenesis, Center for Research and Advanced Studies (CINVESTAV-IPN), Mexico City, Mexico
| | - Carlos Daniel Cordero-Rivera
- Department of Infectomics and Molecular Pathogenesis, Center for Research and Advanced Studies (CINVESTAV-IPN), Mexico City, Mexico
| | - Carlos Noe Farfan-Morales
- Department of Infectomics and Molecular Pathogenesis, Center for Research and Advanced Studies (CINVESTAV-IPN), Mexico City, Mexico
| | - Juan Fidel Osuna-Ramos
- Department of Infectomics and Molecular Pathogenesis, Center for Research and Advanced Studies (CINVESTAV-IPN), Mexico City, Mexico
| | - Gustavo Martínez-Mier
- Unidad Médica de Alta Especialidad, Hospital de Especialidades No. 14, Centro Médico Nacional “Adolfo Ruiz Cortines”, Instituto Mexicano del Seguro Social (IMSS) Veracruz Norte, Veracruz, Mexico
| | - Judith Quistián-Galván
- Unidad Médica de Alta Especialidad, Hospital de Especialidades No. 14, Centro Médico Nacional “Adolfo Ruiz Cortines”, Instituto Mexicano del Seguro Social (IMSS) Veracruz Norte, Veracruz, Mexico
| | - Armando Muñoz-Pérez
- Unidad Médica de Alta Especialidad, Hospital de Especialidades No. 14, Centro Médico Nacional “Adolfo Ruiz Cortines”, Instituto Mexicano del Seguro Social (IMSS) Veracruz Norte, Veracruz, Mexico
| | - Víctor Bernal-Dolores
- Unidad Médica de Alta Especialidad, Hospital de Especialidades No. 14, Centro Médico Nacional “Adolfo Ruiz Cortines”, Instituto Mexicano del Seguro Social (IMSS) Veracruz Norte, Veracruz, Mexico
| | - Rosa María del Ángel
- Department of Infectomics and Molecular Pathogenesis, Center for Research and Advanced Studies (CINVESTAV-IPN), Mexico City, Mexico
| | - José Manuel Reyes-Ruiz
- Unidad Médica de Alta Especialidad, Hospital de Especialidades No. 14, Centro Médico Nacional “Adolfo Ruiz Cortines”, Instituto Mexicano del Seguro Social (IMSS) Veracruz Norte, Veracruz, Mexico
| |
Collapse
|
36
|
Hasankhani A, Bahrami A, Sheybani N, Aria B, Hemati B, Fatehi F, Ghaem Maghami Farahani H, Javanmard G, Rezaee M, Kastelic JP, Barkema HW. Differential Co-Expression Network Analysis Reveals Key Hub-High Traffic Genes as Potential Therapeutic Targets for COVID-19 Pandemic. Front Immunol 2021; 12:789317. [PMID: 34975885 PMCID: PMC8714803 DOI: 10.3389/fimmu.2021.789317] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 11/26/2021] [Indexed: 01/08/2023] Open
Abstract
Background The recent emergence of COVID-19, rapid worldwide spread, and incomplete knowledge of molecular mechanisms underlying SARS-CoV-2 infection have limited development of therapeutic strategies. Our objective was to systematically investigate molecular regulatory mechanisms of COVID-19, using a combination of high throughput RNA-sequencing-based transcriptomics and systems biology approaches. Methods RNA-Seq data from peripheral blood mononuclear cells (PBMCs) of healthy persons, mild and severe 17 COVID-19 patients were analyzed to generate a gene expression matrix. Weighted gene co-expression network analysis (WGCNA) was used to identify co-expression modules in healthy samples as a reference set. For differential co-expression network analysis, module preservation and module-trait relationships approaches were used to identify key modules. Then, protein-protein interaction (PPI) networks, based on co-expressed hub genes, were constructed to identify hub genes/TFs with the highest information transfer (hub-high traffic genes) within candidate modules. Results Based on differential co-expression network analysis, connectivity patterns and network density, 72% (15 of 21) of modules identified in healthy samples were altered by SARS-CoV-2 infection. Therefore, SARS-CoV-2 caused systemic perturbations in host biological gene networks. In functional enrichment analysis, among 15 non-preserved modules and two significant highly-correlated modules (identified by MTRs), 9 modules were directly related to the host immune response and COVID-19 immunopathogenesis. Intriguingly, systemic investigation of SARS-CoV-2 infection identified signaling pathways and key genes/proteins associated with COVID-19's main hallmarks, e.g., cytokine storm, respiratory distress syndrome (ARDS), acute lung injury (ALI), lymphopenia, coagulation disorders, thrombosis, and pregnancy complications, as well as comorbidities associated with COVID-19, e.g., asthma, diabetic complications, cardiovascular diseases (CVDs), liver disorders and acute kidney injury (AKI). Topological analysis with betweenness centrality (BC) identified 290 hub-high traffic genes, central in both co-expression and PPI networks. We also identified several transcriptional regulatory factors, including NFKB1, HIF1A, AHR, and TP53, with important immunoregulatory roles in SARS-CoV-2 infection. Moreover, several hub-high traffic genes, including IL6, IL1B, IL10, TNF, SOCS1, SOCS3, ICAM1, PTEN, RHOA, GDI2, SUMO1, CASP1, IRAK3, HSPA5, ADRB2, PRF1, GZMB, OASL, CCL5, HSP90AA1, HSPD1, IFNG, MAPK1, RAB5A, and TNFRSF1A had the highest rates of information transfer in 9 candidate modules and central roles in COVID-19 immunopathogenesis. Conclusion This study provides comprehensive information on molecular mechanisms of SARS-CoV-2-host interactions and identifies several hub-high traffic genes as promising therapeutic targets for the COVID-19 pandemic.
Collapse
Affiliation(s)
- Aliakbar Hasankhani
- Department of Animal Science, College of Agriculture and Natural Resources, University of Tehran, Karaj, Iran
| | - Abolfazl Bahrami
- Department of Animal Science, College of Agriculture and Natural Resources, University of Tehran, Karaj, Iran
- Biomedical Center for Systems Biology Science Munich, Ludwig-Maximilians-University, Munich, Germany
| | - Negin Sheybani
- Department of Animal and Poultry Science, College of Aburaihan, University of Tehran, Tehran, Iran
| | - Behzad Aria
- Department of Physical Education and Sports Science, School of Psychology and Educational Sciences, Yazd University, Yazd, Iran
| | - Behzad Hemati
- Biotechnology Research Center, Karaj Branch, Islamic Azad University, Karaj, Iran
| | - Farhang Fatehi
- Department of Animal Science, College of Agriculture and Natural Resources, University of Tehran, Karaj, Iran
| | | | - Ghazaleh Javanmard
- Department of Animal Science, College of Agriculture and Natural Resources, University of Tehran, Karaj, Iran
| | - Mahsa Rezaee
- Department of Medical Mycology, School of Medical Science, Tarbiat Modares University, Tehran, Iran
| | - John P. Kastelic
- Department of Production Animal Health, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
| | - Herman W. Barkema
- Department of Production Animal Health, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
37
|
Zhang J, Li Q, Cruz Cosme RS, Gerzanich V, Tang Q, Simard JM, Zhao RY. Genome-wide characterization of SARS-CoV-2 cytopathogenic proteins in the search of antiviral targets. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2021:2021.11.23.469747. [PMID: 34845452 PMCID: PMC8629195 DOI: 10.1101/2021.11.23.469747] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Therapeutic inhibition of critical viral functions is important for curtailing coronavirus disease-2019 (COVID-19). We sought to identify antiviral targets through genome-wide characterization of SARS-CoV-2 proteins that are crucial for viral pathogenesis and that cause harmful cytopathic effects. All twenty-nine viral proteins were tested in a fission yeast cell-based system using inducible gene expression. Twelve proteins including eight non-structural proteins (NSP1, NSP3, NSP4, NSP5, NSP6, NSP13, NSP14 and NSP15) and four accessory proteins (ORF3a, ORF6, ORF7a and ORF7b) were identified that altered cellular proliferation and integrity, and induced cell death. Cell death correlated with the activation of cellular oxidative stress. Of the twelve proteins, ORF3a was chosen for further study in mammalian cells. In human pulmonary and kidney epithelial cells, ORF3a induced cellular oxidative stress associated with apoptosis and necrosis, and caused activation of pro-inflammatory response with production of the cytokines TNF-α, IL-6, and IFN-β1, possibly through the activation of NF-κB. To further characterize the mechanism, we tested a natural ORF3a Beta variant, Q57H, and a mutant with deletion of the highly conserved residue, ΔG188. Compared to wild type ORF3a, the ΔG188 variant yielded more robust activation of cellular oxidative stress, cell death, and innate immune response. Since cellular oxidative stress and inflammation contribute to cell death and tissue damage linked to the severity of COVID-19, our findings suggest that ORF3a is a promising, novel therapeutic target against COVID-19.
Collapse
Affiliation(s)
- Jiantao Zhang
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD 21201
| | - Qi Li
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD 21201
| | - Ruth S. Cruz Cosme
- Institute of Global Health, University of Maryland School of Medicine, Baltimore, MD 21201
| | - Volodymyr Gerzanich
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD 21201
| | - Qiyi Tang
- Institute of Global Health, University of Maryland School of Medicine, Baltimore, MD 21201
| | - J. Marc Simard
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD 21201
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD 21201
| | - Richard Y. Zhao
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD 21201
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD 21201
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD 21201
- Institute of Global Health, University of Maryland School of Medicine, Baltimore, MD 21201
| |
Collapse
|
38
|
Taha SI, Shata AK, Baioumy SA, Fouad SH, Anis SG, Mossad IM, Moustafa NM, Abdou DM, Youssef MK. Toll-Like Receptor 4 Polymorphisms (896A/G and 1196C/T) as an Indicator of COVID-19 Severity in a Convenience Sample of Egyptian Patients. J Inflamm Res 2021; 14:6293-6303. [PMID: 34866927 PMCID: PMC8636845 DOI: 10.2147/jir.s343246] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 11/17/2021] [Indexed: 12/13/2022] Open
Abstract
Background The clinical spectrum of COVID-19 is extremely variable. Thus, it is likely that the heterogeneity in the genetic make-up of the host may contribute to disease severity. Toll‐like receptor (TLR)-4 plays a vital role in the innate immune response to SARS-CoV-2 infection. The susceptibility of humans to severe COVID-19 concerning TLR-4 single nucleotide polymorphisms (SNPs) has not been well examined. Objective The goal of this research was to investigate the association between TLR-4 (Asp299Gly and Thr399Ile) SNPs and COVID-19 severity and progression as well as the cytokine storm in Egyptian patients. Methods We genotyped 300 adult COVID-19 Egyptian patients for TLR-4 (Asp299Gly and Thr399Ile) SNPs using PCR-restriction fragment length polymorphism (PCR-RFLP). We also measured interleukin (IL)-6 levels by enzyme-linked immunosorbent assay (ELISA) as an indicator of the cytokine storm. Results The minor 299Gly (G) and 399Ile (T) alleles were associated with a significant (P < 0.001) positive risk of severe COVID-19 (OR = 3.14; 95% CI = 2.02–4.88 and OR = 2.75; 95% CI = 1.66–4.57), their frequency in the severe group were 71.8% (84/150) and 70.7% (58/150), respectively. We detected significant differences between TLR-4 (Asp299Gly, Thr399Ile) genotypes with regard to serum levels of IL-6. Levels of IL-6 increased significantly with the presence of the mutant 299Gly (G) and 399Ile (T) alleles to reach the highest levels in the Gly299Gly (GG) and the Ile399Ile (TT) genotypes (170 pg/mL (145–208.25) and 112 pg/mL (24–284.75), respectively). Conclusion The TLR-4 (Asp299Gly and Thr399Ile) minor alleles 299Gly (G) and 399Ile (T) are associated with COVID-19 severity, mortality, and the cytokine storm.
Collapse
Affiliation(s)
- Sara I Taha
- Department of Clinical Pathology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Aalaa K Shata
- Department of Pulmonary Medicine, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Shereen A Baioumy
- Department of Microbiology and Immunology, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Shaimaa H Fouad
- Department of Internal Medicine/Allergy and Clinical Immunology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Sherif G Anis
- Department of Anesthesia and Intensive Care, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Isis M Mossad
- Department of Community, Environmental and Occupational Medicine, Faculty of Medicine Ain Shams University, Cairo, Egypt
| | - Nouran M Moustafa
- Department of Basic Medical Science, Faculty of Medicine, Dar Al Uloom University, Riyadh, Saudi Arabia.,Department of Microbiology and Immunology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Dina M Abdou
- Department of Internal Medicine/Hematology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Mariam K Youssef
- Department of Clinical Pathology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| |
Collapse
|
39
|
Aslani M, Mortazavi-Jahromi SS, Mirshafiey A. Cytokine storm in the pathophysiology of COVID-19: Possible functional disturbances of miRNAs. Int Immunopharmacol 2021; 101:108172. [PMID: 34601331 PMCID: PMC8452524 DOI: 10.1016/j.intimp.2021.108172] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 09/15/2021] [Accepted: 09/16/2021] [Indexed: 02/07/2023]
Abstract
SARS-CoV-2, as the causative agent of COVID-19, is an enveloped positives-sense single-stranded RNA virus that belongs to the Beta-CoVs sub-family. A sophisticated hyper-inflammatory reaction named cytokine storm is occurred in patients with severe/critical COVID-19, following an imbalance in immune-inflammatory processes and inhibition of antiviral responses by SARS-CoV-2, which leads to pulmonary failure, ARDS, and death. The miRNAs are small non-coding RNAs with an average length of 22 nucleotides which play various roles as one of the main modulators of genes expression and maintenance of immune system homeostasis. Recent evidence has shown that Homo sapiens (hsa)-miRNAs have the potential to work in three pivotal areas including targeting the virus genome, regulating the inflammatory signaling pathways, and reinforcing the production/signaling of IFNs-I. However, it seems that several SARS-CoV-2-induced interfering agents such as viral (v)-miRNAs, cytokine content, competing endogenous RNAs (ceRNAs), etc. preclude efficient function of hsa-miRNAs in severe/critical COVID-19. This subsequently leads to increased virus replication, intense inflammatory processes, and secondary complications development. In this review article, we provide an overview of hsa-miRNAs roles in viral genome targeting, inflammatory pathways modulation, and IFNs responses amplification in severe/critical COVID-19 accompanied by probable interventional factors and their function. Identification and monitoring of these interventional elements can help us in designing the miRNAs-based therapy for the reduction of complications/mortality rate in patients with severe/critical forms of the disease.
Collapse
Affiliation(s)
- Mona Aslani
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Abbas Mirshafiey
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
40
|
Alam MS, Czajkowsky DM. SARS-CoV-2 infection and oxidative stress: Pathophysiological insight into thrombosis and therapeutic opportunities. Cytokine Growth Factor Rev 2021; 63:44-57. [PMID: 34836751 PMCID: PMC8591899 DOI: 10.1016/j.cytogfr.2021.11.001] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 11/07/2021] [Accepted: 11/08/2021] [Indexed: 01/08/2023]
Abstract
The current coronavirus disease 2019 (COVID-19) pandemic has presented unprecedented challenges to global health. Although the majority of COVID-19 patients exhibit mild-to-no symptoms, many patients develop severe disease and need immediate hospitalization, with most severe infections associated with a dysregulated immune response attributed to a cytokine storm. Epidemiological studies suggest that overall COVID-19 severity and morbidity correlate with underlying comorbidities, including diabetes, obesity, cardiovascular diseases, and immunosuppressive conditions. Patients with such comorbidities exhibit elevated levels of reactive oxygen species (ROS) and oxidative stress caused by an increased accumulation of angiotensin II and by activation of the NADPH oxidase pathway. Moreover, accumulating evidence suggests that oxidative stress coupled with the cytokine storm contribute to COVID-19 pathogenesis and immunopathogenesis by causing endotheliitis and endothelial cell dysfunction and by activating the blood clotting cascade that results in blood coagulation and microvascular thrombosis. In this review, we survey the mechanisms of how severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) induces oxidative stress and the consequences of this stress on patient health. We further shed light on aspects of the host immunity that are crucial to prevent the disease during the early phase of infection. A better understanding of the disease pathophysiology as well as preventive measures aimed at lowering ROS levels may pave the way to mitigate SARS-CoV-2-induced complications and decrease mortality.
Collapse
Affiliation(s)
- Mohammad Shah Alam
- Department of Anatomy and Histology, Bangabandhu Sheikh Mujibur Rahman Agricultural University, Gazipur 1706, Bangladesh.
| | - Daniel M Czajkowsky
- Bio-ID Centre, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| |
Collapse
|
41
|
Tvedt THA, Vo AK, Bruserud Ø, Reikvam H. Cytokine Release Syndrome in the Immunotherapy of Hematological Malignancies: The Biology behind and Possible Clinical Consequences. J Clin Med 2021; 10:jcm10215190. [PMID: 34768710 PMCID: PMC8585070 DOI: 10.3390/jcm10215190] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 11/01/2021] [Accepted: 11/03/2021] [Indexed: 02/07/2023] Open
Abstract
Cytokine release syndrome (CRS) is an acute systemic inflammatory syndrome characterized by fever and multiple organ dysfunction associated with (i) chimeric antigen receptor (CAR)-T cell therapy, (ii) therapeutic antibodies, and (iii) haploidentical allogeneic stem cell transplantation (haplo-allo-HSCT). Severe CRS can be life-threatening in some cases and requires prompt management of those toxicities and is still a great challenge for physicians. The pathophysiology of CRS is still not fully understood, which also applies to the identifications of predictive biomarkers that can forecast these features in advance. However, a broad range of cytokines are involved in the dynamics of CRS. Treatment approaches include both broad spectrum of immunosuppressant, such as corticosteroids, as well as more specific inhibition of cytokine release. In the present manuscript we will try to review an update regarding pathophysiology, etiology, diagnostics, and therapeutic options for this serious complication.
Collapse
Affiliation(s)
| | - Anh Khoi Vo
- Department of Clinical Science, University of Bergen, 5020 Bergen, Norway; (A.K.V.); (Ø.B.)
| | - Øystein Bruserud
- Department of Clinical Science, University of Bergen, 5020 Bergen, Norway; (A.K.V.); (Ø.B.)
- Clinic for Medicine, Haukeland University Hospital, 5020 Bergen, Norway
| | - Håkon Reikvam
- Department of Clinical Science, University of Bergen, 5020 Bergen, Norway; (A.K.V.); (Ø.B.)
- Clinic for Medicine, Haukeland University Hospital, 5020 Bergen, Norway
- Correspondence:
| |
Collapse
|
42
|
Soni S, Namdeo Pudake R, Jain U, Chauhan N. A systematic review on SARS-CoV-2-associated fungal coinfections. J Med Virol 2021; 94:99-109. [PMID: 34570905 PMCID: PMC8661750 DOI: 10.1002/jmv.27358] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Revised: 08/16/2021] [Accepted: 09/23/2021] [Indexed: 01/08/2023]
Abstract
A severe pandemic of Coronavirus Disease (COVID‐19) has been sweeping the globe since 2019, and this time, it did not stop, with frequent mutations transforming into virulent strains, for instance, B.1.1.7, B.1.351, and B.1.427. In recent months, a fungal infection, mucormycosis has emerged with more fatal responses and significantly increased mortality rate. To measure the severity and potential alternative approaches against black fungus coinfection in COVID‐19 patients, PubMed, Google Scholar, World Health Organization (WHO) newsletters, and other online resources, based on the cases reported and retrospective observational analysis were searched from the years 2015–2021. The studies reporting mucormycosis with Severe Acute Respiratory Syndrome Coronavirus 2 (SARS‐CoV‐2) coinfection and/or demonstrating potential risk factors, such as a history of diabetes mellitus or suppressed immune system were included, and reports published in non‐English language were excluded. More than 20 case reports and observational studies on black fungus coinfection in COVID‐19 patients were eligible for inclusion. The results indicated that diabetes mellitus, hyperglycemic, and immunocompromised COVID‐19 patients with mucormycosis were at a higher risk. We found that it was prudent to assess the potential risk factors and severity of invasive mycosis via standardized diagnostic and clinical settings. Large‐scale studies need to be conducted to identify early biomarkers and optimization of diagnostic methods has to be established per population and geographical variation. This will not only help clinicians around the world to detect the coinfection in time but also will prepare them for future outbreaks of other potential pandemics. Observational studies and case reports of Post‐COVID black fungus co‐infection highlighted in review. Along with risk factors, radiological interventions of black fungus co‐infection in COVID‐19 patients and challenges for accurate diagnosis were elaborated. The review discusses interconnection between fungus and SARS‐CoV‐2 co‐infection of mechanism to identify potential biomarkers. Our conclusion will contribute to motivate researchers to design follow‐up plans for black fungus infection in COVID‐19.
Collapse
Affiliation(s)
- Shringika Soni
- Amity Institute of Nanotechnology (AINT), Amity University Uttar Pradesh (AUUP), Noida, India
| | - Ramesh Namdeo Pudake
- Amity Institute of Nanotechnology (AINT), Amity University Uttar Pradesh (AUUP), Noida, India
| | - Utkarsh Jain
- Amity Institute of Nanotechnology (AINT), Amity University Uttar Pradesh (AUUP), Noida, India
| | - Nidhi Chauhan
- Amity Institute of Nanotechnology (AINT), Amity University Uttar Pradesh (AUUP), Noida, India
| |
Collapse
|
43
|
Vázquez-Jiménez A, Avila-Ponce De León UE, Matadamas-Guzman M, Muciño-Olmos EA, Martínez-López YE, Escobedo-Tapia T, Resendis-Antonio O. On Deep Landscape Exploration of COVID-19 Patients Cells and Severity Markers. Front Immunol 2021; 12:705646. [PMID: 34603282 PMCID: PMC8481922 DOI: 10.3389/fimmu.2021.705646] [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: 05/05/2021] [Accepted: 08/23/2021] [Indexed: 12/12/2022] Open
Abstract
COVID-19 is a disease with a spectrum of clinical responses ranging from moderate to critical. To study and control its effects, a large number of researchers are focused on two substantial aims. On the one hand, the discovery of diverse biomarkers to classify and potentially anticipate the disease severity of patients. These biomarkers could serve as a medical criterion to prioritize attention to those patients with higher prone to severe responses. On the other hand, understanding how the immune system orchestrates its responses in this spectrum of disease severities is a fundamental issue required to design new and optimized therapeutic strategies. In this work, using single-cell RNAseq of bronchoalveolar lavage fluid of nine patients with COVID-19 and three healthy controls, we contribute to both aspects. First, we presented computational supervised machine-learning models with high accuracy in classifying the disease severity (moderate and severe) in patients with COVID-19 starting from single-cell data from bronchoalveolar lavage fluid. Second, we identified regulatory mechanisms from the heterogeneous cell populations in the lungs microenvironment that correlated with different clinical responses. Given the results, patients with moderate COVID-19 symptoms showed an activation/inactivation profile for their analyzed cells leading to a sequential and innocuous immune response. In comparison, severe patients might be promoting cytotoxic and pro-inflammatory responses in a systemic fashion involving epithelial and immune cells without the possibility to develop viral clearance and immune memory. Consequently, we present an in-depth landscape analysis of how transcriptional factors and pathways from these heterogeneous populations can regulate their expression to promote or restrain an effective immune response directly linked to the patients prognosis.
Collapse
Affiliation(s)
- Aarón Vázquez-Jiménez
- Human Systems Biology Laboratory, Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City, Mexico
| | - Ugo Enrique Avila-Ponce De León
- Human Systems Biology Laboratory, Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City, Mexico
- Programa de Doctorado en Ciencias Biológicas, UNAM, Mexico City, Mexico
| | - Meztli Matadamas-Guzman
- Human Systems Biology Laboratory, Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City, Mexico
- Programa de Doctorado en Ciencias Biomédicas, UNAM, Mexico City, Mexico
| | - Erick Andrés Muciño-Olmos
- Human Systems Biology Laboratory, Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City, Mexico
- Programa de Doctorado en Ciencias Biomédicas, UNAM, Mexico City, Mexico
| | - Yoscelina E. Martínez-López
- Human Systems Biology Laboratory, Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City, Mexico
- Programa de Doctorado en Ciencias Médicas y de la Salud, UNAM, Mexico City, Mexico
| | - Thelma Escobedo-Tapia
- Human Systems Biology Laboratory, Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City, Mexico
- Programa de Maestría y Doctorado en Ciencias Bioquímicas, UNAM, Mexico City, Mexico
| | - Osbaldo Resendis-Antonio
- Human Systems Biology Laboratory, Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City, Mexico
- Coordinación de la Investigación Científica - Red de Apoyo a la Investigación, UNAM, Mexico City, Mexico
| |
Collapse
|
44
|
Ultramicronized Palmitoylethanolamide Inhibits NLRP3 Inflammasome Expression and Pro-Inflammatory Response Activated by SARS-CoV-2 Spike Protein in Cultured Murine Alveolar Macrophages. Metabolites 2021; 11:metabo11090592. [PMID: 34564408 PMCID: PMC8472716 DOI: 10.3390/metabo11090592] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 08/29/2021] [Accepted: 08/30/2021] [Indexed: 02/06/2023] Open
Abstract
Despite its possible therapeutic potential against COVID-19, the exact mechanism(s) by which palmitoylethanolamide (PEA) exerts its beneficial activity is still unclear. PEA has demonstrated analgesic, anti-allergic, and anti-inflammatory activities. Most of the anti-inflammatory properties of PEA arise from its ability to antagonize nuclear factor-κB (NF-κB) signalling pathway via the selective activation of the PPARα receptors. Acting at this site, PEA can downstream several genes involved in the inflammatory response, including cytokines (TNF-α, Il-1β) and other signal mediators, such as inducible nitric oxide synthase (iNOS) and COX2. To shed light on this, we tested the anti-inflammatory and immunomodulatory activity of ultramicronized(um)-PEA, both alone and in the presence of specific peroxisome proliferator-activated receptor alpha (PPAR-α) antagonist MK886, in primary cultures of murine alveolar macrophages exposed to SARS-CoV-2 spike glycoprotein (SP). SP challenge caused a significant concentration-dependent increase in proinflammatory markers (TLR4, p-p38 MAPK, NF-κB) paralleled to a marked upregulation of inflammasome-dependent inflammatory pathways (NLRP3, Caspase-1) with IL-6, IL-1β, TNF-α over-release, compared to vehicle group. We also observed a significant concentration-dependent increase in angiotensin-converting enzyme-2 (ACE-2) following SP challenge. um-PEA concentration-dependently reduced all the analyzed proinflammatory markers fostering a parallel downregulation of ACE-2. Our data show for the first time that um-PEA, via PPAR-α, markedly inhibits the SP induced NLRP3 signalling pathway outlining a novel mechanism of action of this lipid against COVID-19.
Collapse
|
45
|
Jakobsson J, Cotgreave I, Furberg M, Arnberg N, Svensson M. Potential Physiological and Cellular Mechanisms of Exercise That Decrease the Risk of Severe Complications and Mortality Following SARS-CoV-2 Infection. Sports (Basel) 2021; 9:121. [PMID: 34564326 PMCID: PMC8472997 DOI: 10.3390/sports9090121] [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] [Received: 07/23/2021] [Revised: 08/19/2021] [Accepted: 08/23/2021] [Indexed: 01/08/2023] Open
Abstract
The coronavirus disease 2019 (COVID-19) pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has unmasked mankind's vulnerability to biological threats. Although higher age is a major risk factor for disease severity in COVID-19, several predisposing risk factors for mortality are related to low cardiorespiratory and metabolic fitness, including obesity, cardiovascular disease, diabetes, and hypertension. Reaching physical activity (PA) guideline goals contribute to protect against numerous immune and inflammatory disorders, in addition to multi-morbidities and mortality. Elevated levels of cardiorespiratory fitness, being non-obese, and regular PA improves immunological function, mitigating sustained low-grade systemic inflammation and age-related deterioration of the immune system, or immunosenescence. Regular PA and being non-obese also improve the antibody response to vaccination. In this review, we highlight potential physiological, cellular, and molecular mechanisms that are affected by regular PA, increase the host antiviral defense, and may determine the course and outcome of COVID-19. Not only are the immune system and regular PA in relation to COVID-19 discussed, but also the cardiovascular, respiratory, renal, and hormonal systems, as well as skeletal muscle, epigenetics, and mitochondrial function.
Collapse
Affiliation(s)
- Johan Jakobsson
- Section of Sports Medicine, Department of Community Medicine and Rehabilitation, Umeå University, 901 87 Umeå, Sweden;
| | - Ian Cotgreave
- Division of Biomaterials and Health, Department of Pharmaceutical and Chemical Safety, Research Institutes of Sweden, 151 36 Södertälje, Sweden;
| | - Maria Furberg
- Department of Clinical Microbiology, Umeå University, 901 87 Umeå, Sweden; (M.F.); (N.A.)
| | - Niklas Arnberg
- Department of Clinical Microbiology, Umeå University, 901 87 Umeå, Sweden; (M.F.); (N.A.)
| | - Michael Svensson
- Section of Sports Medicine, Department of Community Medicine and Rehabilitation, Umeå University, 901 87 Umeå, Sweden;
| |
Collapse
|
46
|
Pesce M, Agostoni P, Bøtker HE, Brundel B, Davidson SM, Caterina RD, Ferdinandy P, Girao H, Gyöngyösi M, Hulot JS, Lecour S, Perrino C, Schulz R, Sluijter JP, Steffens S, Tancevski I, Gollmann-Tepeköylü C, Tschöpe C, Linthout SV, Madonna R. COVID-19-related cardiac complications from clinical evidences to basic mechanisms: opinion paper of the ESC Working Group on Cellular Biology of the Heart. Cardiovasc Res 2021; 117:2148-2160. [PMID: 34117887 DOI: 10.1093/cvr/cvab201] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 06/09/2021] [Indexed: 12/15/2022] Open
Abstract
The pandemic of coronavirus disease (COVID)-19 is a global threat, causing high mortality, especially in the elderly. The main symptoms and the primary cause of death are related to interstitial pneumonia. Viral entry also into myocardial cells mainly via the angiotensin converting enzyme type 2 (ACE2) receptor and excessive production of pro-inflammatory cytokines, however, also make the heart susceptible to injury. In addition to the immediate damage caused by the acute inflammatory response, the heart may also suffer from long-term consequences of COVID-19, potentially causing a post-pandemic increase in cardiac complications. Although the main cause of cardiac damage in COVID-19 remains coagulopathy with micro- (and to a lesser extent macro-) vascular occlusion, open questions remain about other possible modalities of cardiac dysfunction, such as direct infection of myocardial cells, effects of cytokines storm, and mechanisms related to enhanced coagulopathy. In this opinion paper, we focus on these lesser appreciated possibilities and propose experimental approaches that could provide a more comprehensive understanding of the cellular and molecular bases of cardiac injury in COVID-19 patients. We first discuss approaches to characterize cardiac damage caused by possible direct viral infection of cardiac cells, followed by formulating hypotheses on how to reproduce and investigate the hyperinflammatory and pro-thrombotic conditions observed in the heart of COVID-19 patients using experimental in vitro systems. Finally, we elaborate on strategies to discover novel pathology biomarkers using omics platforms.
Collapse
Affiliation(s)
| | - Piergiuseppe Agostoni
- Centro Cardiologico Monzino, IRCCS, Milan, Italy
- Dipartimento di Scienze Cliniche e di Comunità, University of Milan, Milan, Italy
| | - Hans-Erik Bøtker
- Department of Cardiology, Aarhus University Hospital, Aarhus N, Denmark
| | - Bianca Brundel
- Department of Physiology, Amsterdam University Medical Centers (UMC), Vrije Universiteit, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands
| | - Sean M Davidson
- The Hatter Cardiovascular Institute, University College London, London, UK
| | | | - Peter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- Pharmahungary Group, Szeged, Hungary
| | - Henrique Girao
- Center for Innovative Biomedicine and Biotechnology (CIBB), Clinical Academic Centre of Coimbra (CACC), Faculty of Medicine, Univ Coimbra, Institute for Clinical and Biomedical Research (iCBR), Coimbra, Portugal
| | - Mariann Gyöngyösi
- Department of Cardiology, Medical University of Vienna, Vienna, Austria
| | - Jean-Sebastien Hulot
- Université de Paris, PARCC, INSERM, Paris, France
- CIC1418 and DMU CARTE, AP-HP, Hôpital Européen Georges-Pompidou, Paris, France
| | - Sandrine Lecour
- Faculty of Health Sciences, Hatter Institute for Cardiovascular Research in Africa and Cape Heart Institute, University of Cape Town, Cape Town, South Africa
| | - Cinzia Perrino
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy
| | - Rainer Schulz
- Institute of Physiology, Justus-Liebig University Giessen, Giessen, Germany
| | - Joost Pg Sluijter
- Laboratory for Experimental Cardiology, Department of Cardiology, Utrecht Regenerative Medicine Center, Circulatory Health Laboratory, University Utrecht, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Sabine Steffens
- Institute for Cardiovascular Prevention, German Centre for Cardiovascular Research (DZHK), Ludwig-Maximilians-University (LMU) Munich, Partner Site Munich Heart Alliance, Munich, Germany
| | - Ivan Tancevski
- Department of Internal Medicine II, Medical University of Innsbruck, Innsbruck, Austria
| | | | - Carsten Tschöpe
- Department of Cardiology, Charité, Campus Virchow Klinikum, Berlin, Germany
- BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health at Charité-Universitätmedizin Berlin, Berlin, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany
| | - Sophie van Linthout
- Department of Cardiology, Charité, Campus Virchow Klinikum, Berlin, Germany
- BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health at Charité-Universitätmedizin Berlin, Berlin, Germany
| | - Rosalinda Madonna
- Cardiology Chair, University of Pisa, Pisa University Hospital, Pisa, Italy
- Department of Internal Medicine, University of Texas Medical School in Houston, Houston, TX, USA
| |
Collapse
|
47
|
Uberti F, Ruga S, Farghali M, Galla R, Molinari C. A Combination of α-Lipoic Acid (ALA) and Palmitoylethanolamide (PEA) Blocks Endotoxin-Induced Oxidative Stress and Cytokine Storm: A Possible Intervention for COVID-19. J Diet Suppl 2021; 20:133-155. [PMID: 34405764 DOI: 10.1080/19390211.2021.1966152] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The global scientific community is striving to understand the pathophysiological mechanisms and develop effective therapeutic strategies for COVID-19. Despite overwhelming data, there is limited knowledge about the molecular mechanisms involved in the prominent cytokine storm syndrome and multiple organ failure and fatality in COVID-19 cases. The aim of this work is to investigate the possible role of of α-lipoic acid (ALA) and palmitoylethanolamide (PEA), in countering the mechanisms in overproduction of reactive oxygen species (ROS), and inflammatory cytokines. An in vitro model of lipopolysaccharide (LPS)-stimulated human epithelial lung cells that mimics the pathogen-associated molecular pattern and reproduces the cell signaling pathways in cytokine storm syndrome has been used. In this model of acute lung injury, the combination effects of ALAPEA, administered before and after LPS injury, were investigated. Our data demonstrated that a combination of 50 µM ALA + 5 µM PEA can reduce ROS and nitric oxide (NO) levels modulating the major cytokines involved on COVID-19 infection when administered either before or after LPS-induced damage. The best outcome was observed when administered after LPS, thus reinforcing the hypothesis that ALA combined with PEA to modulate the key point of cytokine storm syndrome. This work supports for the first time that the combination of ALA with PEA may represent a novel intervention strategy to counteract inflammatory damage related to COVID-19 by restoring the cascade activation of the immune response and acting as a powerful antioxidant.
Collapse
Affiliation(s)
- Francesca Uberti
- Laboratory Physiology, Department of Translational Medicine, UPO, Novara, Italy
| | - Sara Ruga
- Laboratory Physiology, Department of Translational Medicine, UPO, Novara, Italy
| | - Mahitab Farghali
- Laboratory Physiology, Department of Translational Medicine, UPO, Novara, Italy
| | - Rebecca Galla
- Laboratory Physiology, Department of Translational Medicine, UPO, Novara, Italy
| | - Claudio Molinari
- Laboratory Physiology, Department of Translational Medicine, UPO, Novara, Italy
| |
Collapse
|
48
|
de Oliveira AA, Nunes KP. Crosstalk of TLR4, vascular NADPH oxidase, and COVID-19 in diabetes: What are the potential implications? Vascul Pharmacol 2021; 139:106879. [PMID: 34051372 PMCID: PMC8152239 DOI: 10.1016/j.vph.2021.106879] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 05/10/2021] [Accepted: 05/24/2021] [Indexed: 02/08/2023]
Abstract
Toll-like receptor 4 (TLR4) contributes to the pathophysiology of diabetes. This happens, at least in part, because TLR4 modulates the enzyme NADPH oxidase, a primary source of ROS in vascular structures. Increased oxidative stress disrupts key vascular signaling mechanisms and drives the progression of diabetes, elevating the likelihood of cardiovascular diseases. Recently, it has been shown that patients with diabetes are also at a higher risk of developing severe coronavirus disease 2019 (COVID-19). Given the importance of the interaction between TLR4 and NADPH oxidase to the disrupted diabetic vascular system, we put forward the hypothesis that TLR4-mediated NADPH oxidase-derived ROS might be a critical mechanism to help explain why this disparity appears in diabetic patients, but unfortunately, conclusive experimental evidence still lacks in the literature. Herein, we focus on discussing the pathological implications of this signaling communication in the diabetic vasculature and exploring this crosstalk in the context of diabetes-associated severe COVID-19.
Collapse
Affiliation(s)
- Amanda Almeida de Oliveira
- Laboratory of Vascular Biology, Department of Biomedical and Chemical Engineering and Sciences, Florida Institute of Technology, Melbourne, United States
| | - Kenia Pedrosa Nunes
- Laboratory of Vascular Biology, Department of Biomedical and Chemical Engineering and Sciences, Florida Institute of Technology, Melbourne, United States.
| |
Collapse
|
49
|
Shabani Z. Demyelination as a result of an immune response in patients with COVID-19. Acta Neurol Belg 2021; 121:859-866. [PMID: 33934300 PMCID: PMC8088756 DOI: 10.1007/s13760-021-01691-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 04/23/2021] [Indexed: 01/08/2023]
Abstract
The coronavirus disease of 2019 (COVID-19) is caused by the severe acute respiratory syndrome coronavirus-2 (SARS CoV-2), that already appeared as a global pandemic. Presentation of the disease often includes upper respiratory symptoms like dry cough, dyspnea, chest pain, and rhinorrhea that can develop to respiratory failure, needing intubation. Furthermore, the occurrence of acute and subacute neurological manifestations such as stroke, encephalitis, headache, and seizures are frequently stated in patients with COVID-19. One of the reported neurological complications of severe COVID-19 is the demolition of the myelin sheath. Indeed, the complex immunological dysfunction provides a substrate for the development of demyelination. Nevertheless, few published reports in the literature describe demyelination in subjects with COVID-19. In this short narrative review, we discuss probable pathological mechanisms that may trigger demyelination in patients with SARS-CoV-2 infection and summarize the clinical evidence, confirming SARS-CoV-2 condition as a risk factor for the destruction of myelin.
Collapse
Affiliation(s)
- Zahra Shabani
- Department of Neurosciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
50
|
Conte C. Possible Link between SARS-CoV-2 Infection and Parkinson's Disease: The Role of Toll-Like Receptor 4. Int J Mol Sci 2021; 22:7135. [PMID: 34281186 PMCID: PMC8269350 DOI: 10.3390/ijms22137135] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 06/19/2021] [Accepted: 06/29/2021] [Indexed: 12/13/2022] Open
Abstract
Parkinson's disease (PD) is the most common neurodegenerative motor disorder characterized by selective degeneration of dopaminergic neurons in the substantia nigra pars compacta (SNpc) of the midbrain, depletion of dopamine (DA), and impaired nigrostriatal pathway. The pathological hallmark of PD includes the aggregation and accumulation α-synuclein (α-SYN). Although the precise mechanisms underlying the pathogenesis of PD are still unknown, the activation of toll-like receptors (TLRs), mainly TLR4 and subsequent neuroinflammatory immune response, seem to play a significant role. Mounting evidence suggests that viral infection can concur with the precipitation of PD or parkinsonism. The recently identified coronavirus named severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the causative agent of ongoing pandemic coronavirus disease 2019 (COVID-19), responsible for 160 million cases that led to the death of more than three million individuals worldwide. Studies have reported that many patients with COVID-19 display several neurological manifestations, including acute cerebrovascular diseases, conscious disturbance, and typical motor and non-motor symptoms accompanying PD. In this review, the neurotropic potential of SARS-CoV-2 and its possible involvement in the pathogenesis of PD are discussed. Specifically, the involvement of the TLR4 signaling pathway in mediating the virus entry, as well as the massive immune and inflammatory response in COVID-19 patients is explored. The binding of SARS-CoV-2 spike (S) protein to TLR4 and the possible interaction between SARS-CoV-2 and α-SYN as contributing factors to neuronal death are also considered.
Collapse
Affiliation(s)
- Carmela Conte
- Department of Pharmaceutical Sciences, University of Perugia, via Fabretti, 06123 Perugia, Italy
| |
Collapse
|