1
|
Zhang X, Lin J, Zuo D, Chen X, Xu G, Su J, Zhang W. The Tan-Re-Qing Capsule mitigates acute lung injury by suppressing the NLRP3 inflammasome and MAPK/NF-κB signaling pathways. Gene 2025; 933:149001. [PMID: 39401735 DOI: 10.1016/j.gene.2024.149001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 10/09/2024] [Accepted: 10/10/2024] [Indexed: 10/17/2024]
Abstract
OBJECTIVE The Tan-Re-Qing Capsule (TRQC), a traditional Chinese medicine (TCM) preparation, has been historically utilized in treating acute lung injury (ALI) and COVID-19-induced pulmonary diseases. This study aimed to explore the effect and underlying mechanisms of TRQC in lipopolysaccharide (LPS)-induced ALI models. METHODS The changes of acute lung injury and inflammatory response were observed after TRQC treatment of the LPS-induced ALI mouse model. Based on active compounds in TRQC and network pharmacology analysis, potential targeting signals were identified. The effects of TRQC on signaling in LPS-stimulated BMDMs were investigated. Additionally, the defecatory status of mice and the mechanism of Cl- secretion in HBE cells and T84 colonic epithelial cells were examined. RESULTS TRQC exhibited a notable amelioration of inflammatory injuries in ALI mice. Utilizing a systems-pharmacology approach based on active chemical compounds, TRQC was found to regulate inflammation-related pathways, including NF-κB, NOD-like signaling, and MAPK signaling. In vitro experiments demonstrated that TRQC effectively suppressed LPS-induced activation of macrophages and the assembly of the NLRP3 inflammasome induced by LPS and Nigericin. These effects were attributed to the suppression of NF-κB and NOD-like signaling pathways. Furthermore, TRQC blocked MAPK signaling, thereby mitigating the inhibitory effects of LPS and Nigericin on Ca2+-dependent Cl- efflux across colonic epithelial cells. This mechanism generated a cathartic effect, potentially aiding in the removal of harmful substances and pathogenic bacteria. CONCLUSION Our study demonstrates that TRQC significantly mitigates ALI by effectively suppressing the NLRP3 inflammasome and MAPK/NF-κB signaling pathways. These findings suggest that TRQC could serve as a promising therapeutic candidate for inflammatory lung diseases, offering a novel approach to managing conditions like ALI and potentially extending to other inflammatory diseases.
Collapse
Affiliation(s)
- Xing Zhang
- Department of Respiratory Disease, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Division of Pulmonary, Critical Care, Allergy, and Sleep, Department of Medicine, University of California, San Francisco, CA 94143, USA.
| | - Jiacheng Lin
- Central Laboratory, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Dongliang Zuo
- Shanghai Institute for Advanced Immunochemical Studies, Shanghai Tech University, Shanghai 201210, China.
| | - Xuan Chen
- Department of Respiratory Disease, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Guihua Xu
- Department of Respiratory Disease, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Jie Su
- School of Life Sciences and Biotechnology and State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, National Research Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Jiao Tong University, Shanghai 200025, China.
| | - Wei Zhang
- Department of Respiratory Disease, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
2
|
Cahuapaza-Gutierrez NL. Aplastic Anemia in the light of the COVID-19 pandemic: infection, vaccination, and pathophysiologic mechanisms. Ann Hematol 2024:10.1007/s00277-024-06052-9. [PMID: 39441353 DOI: 10.1007/s00277-024-06052-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 10/18/2024] [Indexed: 10/25/2024]
Abstract
Patients infected with SARS-CoV-2 and vaccinated against COVID-19 could develop aplastic anemia (AA). Comprehensive review and analysis were conducted through a selective literature search in PubMed, Scopus, EMBASE, and Web of Science. For this analysis, 26 studies were included, comprising 16 case reports, 7 case series, and 3 observational studies, totaling 53 patients. The causes of acquired or idiopathic AA are diverse; this review presents recent findings, including possible new etiologies such as SARS-CoV-2 infection and COVID-19 vaccines. This possible association is explored, addressing the existing gap, and aiming to improve daily medical practice. This article reviews the relationship between AA and SARS-CoV-2 infection, as well as COVID-19 vaccines, analyzing cases of de novo occurrence and relapses of AA. Although a definitive mechanistic link has not yet been established, possible underlying pathophysiological mechanisms are explored.
Collapse
Affiliation(s)
- Nelson Luis Cahuapaza-Gutierrez
- Facultad de Ciencias de La Salud, Carrera de Medicina Humana, Universidad Científica del Sur, Lima, Perú.
- CHANGE Research Working Group, Universidad Científica del Sur, Lima, Perú.
| |
Collapse
|
3
|
Zhang C, Gerzanich V, Cruz-Cosme R, Zhang J, Tsymbalyuk O, Tosun C, Sallapalli BT, Liu D, Keledjian K, Papadimitriou JC, Drachenberg CB, Nasr M, Zhang Y, Tang Q, Simard JM, Zhao RY. SARS-CoV-2 ORF3a induces COVID-19-associated kidney injury through HMGB1-mediated cytokine production. mBio 2024:e0230824. [PMID: 39345136 DOI: 10.1128/mbio.02308-24] [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: 07/31/2024] [Accepted: 08/12/2024] [Indexed: 10/01/2024] Open
Abstract
The primary challenge posed by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection is COVID-19-related mortality, often exacerbated by additional medical complications, such as COVID-19-associated kidney injuries (CAKIs). Up to half of COVID-19 patients experience kidney complications, with those facing acute respiratory failure and kidney injury having the worst overall prognosis. Despite the significant impact of CAKI on COVID-19-related mortality and its enduring effects in long COVID, the underlying causes and molecular mechanisms of CAKI remain elusive. In this study, we identified a functional relationship between the expression of the SARS-CoV-2 ORF3a protein and inflammation-driven apoptotic death of renal tubular epithelial cells in patients with CAKI. We demonstrate in vitro that ORF3a independently induces renal cell-specific apoptotic cell death, as evidenced by the elevation of kidney injury molecule-1 (KIM-1) and the activation of NF-kB-mediated proinflammatory cytokine (TNFα and IL-6) production. By examining kidney tissues of SARS-CoV-2-infected K18-ACE2 transgenic mice, we observed a similar correlation between ORF3a-induced cytopathic changes and kidney injury. This correlation was further validated through reconstitution of the ORF3a effects via direct adenoviral injection into mouse kidneys. Through medicinal analysis, we identified a natural compound, glycyrrhizin (GL4419), which not only blocks viral replication in renal cells, but also mitigates ORF3a-induced renal cell death by inhibiting activation of a high mobility group box 1 (HMGB1) protein, leading to a reduction of KIM-1. Moreover, ORF3a interacts with HMGB1. Overproduction or downregulation of hmgb1 expression results in correlative changes in renal cellular KIM-1 response and respective cytokine production, implicating a crucial role of HMGB1 in ORF3a-inflicted kidney injuries. Our data suggest a direct functional link between ORF3a and kidney injury, highlighting ORF3a as a unique therapeutic target contributing to CAKI. IMPORTANCE The major challenge of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection during the pandemic is COVID-19-related mortality, which has tragically claimed millions of lives. COVID-19-associated morbidity and mortality are often exacerbated by pre-existing medical conditions, such as chronic kidney diseases (CKDs), or the development of acute kidney injury (AKI) due to COVID-19, collectively known as COVID-19-associated kidney injuries (CAKIs). Patients who experience acute respiratory failure with CAKI have the poorest clinical outcomes, including increased mortality. Despite these alarming clinical findings, there is a critical gap in our understanding of the underlying causes of CAKI. Our study establishes a direct correlation between the expression of the SARS-CoV-2 viral ORF3a protein and kidney injury induced by ORF3a linking to CAKI. This functional relationship was initially observed in our clinical studies of COVID-19 patients with AKI and was further validated through animal and in vitro cellular studies, either by expressing ORF3a alone or in the context of viral infection. By elucidating this functional relationship and its underlying mechanistic pathways, our research deepens the understanding of COVID-19-associated kidney diseases and presents potential therapeutic avenues to address the healthcare challenges faced by individuals with underlying conditions.
Collapse
Affiliation(s)
- Chenyu Zhang
- Department of Pathology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Volodymyr Gerzanich
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Ruth Cruz-Cosme
- Department of Microbiology, Howard University College of Medicine, Washington, DC, USA
| | - Jiantao Zhang
- Department of Pathology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Orest Tsymbalyuk
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Cigdem Tosun
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | | | - Dongxiao Liu
- Department of Microbiology, Howard University College of Medicine, Washington, DC, USA
| | - Kaspar Keledjian
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - John C Papadimitriou
- Department of Pathology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Cinthia B Drachenberg
- Department of Pathology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Mohamed Nasr
- Division of AIDS, NIAID, NIH, Drug Development and Clinical Sciences Branch, Bethesda, Maryland, USA
| | - Yanjin Zhang
- Department of Veterinary Medicine, University of Maryland, College Park, Maryland, USA
| | - Qiyi Tang
- Department of Microbiology, Howard University College of Medicine, Washington, DC, USA
| | - J Marc Simard
- Department of Pathology, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Research and Development Service, VA Maryland Health Care System, Baltimore, Maryland, USA
| | - Richard Y Zhao
- Department of Pathology, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Research and Development Service, VA Maryland Health Care System, Baltimore, Maryland, USA
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Institute of Global Health, University of Maryland School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
4
|
Jaiswal A, Shrivastav S, Kushwaha HR, Chaturvedi R, Singh RP. Oncogenic potential of SARS-CoV-2-targeting hallmarks of cancer pathways. Cell Commun Signal 2024; 22:447. [PMID: 39327555 PMCID: PMC11426004 DOI: 10.1186/s12964-024-01818-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 09/04/2024] [Indexed: 09/28/2024] Open
Abstract
The 2019 outbreak of SARS-CoV-2 has caused a major worldwide health crisis with high rates of morbidity and death. Interestingly, it has also been linked to cancer, which begs the issue of whether it plays a role in carcinogenesis. Recent studies have revealed various mechanisms by which SARS-CoV-2 can influence oncogenic pathways, potentially promoting cancer development. The virus encodes several proteins that alter key signaling pathways associated with cancer hallmarks. Unlike classical oncogenic viruses, which transform cells through viral oncogenes or by activating host oncogenes, SARS-CoV-2 appears to promote tumorigenesis by inhibiting tumor suppressor genes and pathways while activating survival, proliferation, and inflammation-associated signaling cascades. Bioinformatic analyses and experimental studies have identified numerous interactions between SARS-CoV-2 proteins and cellular components involved in cancer-related processes. This review explores the intricate relationship between SARS-CoV-2 infection and cancer, focusing on the regulation of key hallmarks driving initiation, promotion and progression of cancer by viral proteins. By elucidating the underlying mechanisms driving cellular transformation, the potential of SARS-CoV-2 as an oncovirus is highlighted. Comprehending these interplays is essential to enhance our understanding of COVID-19 and cancer biology and further formulating strategies to alleviate SARS-CoV-2 influence on cancer consequences.
Collapse
Affiliation(s)
- Aishwarya Jaiswal
- Cancer Biology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Sanah Shrivastav
- SRM Institute of Science and Technology, Delhi-NCR Campus, Ghaziabad, Uttar Pradesh, India
| | - Hemant R Kushwaha
- School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
- Special Centre for Systems Medicine, Jawaharlal Nehru University, New Delhi, India
| | - Rupesh Chaturvedi
- School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
- Special Centre for Systems Medicine, Jawaharlal Nehru University, New Delhi, India
| | - Rana P Singh
- Cancer Biology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi, 110067, India.
- Special Centre for Systems Medicine, Jawaharlal Nehru University, New Delhi, India.
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA.
| |
Collapse
|
5
|
Vana F, Szabo Z, Masarik M, Kratochvilova M. The interplay of transition metals in ferroptosis and pyroptosis. Cell Div 2024; 19:24. [PMID: 39097717 PMCID: PMC11297737 DOI: 10.1186/s13008-024-00127-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 07/08/2024] [Indexed: 08/05/2024] Open
Abstract
Cell death is one of the most important mechanisms of maintaining homeostasis in our body. Ferroptosis and pyroptosis are forms of necrosis-like cell death. These cell death modalities play key roles in the pathophysiology of cancer, cardiovascular, neurological diseases, and other pathologies. Transition metals are abundant group of elements in all living organisms. This paper presents a summary of ferroptosis and pyroptosis pathways and their connection to significant transition metals, namely zinc (Zn), copper (Cu), molybdenum (Mo), lead (Pb), cobalt (Co), iron (Fe), cadmium (Cd), nickel (Ni), mercury (Hg), uranium (U), platinum (Pt), and one crucial element, selenium (Se). Authors aim to summarize the up-to-date knowledge of this topic.In this review, there are categorized and highlighted the most common patterns in the alterations of ferroptosis and pyroptosis by transition metals. Special attention is given to zinc since collected data support its dual nature of action in both ferroptosis and pyroptosis. All findings are presented together with a brief description of major biochemical pathways involving mentioned metals and are visualized in attached comprehensive figures.This work concludes that the majority of disruptions in the studied metals' homeostasis impacts cell fate, influencing both death and survival of cells in the complex system of altered pathways. Therefore, this summary opens up the space for further research.
Collapse
Affiliation(s)
- Frantisek Vana
- Department of Pathological Physiology, Faculty of Medicine, Masaryk University, Kamenice 5, Brno, CZ-625 00, Czech Republic
| | - Zoltan Szabo
- Department of Pathological Physiology, Faculty of Medicine, Masaryk University, Kamenice 5, Brno, CZ-625 00, Czech Republic
- Regional Centre for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, Zluty kopec 7, Brno, 656 53, Czech Republic
| | - Michal Masarik
- Department of Physiology, Faculty of Medicine, Masaryk University, Kamenice 5, Brno, CZ-625 00, Czech Republic
- Department of Pathological Physiology, Faculty of Medicine, Masaryk University, Kamenice 5, Brno, CZ-625 00, Czech Republic
- First Faculty of Medicine, BIOCEV, Charles University, Prumyslova 595, Vestec, CZ-252 50, Czech Republic
| | - Monika Kratochvilova
- Department of Physiology, Faculty of Medicine, Masaryk University, Kamenice 5, Brno, CZ-625 00, Czech Republic.
| |
Collapse
|
6
|
Aghajani Mir M. Illuminating the pathogenic role of SARS-CoV-2: Insights into competing endogenous RNAs (ceRNAs) regulatory networks. INFECTION, GENETICS AND EVOLUTION : JOURNAL OF MOLECULAR EPIDEMIOLOGY AND EVOLUTIONARY GENETICS IN INFECTIOUS DISEASES 2024; 122:105613. [PMID: 38844190 DOI: 10.1016/j.meegid.2024.105613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 05/20/2024] [Accepted: 05/31/2024] [Indexed: 06/10/2024]
Abstract
The appearance of SARS-CoV-2 in 2019 triggered a significant economic and health crisis worldwide, with heterogeneous molecular mechanisms that contribute to its development are not yet fully understood. Although substantial progress has been made in elucidating the mechanisms behind SARS-CoV-2 infection and therapy, it continues to rank among the top three global causes of mortality due to infectious illnesses. Non-coding RNAs (ncRNAs), being integral components across nearly all biological processes, demonstrate effective importance in viral pathogenesis. Regarding viral infections, ncRNAs have demonstrated their ability to modulate host reactions, viral replication, and host-pathogen interactions. However, the complex interactions of different types of ncRNAs in the progression of COVID-19 remains understudied. In recent years, a novel mechanism of post-transcriptional gene regulation known as "competing endogenous RNA (ceRNA)" has been proposed. Long non-coding RNAs (lncRNAs), circular RNAs (circRNAs), and viral ncRNAs function as ceRNAs, influencing the expression of associated genes by sequestering shared microRNAs. Recent research on SARS-CoV-2 has revealed that disruptions in specific ceRNA regulatory networks (ceRNETs) contribute to the abnormal expression of key infection-related genes and the establishment of distinctive infection characteristics. These findings present new opportunities to delve deeper into the underlying mechanisms of SARS-CoV-2 pathogenesis, offering potential biomarkers and therapeutic targets. This progress paves the way for a more comprehensive understanding of ceRNETs, shedding light on the intricate mechanisms involved. Further exploration of these mechanisms holds promise for enhancing our ability to prevent viral infections and develop effective antiviral treatments.
Collapse
Affiliation(s)
- Mahsa Aghajani Mir
- Deputy of Research and Technology, Babol University of Medical Sciences, Babol, Iran.
| |
Collapse
|
7
|
Zhi J, Zhao KX, Liu JH, Yang D, Deng XM, Xu J, Zhang H. The therapeutic potential of gelsolin in attenuating cytokine storm, ARDS, and ALI in severe COVID-19. Front Pharmacol 2024; 15:1447403. [PMID: 39130641 PMCID: PMC11310015 DOI: 10.3389/fphar.2024.1447403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 07/01/2024] [Indexed: 08/13/2024] Open
Abstract
Severe COVID-19 cases often progress to life-threatening conditions such as acute respiratory distress syndrome (ARDS), sepsis, and multiple organ dysfunction syndrome (MODS). Gelsolin (GSN), an actin-binding protein with anti-inflammatory and immunomodulatory properties, is a promising therapeutic target for severe COVID-19. Plasma GSN levels are significantly decreased in critical illnesses, including COVID-19, correlating with dysregulated immune responses and poor outcomes. GSN supplementation may mitigate acute lung injury, ARDS, and sepsis, which share pathophysiological features with severe COVID-19, by scavenging actin, modulating cytokine production, enhancing macrophage phagocytosis, and stabilizing the alveolar-capillary barrier. Preliminary data indicate that recombinant human plasma GSN improves oxygenation and lung function in severe COVID-19 patients with ARDS. Although further research is needed to optimize GSN therapy, current evidence supports its potential to mitigate severe consequences of COVID-19 and improve patient outcomes. This review provides a comprehensive analysis of the biological characteristics, mechanisms, and therapeutic value of GSN in severe COVID-19.
Collapse
Affiliation(s)
| | | | | | - Dong Yang
- Department of Anesthesiology at the Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | | | | | | |
Collapse
|
8
|
Zheng HY, Song TZ, Zheng YT. Immunobiology of COVID-19: Mechanistic and therapeutic insights from animal models. Zool Res 2024; 45:747-766. [PMID: 38894519 PMCID: PMC11298684 DOI: 10.24272/j.issn.2095-8137.2024.062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 04/22/2024] [Indexed: 06/21/2024] Open
Abstract
The distribution of the immune system throughout the body complicates in vitro assessments of coronavirus disease 2019 (COVID-19) immunobiology, often resulting in a lack of reproducibility when extrapolated to the whole organism. Consequently, developing animal models is imperative for a comprehensive understanding of the pathology and immunology of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. This review summarizes current progress related to COVID-19 animal models, including non-human primates (NHPs), mice, and hamsters, with a focus on their roles in exploring the mechanisms of immunopathology, immune protection, and long-term effects of SARS-CoV-2 infection, as well as their application in immunoprevention and immunotherapy of SARS-CoV-2 infection. Differences among these animal models and their specific applications are also highlighted, as no single model can fully encapsulate all aspects of COVID-19. To effectively address the challenges posed by COVID-19, it is essential to select appropriate animal models that can accurately replicate both fatal and non-fatal infections with varying courses and severities. Optimizing animal model libraries and associated research tools is key to resolving the global COVID-19 pandemic, serving as a robust resource for future emerging infectious diseases.
Collapse
Affiliation(s)
- Hong-Yi Zheng
- State Key Laboratory of Genetic Evolution & Animal Models, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Center for Biosafety Mega-Science, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China
| | - Tian-Zhang Song
- State Key Laboratory of Genetic Evolution & Animal Models, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Center for Biosafety Mega-Science, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China
| | - Yong-Tang Zheng
- State Key Laboratory of Genetic Evolution & Animal Models, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Center for Biosafety Mega-Science, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China
- National Resource Center for Non-Human Primates, National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650107, China. E-mail:
| |
Collapse
|
9
|
Kopera K, Gromowski T, Wydmański W, Skonieczna-Żydecka K, Muszyńska A, Zielińska K, Wierzbicka-Woś A, Kaczmarczyk M, Kadaj-Lipka R, Cembrowska-Lech D, Januszkiewicz K, Kotfis K, Witkiewicz W, Nalewajska M, Feret W, Marlicz W, Łoniewski I, Łabaj PP, Rydzewska G, Kosciolek T. Gut microbiome dynamics and predictive value in hospitalized COVID-19 patients: a comparative analysis of shallow and deep shotgun sequencing. Front Microbiol 2024; 15:1342749. [PMID: 38962119 PMCID: PMC11219902 DOI: 10.3389/fmicb.2024.1342749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 05/20/2024] [Indexed: 07/05/2024] Open
Abstract
The COVID-19 pandemic caused by SARS-CoV-2 has led to a wide range of clinical presentations, with respiratory symptoms being common. However, emerging evidence suggests that the gastrointestinal (GI) tract is also affected, with angiotensin-converting enzyme 2, a key receptor for SARS-CoV-2, abundantly expressed in the ileum and colon. The virus has been detected in GI tissues and fecal samples, even in cases with negative results of the reverse transcription polymerase chain reaction in the respiratory tract. GI symptoms have been associated with an increased risk of ICU admission and mortality. The gut microbiome, a complex ecosystem of around 40 trillion bacteria, plays a crucial role in immunological and metabolic pathways. Dysbiosis of the gut microbiota, characterized by a loss of beneficial microbes and decreased microbial diversity, has been observed in COVID-19 patients, potentially contributing to disease severity. We conducted a comprehensive gut microbiome study in 204 hospitalized COVID-19 patients using both shallow and deep shotgun sequencing methods. We aimed to track microbiota composition changes induced by hospitalization, link these alterations to clinical procedures (antibiotics administration) and outcomes (ICU referral, survival), and assess the predictive potential of the gut microbiome for COVID-19 prognosis. Shallow shotgun sequencing was evaluated as a cost-effective diagnostic alternative for clinical settings. Our study demonstrated the diverse effects of various combinations of clinical parameters, microbiome profiles, and patient metadata on the precision of outcome prognostication in patients. It indicates that microbiological data possesses greater reliability in forecasting patient outcomes when contrasted with clinical data or metadata. Furthermore, we established that shallow shotgun sequencing presents a viable and cost-effective diagnostic alternative to deep sequencing within clinical environments.
Collapse
Affiliation(s)
- Katarzyna Kopera
- Małopolska Centre of Biotechnology, Jagiellonian University, Kraków, Poland
| | - Tomasz Gromowski
- Małopolska Centre of Biotechnology, Jagiellonian University, Kraków, Poland
- Department of General Biochemistry, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Witold Wydmański
- Małopolska Centre of Biotechnology, Jagiellonian University, Kraków, Poland
- Faculty of Mathematics and Computer Science, Jagiellonian University, Kraków, Poland
| | | | - Agata Muszyńska
- Małopolska Centre of Biotechnology, Jagiellonian University, Kraków, Poland
| | - Kinga Zielińska
- Małopolska Centre of Biotechnology, Jagiellonian University, Kraków, Poland
| | | | - Mariusz Kaczmarczyk
- Sanprobi Sp. z o.o. Sp. k., Szczecin, Poland
- Department of Clinical and Molecular Biochemistry, Pomeranian Medical University, Szczecin, Poland
| | - Roland Kadaj-Lipka
- Department of Internal Medicine and Gastroenterology, Central Clinical Hospital of the Ministry of Interior and Administration, Warsaw, Poland
| | - Danuta Cembrowska-Lech
- Department of Biochemical Science, Pomeranian Medical University, Szczecin, Poland
- Sanprobi Sp. z o.o. Sp. k., Szczecin, Poland
| | | | - Katarzyna Kotfis
- Department of Anesthesiology, Intensive Care and Pain Management, Pomeranian Medical University, Szczecin, Poland
| | | | | | - Wiktoria Feret
- Clinical Department of Nephrology, Transplantology and Internal Medicine, Pomeranian Medical University, Szczecin, Poland
| | - Wojciech Marlicz
- Sanprobi Sp. z o.o. Sp. k., Szczecin, Poland
- Department of Gastroenterology, Pomeranian Medical University, Szczecin, Poland
| | - Igor Łoniewski
- Małopolska Centre of Biotechnology, Jagiellonian University, Kraków, Poland
- Department of Biochemical Science, Pomeranian Medical University, Szczecin, Poland
- Sanprobi Sp. z o.o. Sp. k., Szczecin, Poland
| | - Paweł P. Łabaj
- Małopolska Centre of Biotechnology, Jagiellonian University, Kraków, Poland
| | - Grażyna Rydzewska
- Department of Internal Medicine and Gastroenterology, Central Clinical Hospital of the Ministry of Interior and Administration, Warsaw, Poland
| | - Tomasz Kosciolek
- Małopolska Centre of Biotechnology, Jagiellonian University, Kraków, Poland
- Department of Data Science and Engineering, Silesian University of Technology, Gliwice, Poland
| |
Collapse
|
10
|
Zhu L, Qi Z, Zhang H, Wang N. Nucleic Acid Sensor-Mediated PANoptosis in Viral Infection. Viruses 2024; 16:966. [PMID: 38932258 PMCID: PMC11209569 DOI: 10.3390/v16060966] [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: 04/29/2024] [Revised: 06/04/2024] [Accepted: 06/14/2024] [Indexed: 06/28/2024] Open
Abstract
Innate immunity, the first line of host defense against viral infections, recognizes viral components through different pattern-recognition receptors. Nucleic acids derived from viruses are mainly recognized by Toll-like receptors, nucleotide-binding domain leucine-rich repeat-containing receptors, absent in melanoma 2-like receptors, and cytosolic DNA sensors (e.g., Z-DNA-binding protein 1 and cyclic GMP-AMP synthase). Different types of nucleic acid sensors can recognize specific viruses due to their unique structures. PANoptosis is a unique form of inflammatory cell death pathway that is triggered by innate immune sensors and driven by caspases and receptor-interacting serine/threonine kinases through PANoptosome complexes. Nucleic acid sensors (e.g., Z-DNA-binding protein 1 and absent in melanoma 2) not only detect viruses, but also mediate PANoptosis through providing scaffold for the assembly of PANoptosomes. This review summarizes the structures of different nucleic acid sensors, discusses their roles in viral infections by driving PANoptosis, and highlights the crosstalk between different nucleic acid sensors. It also underscores the promising prospect of manipulating nucleic acid sensors as a therapeutic approach for viral infections.
Collapse
Affiliation(s)
- Lili Zhu
- Department of Pathology, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha 410083, China;
| | - Zehong Qi
- Department of Pathophysiology, School of Basic Medical Science, Central South University, Changsha 410083, China;
- Key Laboratory of Sepsis Translational Medicine of Hunan, Central South University, Changsha 410083, China
- National Medicine Functional Experimental Teaching Center, Central South University, Changsha 410083, China
| | - Huali Zhang
- Department of Pathophysiology, School of Basic Medical Science, Central South University, Changsha 410083, China;
- Key Laboratory of Sepsis Translational Medicine of Hunan, Central South University, Changsha 410083, China
- National Medicine Functional Experimental Teaching Center, Central South University, Changsha 410083, China
| | - Nian Wang
- Department of Pathophysiology, School of Basic Medical Science, Central South University, Changsha 410083, China;
- Key Laboratory of Sepsis Translational Medicine of Hunan, Central South University, Changsha 410083, China
- National Medicine Functional Experimental Teaching Center, Central South University, Changsha 410083, China
| |
Collapse
|
11
|
Li S, Song J, Liu J, Zhou S, Zhao G, Li T, Huang L, Li J, Weng C. African swine fever virus infection regulates pyroptosis by cleaving gasdermin A via active caspase-3 and caspase-4. J Biol Chem 2024; 300:107307. [PMID: 38657868 PMCID: PMC11163174 DOI: 10.1016/j.jbc.2024.107307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 04/09/2024] [Accepted: 04/12/2024] [Indexed: 04/26/2024] Open
Abstract
African swine fever, caused by the African swine fever virus (ASFV), is a viral hemorrhagic disease that affects domestic pigs and wild boars. ASFV infection causes extensive tissue damage, and the associated mechanism is poorly understood. Pyroptosis is characterized by the activation of inflammatory caspases and pore formation in the cellular plasma membrane, resulting in the release of inflammatory cytokines and cell damage. How ASFV infection regulates pyroptosis remains unclear. Here, using siRNA assay and overexpression methods, we report that ASFV infection regulated pyroptosis by cleaving the pyroptosis execution protein gasdermin A (GSDMA). ASFV infection activated caspase-3 and caspase-4, which specifically cleaved GSDMA at D75-P76 and D241-V242 to produce GSDMA into five fragments, including GSDMA-N1-75, GSDMA-N1-241, and GSDMA-N76-241 fragments at the N-terminal end of GSDMA. Only GSDMA-N1-241, which was produced in the late stage of ASFV infection, triggered pyroptosis and inhibited ASFV replication. The fragments, GSDMA-N1-75 and GSDMA-N76-241, lose the ability to induce pyroptosis. Overall ASFV infection differentially regulates pyroptosis by GSDMA in the indicated phase, which may be conducive to its own replication. Our findings reveal a novel molecular mechanism for the regulation of pyroptosis.
Collapse
Affiliation(s)
- Shuai Li
- Division of Fundamental Immunology, National African Swine Fever Para-Reference Laboratory, State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, Heilongjiang, China; Heilongjiang Provincial Key Laboratory of Veterinary Immunology, Harbin, Heilongjiang, China
| | - Jie Song
- Division of Fundamental Immunology, National African Swine Fever Para-Reference Laboratory, State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, Heilongjiang, China; Heilongjiang Provincial Key Laboratory of Veterinary Immunology, Harbin, Heilongjiang, China
| | - Jia Liu
- Division of Fundamental Immunology, National African Swine Fever Para-Reference Laboratory, State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, Heilongjiang, China; Heilongjiang Provincial Key Laboratory of Veterinary Immunology, Harbin, Heilongjiang, China
| | - Shijun Zhou
- Division of Fundamental Immunology, National African Swine Fever Para-Reference Laboratory, State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, Heilongjiang, China; Heilongjiang Provincial Key Laboratory of Veterinary Immunology, Harbin, Heilongjiang, China
| | - Gaihong Zhao
- Division of Fundamental Immunology, National African Swine Fever Para-Reference Laboratory, State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, Heilongjiang, China; Heilongjiang Provincial Key Laboratory of Veterinary Immunology, Harbin, Heilongjiang, China
| | - Tingting Li
- Division of Fundamental Immunology, National African Swine Fever Para-Reference Laboratory, State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, Heilongjiang, China; Heilongjiang Provincial Key Laboratory of Veterinary Immunology, Harbin, Heilongjiang, China
| | - Li Huang
- Division of Fundamental Immunology, National African Swine Fever Para-Reference Laboratory, State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, Heilongjiang, China; Heilongjiang Provincial Key Laboratory of Veterinary Immunology, Harbin, Heilongjiang, China
| | - Jiangnan Li
- Division of Fundamental Immunology, National African Swine Fever Para-Reference Laboratory, State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, Heilongjiang, China; Heilongjiang Provincial Key Laboratory of Veterinary Immunology, Harbin, Heilongjiang, China.
| | - Changjiang Weng
- Division of Fundamental Immunology, National African Swine Fever Para-Reference Laboratory, State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, Heilongjiang, China; Heilongjiang Provincial Key Laboratory of Veterinary Immunology, Harbin, Heilongjiang, China.
| |
Collapse
|
12
|
Kozłowski P, Leszczyńska A, Ciepiela O. Long COVID Definition, Symptoms, Risk Factors, Epidemiology and Autoimmunity: A Narrative Review. AMERICAN JOURNAL OF MEDICINE OPEN 2024; 11:100068. [PMID: 39034937 PMCID: PMC11256271 DOI: 10.1016/j.ajmo.2024.100068] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 01/29/2024] [Accepted: 02/01/2024] [Indexed: 07/23/2024]
Abstract
The virus called SARS-CoV-2 emerged in 2019 and quickly spread worldwide, causing COVID-19. It has greatly impacted on everyday life, healthcare systems, and the global economy. In order to save as many lives as possible, precautions such as social distancing, quarantine, and testing policies were implemented, and effective vaccines were developed. A growing amount of data collected worldwide allowed the characterization of this new disease, which turned out to be more complex than other common respiratory tract infections. An increasing number of convalescents presented with a variety of nonspecific symptoms emerging after the acute infection. This possible new global health problem was identified and labelled as long COVID. Since then, a great effort has been made by clinicians and the scientific community to understand the underlying mechanisms and to develop preventive measures and effective treatment. The role of autoimmunity induced by SARS-CoV-2 infection in the development of long COVID is discussed in this review. We aim to deliver a description of several conditions with an autoimmune background observed in COVID-19 convalescents, including Guillain-Barré syndrome, antiphospholipid syndrome and related thrombosis, and Kawasaki disease highlighting a relationship between SARS-CoV-2 infection and the development of autoimmunity. However, further studies are required to determine its true clinical significance.
Collapse
Affiliation(s)
- Paweł Kozłowski
- Central Laboratory, University Clinical Centre of the Medical University of Warsaw, Warsaw, Poland
| | - Aleksandra Leszczyńska
- Central Laboratory, University Clinical Centre of the Medical University of Warsaw, Warsaw, Poland
| | - Olga Ciepiela
- Central Laboratory, University Clinical Centre of the Medical University of Warsaw, Warsaw, Poland
- Department of Laboratory Medicine, Medical University of Warsaw, Warsaw, Poland
| |
Collapse
|
13
|
Joshi G, Das A, Verma G, Guchhait P. Viral infection and host immune response in diabetes. IUBMB Life 2024; 76:242-266. [PMID: 38063433 DOI: 10.1002/iub.2794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 11/05/2023] [Indexed: 04/24/2024]
Abstract
Diabetes, a chronic metabolic disorder disrupting blood sugar regulation, has emerged as a prominent silent pandemic. Uncontrolled diabetes predisposes an individual to develop fatal complications like cardiovascular disorders, kidney damage, and neuropathies and aggravates the severity of treatable infections. Escalating cases of Type 1 and Type 2 diabetes correlate with a global upswing in diabetes-linked mortality. As a growing global concern with limited preventive interventions, diabetes necessitates extensive research to mitigate its healthcare burden and assist ailing patients. An altered immune system exacerbated by chronic hyperinflammation heightens the susceptibility of diabetic individuals to microbial infections, including notable viruses like SARS-CoV-2, dengue, and influenza. Given such a scenario, we scrutinized the literature and compiled molecular pathways and signaling cascades related to immune compartments in diabetics that escalate the severity associated with the above-mentioned viral infections in them as compared to healthy individuals. The pathogenesis of these viral infections that trigger diabetes compromises both innate and adaptive immune functions and pre-existing diabetes also leads to heightened disease severity. Lastly, this review succinctly outlines available treatments for diabetics, which may hold promise as preventive or supportive measures to effectively combat these viral infections in the former.
Collapse
Affiliation(s)
- Garima Joshi
- Regional Centre for Biotechnology, National Capital Region Biotech Science Cluster, Faridabad, India
| | - Anushka Das
- Regional Centre for Biotechnology, National Capital Region Biotech Science Cluster, Faridabad, India
| | - Garima Verma
- Regional Centre for Biotechnology, National Capital Region Biotech Science Cluster, Faridabad, India
| | - Prasenjit Guchhait
- Regional Centre for Biotechnology, National Capital Region Biotech Science Cluster, Faridabad, India
| |
Collapse
|
14
|
Yu T, Hou D, Zhao J, Lu X, Greentree WK, Zhao Q, Yang M, Conde DG, Linder ME, Lin H. NLRP3 Cys126 palmitoylation by ZDHHC7 promotes inflammasome activation. Cell Rep 2024; 43:114070. [PMID: 38583156 PMCID: PMC11130711 DOI: 10.1016/j.celrep.2024.114070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Revised: 02/14/2024] [Accepted: 03/20/2024] [Indexed: 04/09/2024] Open
Abstract
Nucleotide oligomerization domain (NOD)-like receptor protein 3 (NLRP3) inflammasome hyperactivation contributes to many human chronic inflammatory diseases, and understanding how NLRP3 inflammasome is regulated can provide strategies to treat inflammatory diseases. Here, we demonstrate that NLRP3 Cys126 is palmitoylated by zinc finger DHHC-type palmitoyl transferase 7 (ZDHHC7), which is critical for NLRP3-mediated inflammasome activation. Perturbing NLRP3 Cys126 palmitoylation by ZDHHC7 knockout, pharmacological inhibition, or modification site mutation diminishes NLRP3 activation in macrophages. Furthermore, Cys126 palmitoylation is vital for inflammasome activation in vivo. Mechanistically, ZDHHC7-mediated NLRP3 Cys126 palmitoylation promotes resting NLRP3 localizing on the trans-Golgi network (TGN) and activated NLRP3 on the dispersed TGN, which is indispensable for recruitment and oligomerization of the adaptor ASC (apoptosis-associated speck-like protein containing a CARD). The activation of NLRP3 by ZDHHC7 is different from the termination effect mediated by ZDHHC12, highlighting versatile regulatory roles of S-palmitoylation. Our study identifies an important regulatory mechanism of NLRP3 activation that suggests targeting ZDHHC7 or the NLRP3 Cys126 residue as a potential therapeutic strategy to treat NLRP3-related human disorders.
Collapse
Affiliation(s)
- Tao Yu
- Howard Hughes Medical Institute, Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY 14853, USA
| | - Dan Hou
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY 14853, USA
| | - Jiaqi Zhao
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY 14853, USA
| | - Xuan Lu
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY 14853, USA
| | - Wendy K Greentree
- Department of Molecular Medicine, Cornell University, Ithaca, NY 14853, USA
| | - Qian Zhao
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY 14853, USA
| | - Min Yang
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY 14853, USA
| | - Don-Gerard Conde
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY 14853, USA
| | - Maurine E Linder
- Department of Molecular Medicine, Cornell University, Ithaca, NY 14853, USA
| | - Hening Lin
- Howard Hughes Medical Institute, Department of Chemistry and Chemical Biology, Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY 14853, USA.
| |
Collapse
|
15
|
Bonora BM, Marassi M, Fogar P, Zuin J, Cappellari R, Marinello S, Ferrari A, Cattelan A, Avogaro A, Basso D, Fadini GP. Circulating haematopoietic stem cells and long-term outcomes of COVID-19. Eur J Clin Invest 2024; 54:e14150. [PMID: 38088242 DOI: 10.1111/eci.14150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Revised: 11/19/2023] [Accepted: 11/30/2023] [Indexed: 03/13/2024]
Abstract
BACKGROUND AND AIMS An acute depletion of circulating haematopoietic stem/progenitor cells (HSPCs) occurs during COVID-19, especially among patients with a poorer disease course. We herein examined whether HSPCs levels at hospital admission for COVID-19 predict 1-year mortality and the long-COVID syndrome. MATERIALS AND METHODS Patients hospitalized for COVID-19 in an infectious disease ward were consecutively enrolled. Circulating HSPC levels were assessed by flow cytometry as cells expressing CD34 and/or CD133. Follow-up was performed for 12 months after hospitalization through the review of electronic medical records and demographic local registers. RESULTS The study included 100 patients, 36 of whom reported symptoms of long-COVID and 20 died during follow-up. The reduction of 1-SD of HSPCs was associated with a 3- to 5-fold increase in the risk of 1-year mortality. Age, admission hyperglycaemia, C-reactive protein peak, liver enzymes, the need of high-flow oxygen and/or invasive ventilation were predictors of mortality at univariate analysis. Among pre-existing comorbidities, coronary heart disease and chronic kidney disease, but not diabetes, were associated with 1-year mortality. In multivariate analyses, HSPCs remained significantly associated with 1-year mortality independently of confounders. The development of pneumonia an in-hospital treatment with glucocorticoids and convalescent plasma were associated with long-COVID symptoms at follow-up. HSPCs, diabetes and other comorbidities were not predictors of long-COVID. CONCLUSIONS In a cohort of patients hospitalized for COVID-19, lower HSPC levels at the time of admission were independent predictors of 1-year mortality. However, COVID-19 severity, but not HSPC level, was significantly associated with the development of long-COVID symptoms.
Collapse
Affiliation(s)
- Benedetta Maria Bonora
- Department of Medicine, University Hospital of Padova, Padua, Italy
- Veneto Institute of Molecular Medicine, Padua, Italy
| | - Marella Marassi
- Department of Medicine, University Hospital of Padova, Padua, Italy
| | - Paola Fogar
- Department of Medicine, University Hospital of Padova, Padua, Italy
| | - Jenny Zuin
- Department of Medicine, University Hospital of Padova, Padua, Italy
| | | | - Serena Marinello
- Department of Medicine, University Hospital of Padova, Padua, Italy
| | - Anna Ferrari
- Department of Medicine, University Hospital of Padova, Padua, Italy
| | | | - Angelo Avogaro
- Department of Medicine, University Hospital of Padova, Padua, Italy
| | - Daniela Basso
- Department of Medicine, University Hospital of Padova, Padua, Italy
| | - Gian Paolo Fadini
- Department of Medicine, University Hospital of Padova, Padua, Italy
- Veneto Institute of Molecular Medicine, Padua, Italy
| |
Collapse
|
16
|
Adhikary S, Pathak S, Palani V, Acar A, Banerjee A, Al-Dewik NI, Essa MM, Mohammed SGAA, Qoronfleh MW. Current Technologies and Future Perspectives in Immunotherapy towards a Clinical Oncology Approach. Biomedicines 2024; 12:217. [PMID: 38255322 PMCID: PMC10813720 DOI: 10.3390/biomedicines12010217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 01/08/2024] [Accepted: 01/12/2024] [Indexed: 01/24/2024] Open
Abstract
Immunotherapy is now established as a potent therapeutic paradigm engendering antitumor immune response against a wide range of malignancies and other diseases by modulating the immune system either through the stimulation or suppression of immune components such as CD4+ T cells, CD8+ T cells, B cells, monocytes, macrophages, dendritic cells, and natural killer cells. By targeting several immune checkpoint inhibitors or blockers (e.g., PD-1, PD-L1, PD-L2, CTLA-4, LAG3, and TIM-3) expressed on the surface of immune cells, several monoclonal antibodies and polyclonal antibodies have been developed and already translated clinically. In addition, natural killer cell-based, dendritic cell-based, and CAR T cell therapies have been also shown to be promising and effective immunotherapeutic approaches. In particular, CAR T cell therapy has benefited from advancements in CRISPR-Cas9 genome editing technology, allowing the generation of several modified CAR T cells with enhanced antitumor immunity. However, the emerging SARS-CoV-2 infection could hijack a patient's immune system by releasing pro-inflammatory interleukins and cytokines such as IL-1β, IL-2, IL-6, and IL-10, and IFN-γ and TNF-α, respectively, which can further promote neutrophil extravasation and the vasodilation of blood vessels. Despite the significant development of advanced immunotherapeutic technologies, after a certain period of treatment, cancer relapses due to the development of resistance to immunotherapy. Resistance may be primary (where tumor cells do not respond to the treatment), or secondary or acquired immune resistance (where tumor cells develop resistance gradually to ICIs therapy). In this context, this review aims to address the existing immunotherapeutic technologies against cancer and the resistance mechanisms against immunotherapeutic drugs, and explain the impact of COVID-19 on cancer treatment. In addition, we will discuss what will be the future implementation of these strategies against cancer drug resistance. Finally, we will emphasize the practical steps to lay the groundwork for enlightened policy for intervention and resource allocation to care for cancer patients.
Collapse
Affiliation(s)
- Subhamay Adhikary
- Medical Biotechnology, Faculty of Allied Health Sciences, Chettinad Academy of Research and Education (CARE), Chettinad Hospital and Research Institute (CHRI), Chennai 603103, India
| | - Surajit Pathak
- Medical Biotechnology, Faculty of Allied Health Sciences, Chettinad Academy of Research and Education (CARE), Chettinad Hospital and Research Institute (CHRI), Chennai 603103, India
| | - Vignesh Palani
- Faculty of Medicine, Chettinad Hospital and Research Institute (CHRI), Chennai 603103, India
| | - Ahmet Acar
- Department of Biological Sciences, Middle East Technical University, 06800 Ankara, Türkiye;
| | - Antara Banerjee
- Medical Biotechnology, Faculty of Allied Health Sciences, Chettinad Academy of Research and Education (CARE), Chettinad Hospital and Research Institute (CHRI), Chennai 603103, India
| | - Nader I. Al-Dewik
- Department of Pediatrics, Women’s Wellness and Research Center, Hamad Medical Corporation, Doha 00974, Qatar;
| | - Musthafa Mohamed Essa
- College of Agricultural and Marine Sciences, Sultan Qaboos University, Muscat 123, Oman
| | | | - M. Walid Qoronfleh
- Research & Policy Division, Q3 Research Institute (QRI), Ypsilanti, MI 48917, USA
| |
Collapse
|
17
|
Aguilar JJ, Dhillon V, Balasubramanian S. Manifestation of Pancytopenia Associated with COVID-19 as Paroxysmal Nocturnal Hemoglobinuria (PNH) and Aplastic Anemia (AA). Hematol Rep 2024; 16:42-49. [PMID: 38247995 PMCID: PMC10801523 DOI: 10.3390/hematolrep16010005] [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: 08/21/2023] [Revised: 12/29/2023] [Accepted: 01/10/2024] [Indexed: 01/23/2024] Open
Abstract
We report two cases of pancytopenia in patients after recovering from a mild COVID-19, now presenting as paroxysmal nocturnal hemoglobinuria (PNH) and aplastic anemia. These cases illustrate a common pathway whereby a viral trigger causes the clonal expansion of a hematological disorder. Although the association of both cases with COVID-19 is temporal and COVID-19 may be an incidental diagnosis, the growing evidence related to the hematological effects of SARS-CoV-2 infection highlights the need for further investigation into the hematological consequences of COVID-19, particularly in the post-pandemic era.
Collapse
Affiliation(s)
| | - Vikram Dhillon
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI 48201, USA;
| | - Suresh Balasubramanian
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI 48201, USA;
| |
Collapse
|
18
|
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
|
19
|
Zhang J, Hom K, Zhang C, Nasr M, Gerzanich V, Zhang Y, Tang Q, Xue F, Simard JM, Zhao RY. SARS-CoV-2 ORF3a Protein as a Therapeutic Target against COVID-19 and Long-Term Post-Infection Effects. Pathogens 2024; 13:75. [PMID: 38251382 PMCID: PMC10819734 DOI: 10.3390/pathogens13010075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 01/08/2024] [Accepted: 01/11/2024] [Indexed: 01/23/2024] Open
Abstract
The COVID-19 pandemic caused by SARS-CoV-2 has posed unparalleled challenges due to its rapid transmission, ability to mutate, high mortality and morbidity, and enduring health complications. Vaccines have exhibited effectiveness, but their efficacy diminishes over time while new variants continue to emerge. Antiviral medications offer a viable alternative, but their success has been inconsistent. Therefore, there remains an ongoing need to identify innovative antiviral drugs for treating COVID-19 and its post-infection complications. The ORF3a (open reading frame 3a) protein found in SARS-CoV-2, represents a promising target for antiviral treatment due to its multifaceted role in viral pathogenesis, cytokine storms, disease severity, and mortality. ORF3a contributes significantly to viral pathogenesis by facilitating viral assembly and release, essential processes in the viral life cycle, while also suppressing the body's antiviral responses, thus aiding viral replication. ORF3a also has been implicated in triggering excessive inflammation, characterized by NF-κB-mediated cytokine production, ultimately leading to apoptotic cell death and tissue damage in the lungs, kidneys, and the central nervous system. Additionally, ORF3a triggers the activation of the NLRP3 inflammasome, inciting a cytokine storm, which is a major contributor to the severity of the disease and subsequent mortality. As with the spike protein, ORF3a also undergoes mutations, and certain mutant variants correlate with heightened disease severity in COVID-19. These mutations may influence viral replication and host cellular inflammatory responses. While establishing a direct link between ORF3a and mortality is difficult, its involvement in promoting inflammation and exacerbating disease severity likely contributes to higher mortality rates in severe COVID-19 cases. This review offers a comprehensive and detailed exploration of ORF3a's potential as an innovative antiviral drug target. Additionally, we outline potential strategies for discovering and developing ORF3a inhibitor drugs to counteract its harmful effects, alleviate tissue damage, and reduce the severity of COVID-19 and its lingering complications.
Collapse
Affiliation(s)
- Jiantao Zhang
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (J.Z.); (C.Z.)
| | - Kellie Hom
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD 21201, USA; (K.H.); (F.X.)
| | - Chenyu Zhang
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (J.Z.); (C.Z.)
| | - Mohamed Nasr
- Drug Development and Clinical Sciences Branch, Division of AIDS, NIAID, National Institutes of Health, Bethesda, MD 20892, USA;
| | - Volodymyr Gerzanich
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (V.G.); (J.M.S.)
| | - Yanjin Zhang
- Department of Veterinary Medicine, University of Maryland, College Park, MD 20742, USA;
| | - Qiyi Tang
- Department of Microbiology, Howard University College of Medicine, Washington, DC 20059, USA;
| | - Fengtian Xue
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD 21201, USA; (K.H.); (F.X.)
| | - J. Marc Simard
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (V.G.); (J.M.S.)
- Research & Development Service, VA Maryland Health Care System, Baltimore, MD 21201, USA
| | - Richard Y. Zhao
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (J.Z.); (C.Z.)
- Research & Development Service, VA Maryland Health Care System, Baltimore, MD 21201, USA
- Department of Microbiology-Immunology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Institute of Global Health, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| |
Collapse
|
20
|
Yue Z, Zhang X, Gu Y, Liu Y, Lan LM, Liu Y, Li Y, Yang G, Wan P, Chen X. Regulation and functions of the NLRP3 inflammasome in RNA virus infection. Front Cell Infect Microbiol 2024; 13:1309128. [PMID: 38249297 PMCID: PMC10796458 DOI: 10.3389/fcimb.2023.1309128] [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: 10/07/2023] [Accepted: 11/30/2023] [Indexed: 01/23/2024] Open
Abstract
Virus infection is one of the greatest threats to human life and health. In response to viral infection, the host's innate immune system triggers an antiviral immune response mostly mediated by inflammatory processes. Among the many pathways involved, the nucleotide-binding oligomerization domain (NOD)-like receptor protein 3 (NLRP3) inflammasome has received wide attention in the context of viral infection. The NLRP3 inflammasome is an intracellular sensor composed of three components, including the innate immune receptor NLRP3, adaptor apoptosis-associated speck-like protein containing CARD (ASC), and the cysteine protease caspase-1. After being assembled, the NLRP3 inflammasome can trigger caspase-1 to induce gasdermin D (GSDMD)-dependent pyroptosis, promoting the maturation and secretion of proinflammatory cytokines such as interleukin-1 (IL-1β) and interleukin-18 (IL-18). Recent studies have revealed that a variety of viruses activate or inhibit the NLRP3 inflammasome via viral particles, proteins, and nucleic acids. In this review, we present a variety of regulatory mechanisms and functions of the NLRP3 inflammasome upon RNA viral infection and demonstrate multiple therapeutic strategies that target the NLRP3 inflammasome for anti-inflammatory effects in viral infection.
Collapse
Affiliation(s)
- Zhaoyang Yue
- Institute of Medical Microbiology, College of Life Science and Technology, Jinan University, Guangzhou, China
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou, China
| | - Xuelong Zhang
- Institute of Medical Microbiology, College of Life Science and Technology, Jinan University, Guangzhou, China
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou, China
| | - Yu Gu
- Institute of Medical Microbiology, College of Life Science and Technology, Jinan University, Guangzhou, China
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou, China
| | - Ying Liu
- Institute of Medical Microbiology, College of Life Science and Technology, Jinan University, Guangzhou, China
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou, China
| | - Lin-Miaoshen Lan
- Institute of Medical Microbiology, College of Life Science and Technology, Jinan University, Guangzhou, China
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou, China
| | - Yilin Liu
- Institute of Medical Microbiology, College of Life Science and Technology, Jinan University, Guangzhou, China
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou, China
| | - Yongkui Li
- Institute of Medical Microbiology, College of Life Science and Technology, Jinan University, Guangzhou, China
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou, China
| | - Ge Yang
- Foshan Institute of Medical Microbiology, Foshan, China
| | - Pin Wan
- Foshan Institute of Medical Microbiology, Foshan, China
- Wuhan Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan, China
| | - Xin Chen
- Institute of Medical Microbiology, College of Life Science and Technology, Jinan University, Guangzhou, China
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou, China
| |
Collapse
|
21
|
Khanna K, Yan H, Mehra M, Rohatgi N, Mbalaviele G, Mellins ED, Faccio R. Tmem178 Negatively Regulates IL-1β Production Through Inhibition of the NLRP3 Inflammasome. Arthritis Rheumatol 2024; 76:107-118. [PMID: 37534578 PMCID: PMC11421209 DOI: 10.1002/art.42666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Revised: 06/30/2023] [Accepted: 07/13/2023] [Indexed: 08/04/2023]
Abstract
OBJECTIVE Inflammasomes modulate the release of bioactive interleukin (IL)-1β. Excessive IL-1β levels are detected in patients with systemic juvenile idiopathic arthritis (sJIA) and cytokine storm syndrome (CSS) with mutated and unmutated inflammasome components, raising questions on the mechanisms of IL-1β regulation in these disorders. METHODS To investigate how the NLRP3 inflammasome is modulated in sJIA, we focused on Transmembrane protein 178 (Tmem178), a negative regulator of calcium levels in macrophages, and measured IL-1β and caspase-1 activation in wild-type (WT) and Tmem178-/- macrophages after calcium chelators, silencing of Stim1, a component of store-operated calcium entry (SOCE), or by expressing a Tmem178 mutant lacking the Stromal Interaction Molecule 1 (Stim1) binding site. Mitochondrial function in both genotypes was assessed by measuring oxidative respiration, mitochondrial reactive oxygen species (mtROS), and mitochondrial damage. CSS development was analyzed in Perforin-/- /Tmem178-/- mice infected with lymphocytic choriomeningitis virus (LCMV) in which inflammasome or IL-1β signaling was pharmacologically inhibited. Human TMEM178 and IL1B transcripts were analyzed in data sets of whole blood and peripheral blood monocytes from healthy controls and patients with active sJIA. RESULTS TMEM178 levels are reduced in whole blood and monocytes from patients with sJIA while IL1B levels are increased. Accordingly, Tmem178-/- macrophages produce elevated IL-1β compared with WT cells. The elevated intracellular calcium levels after SOCE activation in Tmem178-/- macrophages induce mitochondrial damage, release mtROS, and ultimately promote NLRP3 inflammasome activation. In vivo, inhibition of inflammasome or IL-1β neutralization prolongs Tmem178-/- mouse survival in LCMV-induced CSS. CONCLUSION Down-regulation of TMEM178 levels may represent a marker of disease activity and help identify patients who could benefit from inflammasome targeting.
Collapse
Affiliation(s)
- Kunjan Khanna
- Washington University in St. Louis, St. Louis, Missouri
| | - Hui Yan
- Washington University in St. Louis, St. Louis, Missouri
| | | | - Nidhi Rohatgi
- Washington University in St. Louis, St. Louis, Missouri
| | | | | | - Roberta Faccio
- Washington University in St. Louis and Shriners Hospital for Children, St. Louis, Missouri
| |
Collapse
|
22
|
Tavares-Júnior JWL, Ciurleo GCV, Feitosa EDAAF, Oriá RB, Braga-Neto P. The Clinical Aspects of COVID and Alzheimer's Disease: A Round-Up of Where Things Stand and Are Headed. J Alzheimers Dis 2024; 99:1159-1171. [PMID: 38848177 DOI: 10.3233/jad-231368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2024]
Abstract
The link between long COVID-19 and brain/cognitive impairments is concerning and may foster a worrisome worldwide emergence of novel cases of neurodegenerative diseases with aging. This review aims to update the knowledge, crosstalk, and possible intersections between the Post-COVID Syndrome (PCS) and Alzheimer's disease (AD). References included in this review were obtained from PubMed searches conducted between October 2023 and November 2023. PCS is a very heterogenous and poorly understood disease with recent evidence of a possible association with chronic diseases such as AD. However, more scientific data is required to establish the link between PCS and AD.
Collapse
Affiliation(s)
| | - Gabriella Cunha Vieira Ciurleo
- Department of Clinical Medicine, Neurology Section, Faculty of Medicine, Federal University of Ceará (UFC), Fortaleza, CE, Brazil
- Department of Morphology and Institute of Biomedicine, Laboratory of the Biology of Tissue Healing, Ontogeny and Nutrition, School of Medicine, Federal University of Ceara, Fortaleza, CE, Brazil
| | | | - Reinaldo B Oriá
- Department of Clinical Medicine, Neurology Section, Faculty of Medicine, Federal University of Ceará (UFC), Fortaleza, CE, Brazil
- Department of Morphology and Institute of Biomedicine, Laboratory of the Biology of Tissue Healing, Ontogeny and Nutrition, School of Medicine, Federal University of Ceara, Fortaleza, CE, Brazil
| | - Pedro Braga-Neto
- Department of Clinical Medicine, Neurology Section, Faculty of Medicine, Federal University of Ceará (UFC), Fortaleza, CE, Brazil
- Center of Health Sciences, State University of Ceará, Fortaleza, CE, Brazil
| |
Collapse
|
23
|
Wan N, Shi J, Xu J, Huang J, Gan D, Tang M, Li X, Huang Y, Li P. Gasdermin D: A Potential New Auxiliary Pan-Biomarker for the Detection and Diagnosis of Diseases. Biomolecules 2023; 13:1664. [PMID: 38002346 PMCID: PMC10669528 DOI: 10.3390/biom13111664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Revised: 11/10/2023] [Accepted: 11/14/2023] [Indexed: 11/26/2023] Open
Abstract
Pyroptosis is a form of programmed cell death mediated by gasdermins, particularly gasdermin D (GSDMD), which is widely expressed in tissues throughout the body. GSDMD belongs to the gasdermin family, which is expressed in a variety of cell types including epithelial cells and immune cells. It is involved in the regulation of anti-inflammatory responses, leading to its differential expression in a wide range of diseases. In this review, we provide an overview of the current understanding of the major activation mechanisms and effector pathways of GSDMD. Subsequently, we examine the importance and role of GSDMD in different diseases, highlighting its potential as a pan-biomarker. We specifically focus on the biological characteristics of GSDMD in several diseases and its promising role in diagnosis, early detection, and differential diagnosis. Furthermore, we discuss the application of GSDMD in predicting prognosis and monitoring treatment efficacy in cancer. This review proposes a new strategy to guide therapeutic decision-making and suggests potential directions for further research into GSDMD.
Collapse
Affiliation(s)
- Ningyi Wan
- Department of Clinical Laboratory, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Jing Shi
- Department of Clinical Laboratory, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Jianguo Xu
- Department of Clinical Laboratory, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Juan Huang
- Department of Information Center, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Delu Gan
- Department of Clinical Laboratory, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Min Tang
- Key Laboratory of Medical Diagnostics Designated by Chinese Ministry of Education, Chongqing Medical University, Chongqing 400016, China
| | - Xiaohan Li
- Department of Clinical Laboratory, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Ying Huang
- Department of Clinical Laboratory, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Pu Li
- Department of Clinical Laboratory, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| |
Collapse
|
24
|
Wang H, Chen L, Li R, Lv C, Xu Y, Xiong Y. Polydopamine-coated mesoporous silica nanoparticles co-loaded with Ziyuglycoside I and Oseltamivir for synergistic treatment of viral pneumonia. Int J Pharm 2023; 645:123412. [PMID: 37703956 DOI: 10.1016/j.ijpharm.2023.123412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 08/18/2023] [Accepted: 09/11/2023] [Indexed: 09/15/2023]
Abstract
Viral pneumonia (VP) is a serious health risk to humans, however, there is still a lack of specific treatments for VP. The spread of the virus in the body induces an excessive inflammatory response that can cause chronic or irreversible damage to lungs. Hence, VP treatment requires rapid clearance of the virus and sustained inflammation control. In this study, an innovative mesoporous silica medication delivery system co-loaded with Ziyuglycoside I(ZgI) and Oseltamivirv(OST) in fast and slow monomeric forms ZgI@MSNs-OST@ Polydopamine (PDA) was prepared for targeted treatment of VP. The prepared ZgI@MSNs-OST@PDA nanoparticles had a homogeneous and membrane-encapsulated spherical structure, with an average particle size of approximately 760 nm. in vitro release and in vivo pharmacokinetic studies demonstrated that ZgI@MSNs-OST@PDA achieved immediate release of OST and sustained release of ZgI, which was readily taken up by the cells. In vitro anti-H1N1 virus experiments showed that nanoparticles rapidly killed the virus in host cells, and the anti-inflammatory effect was sustained and long-lasting, providing excellent protection to host cells. In vivo antiviral pneumonia experiments confirmed the rapid clearance of influenza viruses from mouse lungs and the effective control of overactivated immune responses by ZgI@MSNs-OST@PDA nanoparticles. Through a mechanistic study, we found that the treatment of viral pneumonia with nanoparticles was associated with inhibition of the NLRP3 inflammasome pathway. In conclusion, the constructed nanoparticles achieved synergistic therapeutic effects of ZgI and OST on VP, that is, rapid killing of influenza viruses by OST and effective control of the virus-induced hyperinflammatory response by ZgI.
Collapse
Affiliation(s)
- Hong Wang
- Key Laboratory of Basic Pharmacology of Guizhou Province and School of Pharmacy, Zunyi Medical University, Zunyi, Guizhou 563000, China; Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou 563000, China
| | - Lei Chen
- Key Laboratory of Basic Pharmacology of Guizhou Province and School of Pharmacy, Zunyi Medical University, Zunyi, Guizhou 563000, China; Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou 563000, China
| | - Ruidong Li
- Key Laboratory of Basic Pharmacology of Guizhou Province and School of Pharmacy, Zunyi Medical University, Zunyi, Guizhou 563000, China; Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou 563000, China
| | - Chunmei Lv
- Key Laboratory of Basic Pharmacology of Guizhou Province and School of Pharmacy, Zunyi Medical University, Zunyi, Guizhou 563000, China; Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou 563000, China
| | - Yingshu Xu
- Key Laboratory of Basic Pharmacology of Guizhou Province and School of Pharmacy, Zunyi Medical University, Zunyi, Guizhou 563000, China; Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou 563000, China.
| | - Yongai Xiong
- Key Laboratory of Basic Pharmacology of Guizhou Province and School of Pharmacy, Zunyi Medical University, Zunyi, Guizhou 563000, China; Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou 563000, China.
| |
Collapse
|
25
|
Mahalingam SS, Jayaraman S, Arunkumar A, Dudley HM, Anthony DD, Shive CL, Jacobson JM, Pandiyan P. Distinct SARS-CoV-2 specific NLRP3 and IL-1β responses in T cells of aging patients during acute COVID-19 infection. Front Immunol 2023; 14:1231087. [PMID: 37799713 PMCID: PMC10548880 DOI: 10.3389/fimmu.2023.1231087] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 08/31/2023] [Indexed: 10/07/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) causes Coronavirus Disease 2019 (COVID-19) that presents with varied clinical manifestations ranging from asymptomatic or mild infections and pneumonia to severe cases associated with cytokine storm, acute respiratory distress syndrome (ARDS), and even death. The underlying mechanisms contributing to these differences are unclear, although exacerbated inflammatory sequelae resulting from infection have been implicated. While advanced aging is a known risk factor, the precise immune parameters that determine the outcome of SARS-CoV-2 infection in elderly individuals are not understood. Here, we found aging-associated (age ≥61) intrinsic changes in T cell responses when compared to those from individuals aged ≤ 60, even among COVID-positive patients with mild symptoms. Specifically, when stimulated with SARS-CoV-2 peptides in vitro, peripheral blood mononuclear cell (PBMC) CD4+ and CD8+ T cells from individuals aged ≥61 showed a diminished capacity to produce IFN-γ and IL-1β. Although they did not have severe disease, aged individuals also showed a higher frequency of PD-1+ cells and significantly diminished IFN-γ/PD-1 ratios among T lymphocytes upon SARS-CoV-2 peptide stimulation. Impaired T cell IL-1β expression coincided with reduced NLRP3 levels in T lymphocytes. However, the expression of these molecules was not affected in the monocytes of individuals aged ≥61. Together, these data reveal SARS-CoV-2-specific CD4+ and CD8+ T-cell intrinsic cytokine alterations in the individuals older than 61 and may provide new insights into dysregulated COVID-directed immune responses in the elderly.
Collapse
Affiliation(s)
- Shanmuga Sundaram Mahalingam
- Department of Biological Sciences, School of Dental Medicine, Case Western Reserve University, Cleveland, OH, United States
| | - Sangeetha Jayaraman
- Department of Biological Sciences, School of Dental Medicine, Case Western Reserve University, Cleveland, OH, United States
| | - Adhvika Arunkumar
- Department of Biological Sciences, School of Dental Medicine, Case Western Reserve University, Cleveland, OH, United States
| | - Holly M. Dudley
- Department of Molecular Biology and Microbiology, School of Medicine, Case Western Reserve University, Cleveland, OH, United States
| | - Donald D. Anthony
- Department of Rheumatology, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH, United States
- Department of Pathology, Case Western Reserve University, Cleveland, OH, United States
- Center for AIDS Research, School of Medicine, Case Western Reserve University, Cleveland, OH, United States
- Department of Medicine, School of Medicine, University Hospitals, Case Western Reserve University, Cleveland, OH, United States
| | - Carey L. Shive
- Department of Rheumatology, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH, United States
- Department of Pathology, Case Western Reserve University, Cleveland, OH, United States
| | - Jeffrey M. Jacobson
- Center for AIDS Research, School of Medicine, Case Western Reserve University, Cleveland, OH, United States
- Department of Medicine, School of Medicine, University Hospitals, Case Western Reserve University, Cleveland, OH, United States
| | - Pushpa Pandiyan
- Department of Biological Sciences, School of Dental Medicine, Case Western Reserve University, Cleveland, OH, United States
- Department of Rheumatology, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH, United States
- Center for AIDS Research, School of Medicine, Case Western Reserve University, Cleveland, OH, United States
| |
Collapse
|
26
|
Almutawif YA, Al‐kuraishy HM, Al‐Gareeb AI, Alexiou A, Papadakis M, Eid HMA, Saad HM, Batiha GE. Insights on Covid-19 with superimposed pulmonary histoplasmosis: The possible nexus. Immun Inflamm Dis 2023; 11:e989. [PMID: 37773721 PMCID: PMC10540147 DOI: 10.1002/iid3.989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 08/06/2023] [Accepted: 08/09/2023] [Indexed: 10/01/2023] Open
Abstract
A novel coronavirus (CoV) known as severe acute respiratory syndrome CoV type 2 is the causative agent for the development of CoV disease 2019 (Covid-19). Covid-19 may increase the risk of developing pulmonary histoplasmosis due to immune dysregulation. In addition, Covid-19 may enhance the propagation of acute pulmonary histoplasmosis due to lung injury and inflammation, and using corticosteroids in severely affected Covid-19 patients may reactivate latent pulmonary histoplasmosis. Likewise, activation of inflammatory signaling pathways during H. capsulatum infection may increase the severity of Covid-19 and vice versa. Furthermore, lymphopenia in Covid-19 may increase the risk for the progress of pulmonary histoplasmosis besides activation of inflammatory signaling pathways during H. capsulatum infection may increase the severity of Covid-19 and vice versa. Therefore, this critical review aimed to find the potential link between Covid-19 pneumonia and pulmonary histoplasmosis concerning the immunological response.
Collapse
Affiliation(s)
- Yahya A. Almutawif
- Department of Medical Laboratories Technology, College of Applied Medical SciencesTaibah UniversityMadinahSaudi Arabia
| | - Hayder M. Al‐kuraishy
- Department of Clinical Pharmacology and Medicine, College of MedicineAl‐Mustansiriyia UniversityBaghdadIraq
| | - Ali I. Al‐Gareeb
- Department of Clinical Pharmacology and Medicine, College of MedicineAl‐Mustansiriyia UniversityBaghdadIraq
| | - Athanasios Alexiou
- Department of Science and EngineeringNovel Global Community Educational FoundationHebershamNew South WalesAustralia
- AFNP MedWienAustria
| | - Marios Papadakis
- Department of Surgery II, University Hospital Witten‐Herdecke, Heusnerstrasse 40University of Witten‐HerdeckeWuppertalGermany
| | - Hamza M. A. Eid
- Department of Medical Laboratories Technology, College of Applied Medical SciencesTaibah UniversityMadinahSaudi Arabia
| | - Hebatallah M. Saad
- Department of Pathology, Faculty of Veterinary MedicineMatrouh UniversityMarsaMatruhEgypt
| | - Gaber El‐Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary MedicineDamanhour UniversityDamanhourAlBeheiraEgypt
| |
Collapse
|
27
|
Tsiakalos A, Ziakas PD, Polyzou E, Schinas G, Akinosoglou K. Early Fluvoxamine Reduces the Risk for Clinical Deterioration in Symptomatic Outpatients with COVID-19: A Real-World, Retrospective, before-after Analysis. Microorganisms 2023; 11:2073. [PMID: 37630633 PMCID: PMC10459506 DOI: 10.3390/microorganisms11082073] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 08/06/2023] [Accepted: 08/09/2023] [Indexed: 08/27/2023] Open
Abstract
Fluvoxamine, a selective serotonin reuptake inhibitor with anti-inflammatory properties, has gained attention as a repurposed drug to treat COVID-19. We aimed to explore the potential benefit of fluvoxamine on outpatients with early SARS-CoV-2 infection. We performed a retrospective study of fluvoxamine adult outpatients with symptomatic COVID-19 disease of early onset (<5 days), in the context of an infectious diseases private practice, between September-December 2021, in Greece. Patients with disease duration ≥5 days, dyspnea and/or hypoxemia with oxygen saturation <94% in room air and pregnancy were excluded from the analysis. In total, 103 patients, 54 males/49 females with a median age of 47 years (39-56), were included in this study. Patient characteristics were balanced before and after the introduction of fluvoxamine. Two patients in the fluvoxamine arm (3.8%; 95% CI 0.4-13) had clinical deterioration compared to 8 patients in the standard of care group (16%; 95% CI 7.2-29.1, p < 0.04). After controlling for age, sex, body mass index > 30 and vaccination status, fluvoxamine was independently associated with a lower risk of clinical deterioration (adj. OR 0.12; 95% CI 0.02-0.70, p < 0.02). Adding on fluvoxamine to treatment for early symptomatic COVID-19 patients may protect them from clinical deterioration and hospitalization, and it is an appealing low-cost, low-toxicity option in the community setting and warrants further investigation.
Collapse
Affiliation(s)
| | | | - Eleni Polyzou
- Dept of Internal Medicine and Infectious Diseases, Medical School, University General Hospital of Patras, University of Patras, 26504 Rio, Greece; (E.P.); (G.S.); (K.A.)
| | - Georgios Schinas
- Dept of Internal Medicine and Infectious Diseases, Medical School, University General Hospital of Patras, University of Patras, 26504 Rio, Greece; (E.P.); (G.S.); (K.A.)
| | - Karolina Akinosoglou
- Dept of Internal Medicine and Infectious Diseases, Medical School, University General Hospital of Patras, University of Patras, 26504 Rio, Greece; (E.P.); (G.S.); (K.A.)
| |
Collapse
|
28
|
Yan L, Wu J, Fan X, Liu J, Zhang L, Hu J, Li X, Su Y, Zhang F, Xu X, Chen X, Yang H. Analysis of risk factors and short-term prognostic factors of arrhythmia in patients infected with mild/moderate SARS-CoV-2 Omicron variant. Front Med (Lausanne) 2023; 10:1186200. [PMID: 37575983 PMCID: PMC10413106 DOI: 10.3389/fmed.2023.1186200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 07/11/2023] [Indexed: 08/15/2023] Open
Abstract
Background Complications, including arrhythmia, following severe acute respiratory syndrome-coronavirus-2 (SARS-CoV-2) infection continue to be of concern. Omicron is the mainstream SARS-CoV-2 mutant circulating in mainland China. At present, there are few epidemiological studies concerning the relationship between arrhythmia and Omicron variant infection in mainland China. Objectives To investigate the risk factors of arrhythmia in patients infected with the SARS-CoV-2 Omicron variant and the factors influencing prognosis. Methods Data from 192 Omicron infected patients with symptoms of arrhythmia (AH group) and 100 Omicron infected patients without arrhythmia (Control group) were collected. Patients in the AH group were divided into the good and poor prognosis groups, according to the follow-up results 4-6 weeks after infection. The general and clinical data between the AH and Control groups, and between the good and poor prognosis groups were compared. The variables with differences between the groups were included in the multivariate logistic regression analysis, and the quantitative variables were analyzed by receiver operating characteristic curve to obtain their cut-off values. Results Compared with the control group, the body mass index (BMI), proportion of patients with a history of arrhythmia, proportion of antibiotics taken, heart rate, moderate disease severity, white blood cell (WBC) count, and the aspartate aminotransferase, creatine kinase (CK), CK isoenzyme (CK-MB), myoglobin (Mb), high-sensitive troponin I (hs-cTnI), lymphocyte ratio and high sensitivity C-reactive protein (hs-CRP) levels in the AH group were significantly higher (p < 0.05). In addition, obesity (BMI ≥24 kg/m2), fast heart rate (≥100 times/min), moderate disease severity, and WBC, CK-MB and hs-cTnI levels were independent risk factors of arrhythmia for patients with Omicron infection (p < 0.05), and hs-CRP was a protective factor (p < 0.05). Compared with the good prognosis group, the age, proportion of patients with a history of arrhythmia, heart rate, proportion of moderate disease severity, and hs-CRP, CK, Mb and hs-cTnI levels were significantly higher in the poor prognosis group, while the proportion of vaccination was lower in the poor prognosis group (p < 0.05). Advanced age (≥65 years old), proportion of history of arrhythmia, moderate disease severity, vaccination, and hs-CRP, Mb and cTnI levels were independent factors for poor prognosis of patients with arrhythmia (p < 0.05). Conclusion The factors that affect arrhythmia and the prognosis of patients infected with Omicron include obesity, high heart rate, severity of the disease, age. history of arrhythmia, WBC, hs-CRP, and myocardial injury indexes, which could be used to evaluate and prevent arrhythmia complications in patients in the future.
Collapse
Affiliation(s)
- Lijie Yan
- Heart Center of Henan Provincial People’s Hospital, Fuwai Central China Cardiovascular Hospital, Fuwai Central China Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Jintao Wu
- Heart Center of Henan Provincial People’s Hospital, Fuwai Central China Cardiovascular Hospital, Fuwai Central China Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Xianwei Fan
- Heart Center of Henan Provincial People’s Hospital, Fuwai Central China Cardiovascular Hospital, Fuwai Central China Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Jingjing Liu
- Heart Center of Henan Provincial People’s Hospital, Fuwai Central China Cardiovascular Hospital, Fuwai Central China Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Leiming Zhang
- Heart Center of Henan Provincial People’s Hospital, Fuwai Central China Cardiovascular Hospital, Fuwai Central China Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Juan Hu
- Heart Center of Henan Provincial People’s Hospital, Fuwai Central China Cardiovascular Hospital, Fuwai Central China Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Xuejie Li
- Heart Center of Henan Provincial People’s Hospital, Fuwai Central China Cardiovascular Hospital, Fuwai Central China Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yandong Su
- Heart Center of Henan Provincial People’s Hospital, Fuwai Central China Cardiovascular Hospital, Fuwai Central China Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Futao Zhang
- Department of Cardiology, Henan University People’s Hospital, Henan Provincial People’s Hospital, Zhengzhou, Henan, China
| | - Xizheng Xu
- Department of Cardiology, Henan University People’s Hospital, Henan Provincial People’s Hospital, Zhengzhou, Henan, China
| | - Xiaosheng Chen
- Heart Center of Henan Provincial People’s Hospital, Fuwai Central China Cardiovascular Hospital, Fuwai Central China Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Haitao Yang
- Heart Center of Henan Provincial People’s Hospital, Fuwai Central China Cardiovascular Hospital, Fuwai Central China Hospital of Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
29
|
Maity S, Santra A, Vardhan Hebbani A, Pulakuntla S, Chatterjee A, Rao Badri K, Damodara Reddy V. Targeting cytokine storm as the potential anti-viral therapy: Implications in regulating SARS-CoV-2 pathogenicity. Gene 2023:147612. [PMID: 37423400 DOI: 10.1016/j.gene.2023.147612] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 06/18/2023] [Accepted: 07/03/2023] [Indexed: 07/11/2023]
Abstract
The latest global pandemic corona virus disease - 2019 (COVID-19) caused by the virus SARS-CoV-2 is still a matter of worrying concern both for the scientific communities and health care organizations. COVID-19 disease is proved to be a highly contagious disease transmitted through respiratory droplets and even close contact with affected individuals. COVID-19 disease is also understood to exhibit diverse symptoms of ranging severities i.e., from mild fatigue to death. Affected individuals' susceptibility to induce immunologic dysregulation phenomena termed 'cytokine storm' seems to be playing the damaging role of escalating the disease manifestation from mild to severe. Cytokine storm in patients with severe symptoms is understood to be characterized by enhanced serum levels of many cytokines including interleukin-1β, interleukin-6, IL-10, TNF, interferon-γ, MIP-1α, MIP-1β and VEGF. Since cytokine production in general is the most important antiviral defense response, understanding the COVID-19 associated cytokine storm in particular and differentiating it from the regular cytokine production response becomes crucial in developing an effective therapeutic strategy.This review focuses on the potential targeting of COVID-19 associated cytokine storm and its challenges.
Collapse
Affiliation(s)
- Subashish Maity
- Department of Biotechnology, REVA University, Bengaluru-560064, Karnataka, India
| | - Ayantika Santra
- Department of Biochemistry, Indian Academy Degree College, Bengaluru, 560 043, India
| | | | - Swetha Pulakuntla
- Department of Biotechnology, REVA University, Bengaluru-560064, Karnataka, India
| | - Ankita Chatterjee
- Department of Biotechnology, REVA University, Bengaluru-560064, Karnataka, India
| | - Kameswara Rao Badri
- Department of Pharmacology and Toxicology, Cardiovascular Research Institute, Morehouse School of Medicine, GA, Atlanta-30310, USA; Clinical Analytical Chemistry Laboratory, COVID-19 Testing Laboratory, Morehouse School of Medicine, GA, Atlanta-30310, USA.
| | - Vaddi Damodara Reddy
- Department of Biotechnology, REVA University, Bengaluru-560064, Karnataka, India.
| |
Collapse
|
30
|
Zhang Z, Zhang H, Zhang Y, Zhang Q, Liu Q, Hu Y, Chen X, Wang J, Shi Y, Deng C, Gong P, Zhang B, Li X, Zhu B, Ye H. Oridonin inhibits SARS-CoV-2 replication by targeting viral proteinase and polymerase. Virol Sin 2023; 38:470-479. [PMID: 37127212 PMCID: PMC10148713 DOI: 10.1016/j.virs.2023.04.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Accepted: 04/27/2023] [Indexed: 05/03/2023] Open
Abstract
COVID-19 has become a global public health crisis since its outbreak in China in December 2019. Currently there are few clinically effective drugs to combat SARS-CoV-2 infection. The main protein (Mpro), papain-like protease (PLpro) and RNA-dependent RNA polymerase (RdRp) of SARS-CoV-2 are involved in the viral replication, and might be prospective targets for anti-coronavirus drug development. Here, we investigated the antiviral activity of oridonin, a natural small-molecule compound, against SARS-CoV-2 infection in vitro. The time-of-addition analysis showed that oridonin efficiently inhibited SARS-CoV-2 infection by interfering with the genome replication at the post-entry stage. Mechanistically, the inhibition of viral replication by oridonin depends on the oxidation activity of α, β-unsaturated carbonyl. Further experiments showed that oridonin not only effectively inhibited SARS-CoV-2 Mpro activity, but also had some inhibitory effects on PLpro-mediated deubiquitinating and viral polymerase-catalyzed RNA elongation activities at high concentrations. In particular, oridonin could inhibit the bat SARS-like CoV and the newly emerged SARS-CoV-2 omicron variants (BA.1 and BA.2), which highlights its potential as a pan-coronavirus antiviral agent. Overall, our data provide strong evidence that oridonin is an efficient antiviral agent against SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Zherui Zhang
- Virus Laboratory, Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China; Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Hongqing Zhang
- Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yanan Zhang
- Virus Laboratory, Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China; Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Qiuyan Zhang
- Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Qiaojie Liu
- Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Yanyan Hu
- Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xiaoling Chen
- Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Jing Wang
- Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yujia Shi
- Hunan Normal University, School of Medicine, Changsha, 410081, China
| | - Chenglin Deng
- Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Peng Gong
- Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Bo Zhang
- Virus Laboratory, Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China; Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, China; University of Chinese Academy of Sciences, Beijing, 100049, China.
| | - Xiaodan Li
- Hunan Normal University, School of Medicine, Changsha, 410081, China.
| | - Bing Zhu
- Virus Laboratory, Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China.
| | - Hanqing Ye
- Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, China.
| |
Collapse
|
31
|
Wang M, Yu F, Chang W, Zhang Y, Zhang L, Li P. Inflammasomes: a rising star on the horizon of COVID-19 pathophysiology. Front Immunol 2023; 14:1185233. [PMID: 37251383 PMCID: PMC10213254 DOI: 10.3389/fimmu.2023.1185233] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 05/02/2023] [Indexed: 05/31/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a contagious respiratory virus that is the cause of the coronavirus disease 2019 (COVID-19) pandemic which has posed a serious threat to public health. COVID-19 is characterized by a wide spectrum of clinical manifestations, ranging from asymptomatic infection to mild cold-like symptoms, severe pneumonia or even death. Inflammasomes are supramolecular signaling platforms that assemble in response to danger or microbial signals. Upon activation, inflammasomes mediate innate immune defense by favoring the release of proinflammatory cytokines and triggering pyroptotic cell death. Nevertheless, abnormalities in inflammasome functioning can result in a variety of human diseases such as autoimmune disorders and cancer. A growing body of evidence has showed that SARS-CoV-2 infection can induce inflammasome assembly. Dysregulated inflammasome activation and consequent cytokine burst have been associated with COVID-19 severity, alluding to the implication of inflammasomes in COVID-19 pathophysiology. Accordingly, an improved understanding of inflammasome-mediated inflammatory cascades in COVID-19 is essential to uncover the immunological mechanisms of COVID-19 pathology and identify effective therapeutic approaches for this devastating disease. In this review, we summarize the most recent findings on the interplay between SARS-CoV-2 and inflammasomes and the contribution of activated inflammasomes to COVID-19 progression. We dissect the mechanisms involving the inflammasome machinery in COVID-19 immunopathogenesis. In addition, we provide an overview of inflammasome-targeted therapies or antagonists that have potential clinical utility in COVID-19 treatment.
Collapse
Affiliation(s)
- Man Wang
- *Correspondence: Man Wang, ; Peifeng Li,
| | | | | | | | | | - Peifeng Li
- *Correspondence: Man Wang, ; Peifeng Li,
| |
Collapse
|
32
|
Shen J, Fan J, Zhao Y, Jiang D, Niu Z, Zhang Z, Cao G. Innate and adaptive immunity to SARS-CoV-2 and predisposing factors. Front Immunol 2023; 14:1159326. [PMID: 37228604 PMCID: PMC10203583 DOI: 10.3389/fimmu.2023.1159326] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Accepted: 04/27/2023] [Indexed: 05/27/2023] Open
Abstract
The coronavirus disease 2019 (COVID-19) pandemic, caused by severe acute respiratory syndrome coronavirus (SARS-CoV-2), has affected all countries worldwide. Although some symptoms are relatively mild, others are still associated with severe and even fatal clinical outcomes. Innate and adaptive immunity are important for the control of SARS-CoV-2 infections, whereas a comprehensive characterization of the innate and adaptive immune response to COVID-19 is still lacking and the mechanisms underlying immune pathogenesis and host predisposing factors are still a matter of scientific debate. Here, the specific functions and kinetics of innate and adaptive immunity involved in SARS-CoV-2 recognition and resultant pathogenesis are discussed, as well as their immune memory for vaccinations, viral-mediated immune evasion, and the current and future immunotherapeutic agents. We also highlight host factors that contribute to infection, which may deepen the understanding of viral pathogenesis and help identify targeted therapies that attenuate severe disease and infection.
Collapse
Affiliation(s)
- Jiaying Shen
- Tongji University School of Medicine, Tongji University, Shanghai, China
| | - Junyan Fan
- Department of Epidemiology, Shanghai Key Laboratory of Medical Bioprotection, Key Laboratory of Biological Defense, Ministry of Education, Second Military Medical University, Shanghai, China
| | - Yue Zhao
- Department of Epidemiology, Shanghai Key Laboratory of Medical Bioprotection, Key Laboratory of Biological Defense, Ministry of Education, Second Military Medical University, Shanghai, China
| | - Doming Jiang
- Tongji University School of Medicine, Tongji University, Shanghai, China
| | - Zheyun Niu
- Tongji University School of Medicine, Tongji University, Shanghai, China
| | - Zihan Zhang
- Tongji University School of Medicine, Tongji University, Shanghai, China
| | - Guangwen Cao
- Tongji University School of Medicine, Tongji University, Shanghai, China
- Department of Epidemiology, Shanghai Key Laboratory of Medical Bioprotection, Key Laboratory of Biological Defense, Ministry of Education, Second Military Medical University, Shanghai, China
| |
Collapse
|
33
|
Nasiri K, Mohammadzadehsaliani S, Kheradjoo H, Shabestari AM, Eshaghizadeh P, Pakmehr A, Alsaffar MF, Al-Naqeeb BZT, Yasamineh S, Gholizadeh O. Spotlight on the impact of viral infections on Hematopoietic Stem Cells (HSCs) with a focus on COVID-19 effects. Cell Commun Signal 2023; 21:103. [PMID: 37158893 PMCID: PMC10165295 DOI: 10.1186/s12964-023-01122-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 04/05/2023] [Indexed: 05/10/2023] Open
Abstract
Hematopoietic stem cells (HSCs) are known for their significant capability to reconstitute and preserve a functional hematopoietic system in long-term periods after transplantation into conditioned hosts. HSCs are thus crucial cellular targets for the continual repair of inherited hematologic, metabolic, and immunologic disorders. In addition, HSCs can undergo various fates, such as apoptosis, quiescence, migration, differentiation, and self-renewal. Viruses continuously pose a remarkable health risk and request an appropriate, balanced reaction from our immune system, which as well as affects the bone marrow (BM). Therefore, disruption of the hematopoietic system due to viral infection is essential. In addition, patients for whom the risk-to-benefit ratio of HSC transplantation (HSCT) is acceptable have seen an increase in the use of HSCT in recent years. Hematopoietic suppression, BM failure, and HSC exhaustion are all linked to chronic viral infections. Virus infections continue to be a leading cause of morbidity and mortality in HSCT recipients, despite recent advancements in the field. Furthermore, whereas COVID-19 manifests initially as an infection of the respiratory tract, it is now understood to be a systemic illness that significantly impacts the hematological system. Patients with advanced COVID-19 often have thrombocytopenia and blood hypercoagulability. In the era of COVID-19, Hematological manifestations of COVID-19 (i.e., thrombocytopenia and lymphopenia), the immune response, and HSCT may all be affected by the SARS-CoV-2 virus in various ways. Therefore, it is important to determine whether exposure to viral infections may affect HSCs used for HSCT, as this, in turn, may affect engraftment efficiency. In this article, we reviewed the features of HSCs, and the effects of viral infections on HSCs and HSCT, such as SARS-CoV-2, HIV, cytomegalovirus, Epstein-Barr virus, HIV, etc. Video Abstract.
Collapse
Affiliation(s)
- Kamyar Nasiri
- Department of Dentistry, Islamic Azad University, Tehran, Iran
| | | | | | | | - Parisa Eshaghizadeh
- Department of Dental Surgery, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Azin Pakmehr
- Medical Doctor, Tehran University of Medical Science, Tehran, Iran
| | - Marwa Fadhil Alsaffar
- Medical Laboratories Techniques Department / AL-Mustaqbal University College, 51001, Hillah, Babil, Iraq
| | | | - Saman Yasamineh
- Research Center for Clinical Virology, Tehran University of Medical Sciences, Tehran, Iran.
| | - Omid Gholizadeh
- Department of Bacteriology and Virology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
34
|
Moon Y. Gut distress and intervention via communications of SARS-CoV-2 with mucosal exposome. Front Public Health 2023; 11:1098774. [PMID: 37139365 PMCID: PMC10150023 DOI: 10.3389/fpubh.2023.1098774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 03/27/2023] [Indexed: 05/05/2023] Open
Abstract
Acute coronavirus disease 2019 (COVID-19) has been associated with prevalent gastrointestinal distress, characterized by fecal shedding of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) RNA or persistent antigen presence in the gut. Using a meta-analysis, the present review addressed gastrointestinal symptoms, such as nausea, vomiting, abdominal pain, and diarrhea. Despite limited data on the gut-lung axis, viral transmission to the gut and its influence on gut mucosa and microbial community were found to be associated by means of various biochemical mechanisms. Notably, the prolonged presence of viral antigens and disrupted mucosal immunity may increase gut microbial and inflammatory risks, leading to acute pathological outcomes or post-acute COVID-19 symptoms. Patients with COVID-19 exhibit lower bacterial diversity and a higher relative abundance of opportunistic pathogens in their gut microbiota than healthy controls. Considering the dysbiotic changes during infection, remodeling or supplementation with beneficial microbial communities may counteract adverse outcomes in the gut and other organs in patients with COVID-19. Moreover, nutritional status, such as vitamin D deficiency, has been associated with disease severity in patients with COVID-19 via the regulation of the gut microbial community and host immunity. The nutritional and microbiological interventions improve the gut exposome including the host immunity, gut microbiota, and nutritional status, contributing to defense against acute or post-acute COVID-19 in the gut-lung axis.
Collapse
Affiliation(s)
- Yuseok Moon
- Laboratory of Mucosal Exposome and Biomodulation, Department of Integrative Biomedical Sciences, Pusan National University, Yangsan-si, Republic of Korea
- Biomedical Research Institute, Pusan National University, Busan, Republic of Korea
- Graduate Program of Genomic Data Sciences, Pusan National University, Yangsan-si, Republic of Korea
| |
Collapse
|
35
|
Khanna K, Yan H, Mehra M, Rohatgi N, Mbalaviele G, Faccio R. Tmem178 negatively regulates IL-1β production through inhibition of the NLRP3 inflammasome. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.07.531385. [PMID: 36945522 PMCID: PMC10028891 DOI: 10.1101/2023.03.07.531385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/09/2023]
Abstract
Objective Inflammasomes modulate the release of bioactive IL-1β. Excessive IL-1β levels are detected in patients with systemic juvenile idiopathic arthritis (sJIA) and cytokine storm syndrome (CSS) with mutated and unmutated inflammasome components, raising questions on the mechanisms of IL-1β regulation in these disorders. Methods To investigate how the NLRP3 inflammasome is modulated in sJIA, we focused on Tmem178, a negative regulator of calcium levels in macrophages, and measured IL-1β and caspase-1 activation in wild-type (WT) and Tmem178 -/- macrophages following calcium chelators, silencing of Stim1, a component of store-operated calcium entry (SOCE), or by expressing a Tmem178 mutant lacking Stim1 binding site. Mitochondrial function in both genotypes was assessed by measuring oxidative respiration, mitochondrial reactive oxygen species (mtROS), and mitochondrial damage. CSS development was analyzed in Perforin -/- /Tmem178 -/- mice infected with LCMV in which inflammasome or IL-1 signaling was pharmacologically inhibited. Human TMEM178 and IL-1B transcripts were analyzed in a dataset of peripheral blood monocytes from healthy controls and active sJIA patients. Results TMEM178 levels are reduced in monocytes from sJIA patients while IL-1B show increased levels. Accordingly, Tmem178 -/- macrophages produce elevated IL-1β compared to WT cells. The elevated intracellular calcium levels following SOCE activation in Tmem178 -/- macrophages induce mitochondrial damage, release mtROS, and ultimately, promote NLRP3 inflammasome activation. In vivo , inhibition of inflammasome or IL-1 neutralization prolongs Tmem178 -/- mouse survival to LCMV-induced CSS. Conclusion Downregulation of Tmem178 levels may represent a new biomarker to identify sJIA/CSS patients that could benefit from receiving drugs targeting inflammasome signaling.
Collapse
|
36
|
Liu Z, Han Z, Jin X, An J, Kim J, Chen W, Kim JS, Zheng J, Deng J. Regulating the microenvironment with nanomaterials: Potential strategies to ameliorate COVID-19. Acta Pharm Sin B 2023; 13:S2211-3835(23)00054-0. [PMID: 36846153 PMCID: PMC9941074 DOI: 10.1016/j.apsb.2023.02.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 02/08/2023] [Accepted: 02/10/2023] [Indexed: 02/23/2023] Open
Abstract
COVID-19, caused by SARS-CoV-2, has resulted in serious economic and health burdens. Current treatments remain inadequate to extinguish the epidemic, and efficient therapeutic approaches for COVID-19 are urgently being sought. Interestingly, accumulating evidence suggests that microenvironmental disorder plays an important role in the progression of COVID-19 in patients. In addition, recent advances in nanomaterial technologies provide promising opportunities for alleviating the altered homeostasis induced by a viral infection, providing new insight into COVID-19 treatment. Most literature reviews focus only on certain aspects of microenvironment alterations and fail to provide a comprehensive overview of the changes in homeostasis in COVID-19 patients. To fill this gap, this review systematically discusses alterations of homeostasis in COVID-19 patients and potential mechanisms. Next, advances in nanotechnology-based strategies for promoting homeostasis restoration are summarized. Finally, we discuss the challenges and prospects of using nanomaterials for COVID-19 management. This review provides a new strategy and insights into treating COVID-19 and other diseases associated with microenvironment disorders.
Collapse
Affiliation(s)
- Zhicheng Liu
- Department of Urology, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing 400037, China
- Department of Urology, Urological Surgery Research Institute, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Zhuolei Han
- Institute of Burn Research, Southwest Hospital, State Key Lab of Trauma, Burn and Combined Injury, Chongqing Key Laboratory for Disease Proteomics, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Xin Jin
- Tianjin Key Laboratory of Composite and Functional Materials, School of Materials Science and Engineering, Tianjin University, Tianjin 300350, China
| | - Jusung An
- Department of Chemistry, Korea University, Seoul 02841, South Korea
| | - Jaewon Kim
- Department of Chemistry, Korea University, Seoul 02841, South Korea
| | - Wenting Chen
- Department of Rheumatology and Clinical Immunology, Army Medical Center, Third Military Medical University (Army Medical University), Chongqing 400042, China
| | - Jong Seung Kim
- Department of Chemistry, Korea University, Seoul 02841, South Korea
| | - Ji Zheng
- Department of Urology, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing 400037, China
- Department of Urology, Urological Surgery Research Institute, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Jun Deng
- Institute of Burn Research, Southwest Hospital, State Key Lab of Trauma, Burn and Combined Injury, Chongqing Key Laboratory for Disease Proteomics, Third Military Medical University (Army Medical University), Chongqing 400038, China
| |
Collapse
|
37
|
Amini N, Mahdavi M, Choubdar H, Abedini A, Shalbaf A, Lashgari R. Automated prediction of COVID-19 mortality outcome using clinical and laboratory data based on hierarchical feature selection and random forest classifier. Comput Methods Biomech Biomed Engin 2023; 26:160-173. [PMID: 35297747 DOI: 10.1080/10255842.2022.2050906] [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] [Indexed: 01/25/2023]
Abstract
Early prediction of COVID-19 mortality outcome can decrease expiration risk by alerting healthcare personnel to assure efficient resource allocation and treatment planning. This study introduces a machine learning framework for the prediction of COVID-19 mortality using demographics, vital signs, and laboratory blood tests (complete blood count (CBC), coagulation, kidney, liver, blood gas, and general). 41 features from 244 COVID-19 patients were recorded on the first day of admission. In this study, first, the features in each of the eight categories were investigated. Afterward, features that have an area under the receiver operating characteristic curve (AUC) above 0.6 and the p-value criterion from the Wilcoxon rank-sum test below 0.005 were used as selected features for further analysis. Then five feature reduction methods, Forward Feature selection, minimum Redundancy Maximum Relevance, Relieff, Linear Discriminant Analysis, and Neighborhood Component Analysis were utilized to select the best combination of features. Finally, seven classifiers frameworks, random forest (RF), support vector machine, logistic regression (LR), K nearest neighbors, Artifical neural network, bagging, and boosting were used to predict the mortality outcome of COVID-19 patients. The results revealed that the combination of features in CBC and then vital signs had the highest mortality classification parameters, respectively. Furthermore, the RF classifier with hierarchical feature selection algorithms via Forward Feature selection had the highest classification power with an accuracy of 92.08 ± 2.56. Therefore, our proposed method can be confidently used as a valuable assistant prognostic tool to sieve patients with high mortality risks.
Collapse
Affiliation(s)
- Nasrin Amini
- Department of Biomedical Engineering and Medical Physics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahdi Mahdavi
- Institute of Medical Science and Technology (IMSAT), Shahid Beheshti University, Tehran, Iran.,School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hadi Choubdar
- Institute of Medical Science and Technology (IMSAT), Shahid Beheshti University, Tehran, Iran.,School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Atefeh Abedini
- Chronic Respiratory Diseases Research Center, National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ahmad Shalbaf
- Department of Biomedical Engineering and Medical Physics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Reza Lashgari
- Institute of Medical Science and Technology (IMSAT), Shahid Beheshti University, Tehran, Iran
| |
Collapse
|
38
|
Kullappan M, Mary U, Ambrose JM, Veeraraghavan VP, Surapaneni KM. Elucidating the role of N440K mutation in SARS-CoV-2 spike - ACE-2 binding affinity and COVID-19 severity by virtual screening, molecular docking and dynamics approach. J Biomol Struct Dyn 2023; 41:912-929. [PMID: 34904526 DOI: 10.1080/07391102.2021.2014973] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
COVID-19 has become a public health concern around the world. The frequency of N440K variant was higher during the second wave in South India. The mutation was observed in the Receptor Binding Domain region (RBD) of the Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2) spike (S) protein. The binding affinity of SARS-CoV-2-Angiotensin-Converting Enzyme-2 (ACE-2) plays a major role in the transmission and severity of the disease. To understand the binding affinity of the wild and mutant SARS-CoV-2 S with ACE2, molecular modeling studies were carried out. We discovered that the wild SARS-CoV-2 S RBD-ACE-2 complex has a high binding affinity and stability than that of the mutant. The N440K strain escapes from antibody neutralization, which might increase reinfection and decrease vaccine efficiency. To find a potential inhibitor against mutant N440K SARS-CoV-2, a virtual screening process was carried out and found ZINC169293961, ZINC409421825 and ZINC22060839 as the best binding energy compounds. Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Malathi Kullappan
- Department of Research, Panimalar Medical College Hospital & Research Institute, Varadharajapuram, Poonamallee, Chennai, India
| | - Usha Mary
- Department of Chemistry, Panimalar Engineering College, Varadharajapuram, Poonamallee, Chennai, India
| | - Jenifer M Ambrose
- Department of Research, Panimalar Medical College Hospital & Research Institute, Varadharajapuram, Poonamallee, Chennai, India
| | - Vishnu Priya Veeraraghavan
- Department of Biochemistry, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences (SIMATS), Saveetha University, Velappanchavadi, Chennai, Tamil Nadu, India
| | - Krishna Mohan Surapaneni
- Department of Research, Panimalar Medical College Hospital & Research Institute, Varadharajapuram, Poonamallee, Chennai, India.,Department of Biochemistry, Panimalar Medical College Hospital & Research Institute, Varadharajapuram, Poonamallee, Chennai, India.,Department of Molecular Virology, Clinical Skills & Simulation, Research, Panimalar Medical College Hospital & Research Institute, Varadharajapuram, Poonamallee, Chennai, India.,Department of Clinical Skills & Simulation, Panimalar Medical College Hospital & Research Institute, Varadharajapuram, Poonamallee, Chennai, India
| |
Collapse
|
39
|
Wang K, Chen D, Yu B, He J, Mao X, Huang Z, Yan H, Wu A, Luo Y, Zheng P, Yu J, Luo J. Eugenol Alleviates TGEV-Induced Intestinal Injury via Suppressing ROS/NLRP3/GSDMD-Dependent Pyroptosis. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:1477-1487. [PMID: 36642968 DOI: 10.1021/acs.jafc.2c05833] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Transmissible gastroenteritis virus (TGEV), a coronavirus, is one of the main causative agents of diarrhea in piglets and significantly impacts the global swine industry. Pyroptosis is involved in the pathogenesis of coronavirus, but its role in TGEV-induced intestinal injury has yet to be fully elucidated. Eugenol, an essential plant oil, plays a vital role in antiviral innate immune responses. We demonstrate the preventive effect of eugenol on TGEV infection. Eugenol alleviates TGEV-induced intestinal epithelial cell pyroptosis and reduces intestinal injury in TGEV-infected piglets. Mechanistically, eugenol reduces the activation of NLRP3 inflammasome, thereby inhibiting TGEV-induced intestinal epithelial cell pyroptosis. In addition, eugenol scavenges TGEV-induced reactive oxygen species (ROS) increase, which in turn prevents TGEV-induced NLRP3 inflammasome activation and pyroptosis. Overall, eugenol protects the intestine by reducing TGEV-induced pyroptosis through inhibition of NLRP3 inflammasome activation, which may be mediated through intracellular ROS levels. These findings propose that eugenol may be an effective strategy to prevent TGEV infection.
Collapse
Affiliation(s)
- Kang Wang
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan Province 611130, People's Republic of China
- Key Laboratory of Animal Disease-resistant Nutrition, Chengdu, Sichuan Province 611130, People's Republic of China
| | - Daiwen Chen
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan Province 611130, People's Republic of China
- Key Laboratory of Animal Disease-resistant Nutrition, Chengdu, Sichuan Province 611130, People's Republic of China
| | - Bing Yu
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan Province 611130, People's Republic of China
- Key Laboratory of Animal Disease-resistant Nutrition, Chengdu, Sichuan Province 611130, People's Republic of China
| | - Jun He
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan Province 611130, People's Republic of China
- Key Laboratory of Animal Disease-resistant Nutrition, Chengdu, Sichuan Province 611130, People's Republic of China
| | - Xiangbing Mao
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan Province 611130, People's Republic of China
- Key Laboratory of Animal Disease-resistant Nutrition, Chengdu, Sichuan Province 611130, People's Republic of China
| | - Zhiqing Huang
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan Province 611130, People's Republic of China
- Key Laboratory of Animal Disease-resistant Nutrition, Chengdu, Sichuan Province 611130, People's Republic of China
| | - Hui Yan
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan Province 611130, People's Republic of China
- Key Laboratory of Animal Disease-resistant Nutrition, Chengdu, Sichuan Province 611130, People's Republic of China
| | - Aimin Wu
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan Province 611130, People's Republic of China
- Key Laboratory of Animal Disease-resistant Nutrition, Chengdu, Sichuan Province 611130, People's Republic of China
| | - Yuheng Luo
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan Province 611130, People's Republic of China
- Key Laboratory of Animal Disease-resistant Nutrition, Chengdu, Sichuan Province 611130, People's Republic of China
| | - Ping Zheng
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan Province 611130, People's Republic of China
- Key Laboratory of Animal Disease-resistant Nutrition, Chengdu, Sichuan Province 611130, People's Republic of China
| | - Jie Yu
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan Province 611130, People's Republic of China
- Key Laboratory of Animal Disease-resistant Nutrition, Chengdu, Sichuan Province 611130, People's Republic of China
| | - Junqiu Luo
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan Province 611130, People's Republic of China
- Key Laboratory of Animal Disease-resistant Nutrition, Chengdu, Sichuan Province 611130, People's Republic of China
| |
Collapse
|
40
|
Elsebai MF, Habib ESE. Blood pH and COVID-19. Arch Pharm (Weinheim) 2023; 356:e2200558. [PMID: 36690587 DOI: 10.1002/ardp.202200558] [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: 10/24/2022] [Revised: 12/31/2022] [Accepted: 01/02/2023] [Indexed: 01/25/2023]
Abstract
The coronavirus disease 2019 (COVID-19) pandemic is a worldwide war. Raising the blood pH might be a crucial strategy to chase COVID-19. The human blood is slightly alkaline, which is essential for cell metabolism, normal physiology, and balanced immunity since all of these biological processes are pH-dependent. Varieties of physiologic derangements occur when the blood pH is disrupted. Severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) proliferates in acidic blood that magnifies the severity of COVID-19. On the other side, blood acidemia is linked to increased morbidity and mortality because of its complications on immunity, especially in the elderly and in critical diseases such as cancer, musculoskeletal degradation, renal, cardiac, and pulmonary disorders, which result in many pathological disorders such as osteomalacia, and disturbing the hematopoiesis. Additionally, acidemia of the blood facilitates viral infection and progression. Thus, correcting the acid-base balance might be a crucial strategy for the treatment of COVID-19, which might be attributed to the distraction of the viral spike protein to its cognate receptor angiotensin-converting enzyme 2 and supporting the over-taxed immunity.
Collapse
Affiliation(s)
- Mahmoud Fahmi Elsebai
- Department of Pharmacognosy, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| | - El-Sayed E Habib
- Department of Microbiology and Immunology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| |
Collapse
|
41
|
Li J, Lin H, Fan T, Huang L, Zhang X, Tai Y, Fang Y, Li Q, Zhao R, Wang P, Zhou L, Wan L, Wu Y, Zhong H, Wei C, Yang X. BPOZ-2 is a negative regulator of the NLPR3 inflammasome contributing to SARS-CoV-2-induced hyperinflammation. Front Cell Infect Microbiol 2023; 13:1134511. [PMID: 36936774 PMCID: PMC10019892 DOI: 10.3389/fcimb.2023.1134511] [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: 12/30/2022] [Accepted: 02/09/2023] [Indexed: 03/05/2023] Open
Abstract
Introduction Inflammation play important roles in the initiation and progression of acute lung injury (ALI), acute respiratory distress syndrome (ARDS), septic shock, clotting dysfunction, or even death associated with SARS-CoV-2 infection. However, the pathogenic mechanisms underlying SARS-CoV-2-induced hyperinflammation are still largely unknown. Methods The animal model of septic shock and ALI was established after LPS intraperitoneal injection or intratracheal instillation. Bone marrow-derived macrophages (BMDMs) from WT and BPOZ-2 KO mouse strains were harvested from the femurs and tibias of mice. Immunohistology staining, ELISA assay, coimmunoprecipitation, and immunoblot analysis were used to detect the histopathological changes of lung tissues and the expression of inflammatory factors and protein interaction. Results and conclusions We show a distinct mechanism by which the SARS-CoV-2 N (SARS-2-N) protein targets Bood POZ-containing gene type 2 (BPOZ-2), a scaffold protein for the E3 ubiquitin ligase Cullin 3 that we identified as a negative regulator of inflammatory responses, to promote NLRP3 inflammasome activation. We first demonstrated that BPOZ-2 knockout (BPOZ-2 KO) mice were more susceptible to lipopolysaccharide (LPS)-induced septic shock and ALI and showed increased serum IL-1β levels. In addition, BMDMs isolated from BPOZ-2 KO mice showed increased IL-1β production in response to NLRP3 stimuli. Mechanistically, BPOZ-2 interacted with NLRP3 and mediated its degradation by recruiting Cullin 3. In particular, the expression of BPOZ-2 was significantly reduced in lung tissues from mice infected with SARS-CoV-2 and in cells overexpressing SARS-2-N. Importantly, proinflammatory responses triggered by the SARS-2-N were significantly blocked by BPOZ-2 reintroduction. Thus, we concluded that BPOZ-2 is a negative regulator of the NLPR3 inflammasome that likely contributes to SARS-CoV-2-induced hyperinflammation.
Collapse
Affiliation(s)
- Jingfei Li
- Department of Genetic Engineering, Beijing Institute of Biotechnology, Beijing, China
| | - Haotian Lin
- Department of Genetic Engineering, Beijing Institute of Biotechnology, Beijing, China
| | - Tinghui Fan
- Department of Genetic Engineering, Beijing Institute of Biotechnology, Beijing, China
| | - Linfei Huang
- Department of Genetic Engineering, Beijing Institute of Biotechnology, Beijing, China
| | - Xinyong Zhang
- Department of Medical Oncology, Beijing Tuberculosis and Thoracic Tumor Research Institute/Beijing Chest Hospital, Capital Medical University, Beijing, China
| | - Yanhong Tai
- Department of Pathology, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Yi Fang
- Department of Endocrinology, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Qihong Li
- Department of Stomatology, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Ruzhou Zhao
- Department of Genetic Engineering, Beijing Institute of Biotechnology, Beijing, China
| | - Penghao Wang
- Department of Genetic Engineering, Beijing Institute of Biotechnology, Beijing, China
| | - Li Zhou
- Department of Genetic Engineering, Beijing Institute of Biotechnology, Beijing, China
| | - Luming Wan
- Department of Genetic Engineering, Beijing Institute of Biotechnology, Beijing, China
| | - Yuhua Wu
- Department of Medical Oncology, Beijing Tuberculosis and Thoracic Tumor Research Institute/Beijing Chest Hospital, Capital Medical University, Beijing, China
| | - Hui Zhong
- Department of Genetic Engineering, Beijing Institute of Biotechnology, Beijing, China
| | - Congwen Wei
- Department of Genetic Engineering, Beijing Institute of Biotechnology, Beijing, China
| | - Xiaopan Yang
- Department of Genetic Engineering, Beijing Institute of Biotechnology, Beijing, China
- *Correspondence: Xiaopan Yang,
| |
Collapse
|
42
|
Papadopoulos KI, Papadopoulou A, Aw TC. A protective erythropoietin evolutionary landscape, NLRP3 inflammasome regulation, and multisystem inflammatory syndrome in children. Hum Cell 2023; 36:26-40. [PMID: 36310304 PMCID: PMC9618415 DOI: 10.1007/s13577-022-00819-w] [Citation(s) in RCA: 4] [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: 05/18/2022] [Accepted: 10/24/2022] [Indexed: 11/04/2022]
Abstract
The low incidence of pediatric severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) infection and the associated multisystem inflammatory syndrome (MIS-C) lack a unifying pathophysiological explanation, impeding effective prevention and therapy. Activation of the NACHT, LRR, and PYD domains-containing protein (NLRP) 3 inflammasome in SARS-CoV-2 with perturbed regulation in MIS-C, has been reported. We posit that, early age physiological states and genetic determinants, such as certain polymorphisms of renin-angiotensin aldosterone system (RAAS) molecules, promote a controlled RAAS hyperactive state, and form an evolutionary landscape involving an age-dependent erythropoietin (EPO) elevation, mediating ancestral innate immune defenses that, through appropriate NLRP3 regulation, mitigate tissue injury and pathogen invasion. SARS-CoV-2-induced downregulation of angiotensin-converting enzyme (ACE)2 expression in endothelial cells (EC), impairment of endothelial nitric oxide (NO) synthase (eNOS) activity and downstream NO bioavailability, may promote a hyperactive RAAS with elevated angiotensin II and aldosterone that, can trigger, and accelerate NLRP3 inflammasome activation, while EPO-eNOS/NO abrogate it. Young age and a protective EPO evolutionary landscape may successfully inhibit SARS-CoV-2 and contain NLRP3 inflammasome activation. By contrast, increasing age and falling EPO levels, in genetically susceptible children with adverse genetic variants and co-morbidities, may lead to unopposed RAAS hyperactivity, NLRP3 inflammasome dysregulation, severe endotheliitis with pyroptotic cytokine storm, and development of autoantibodies, as already described in MIS-C. Our haplotype estimates, predicted from allele frequencies in population databases, are in concordance with MIS-C incidence reports in Europeans but indicate lower risks for Asians and African Americans. Targeted Mendelian approaches dissecting the influence of relevant genetic variants are needed.
Collapse
Affiliation(s)
- Konstantinos I Papadopoulos
- Department of Research and Development, THAI StemLife Co., Ltd., 566/3 THAI StemLife Bldg., Soi Ramkhamhaeng 39 (Thepleela 1), Prachaouthit Rd., Wangthonglang, 10310, Bangkok, Thailand.
| | - Alexandra Papadopoulou
- Occupational and Environmental Health Services, Feelgood Lund, Ideon Science Park, Scheelevägen 17, 223 63, Lund, Sweden
| | - Tar-Choon Aw
- Department of Laboratory Medicine, Changi General Hospital, Singapore, 529889, Singapore
- Department of Medicine, National University of Singapore, Singapore, 119228, Singapore
| |
Collapse
|
43
|
Tong X, Ping H, Gong X, Zhang K, Chen Z, Cai C, Lu Z, Yang R, Gao S, Wang Y, Wang X, Liu L, Ke H. Pyroptosis in the lung and spleen of patients died from
COVID-19. EUR J INFLAMM 2022; 20:1721727X221140661. [PMCID: PMC9702972 DOI: 10.1177/1721727x221140661] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2024] Open
Abstract
The purpose of this study was to investigate the expression of pyroptosis-related factors (NLRP3, IL-18, NF-κB, HMGB-1, and GSDMD) in patients who died of COVID-19. The expression levels of NLRP3, IL-18, NF-κB, HMGB-1, and GSDMD in lung and spleen tissues of the COVID-19 group and the control group were detected by tissue immunofluorescence. The control group includes lung tissues and spleen tissues of two patients who died unexpectedly without SARS-CoV-2 infection, and the COVID-19 group includes the lung and spleen tissues of three patients who died of SARS-CoV-2 virus infection. The positive rates of NF-κB, NLRP3, IL-18, and GSDMD in the lung tissues from the control group and COVID-19 group were 9.8% vs 73.4% (p = 0.000), 5.5% vs 63.6% (p = 0.000), 24.4% vs 76.2% (p = 0.000), and 17.5% and 46.8% (p = 0.000) respectively. The positive rates of NF-κB, NLRP3, IL-18, HMGB-1, and GSDMD in the spleen tissues from the control group and COVID-19 group were 20.6% vs 71.2% (p = 0.000), 18.9% vs 72.0% (p = 0.000), 15.2% vs 64.8% (p = 0.000), 27.6% vs 69.2% (p = 0.000), and 23% and 48.8% (p = 0.000), respectively. The positive rates of SARS-CoV-2 spike protein in the CD68 positive cells of the lung and spleen in the control group and COVID-19 group were 2.5% vs 56.8% (p = 0.000); 3.0% vs 64.9% (p = 0.000) respectively. The rates of NF-κB positive nuclei in the control group and COVID-19 group were 13.4% vs 51.4% (p = 0.000) in the lung and 38.2% vs 59.3% (p = 0.000) in the spleen. The rates of HMGB-1 positive cytoplasm in the control and the COVID-19 group were 19.7% vs 50.3% (p = 0.000) in the lung and 12.3% vs 45.2% (p = 0.000) in the spleen. The targets of SARS-CoV-2 are the lung and spleen, where increased macrophages could be involved in the up-regulation of pyroptosis-related inflammatory factors such as NF-κB, HMGB-1, NLRP3, IL-18, and GSDMD.
Collapse
Affiliation(s)
- Xin Tong
- Training Center of AIDS prevention
and Cure of Hubei Province, Zhongnan Hospital Wuhan
University, Wuhan, China
| | - Haiqin Ping
- Training Center of AIDS prevention
and Cure of Hubei Province, Zhongnan Hospital Wuhan
University, Wuhan, China
| | - Xiaoming Gong
- Training Center of AIDS prevention
and Cure of Hubei Province, Zhongnan Hospital Wuhan
University, Wuhan, China
| | - Kai Zhang
- Training Center of AIDS prevention
and Cure of Hubei Province, Zhongnan Hospital Wuhan
University, Wuhan, China
| | - Zhaojun Chen
- Training Center of AIDS prevention
and Cure of Hubei Province, Zhongnan Hospital Wuhan
University, Wuhan, China
| | - Caiyun Cai
- Training Center of AIDS prevention
and Cure of Hubei Province, Zhongnan Hospital Wuhan
University, Wuhan, China
| | - Zhiyan Lu
- Department of Radiology, Zhongnan Hospital of Wuhan
University, Wuhan, China
| | - Rongrong Yang
- Department of Radiology, Zhongnan Hospital of Wuhan
University, Wuhan, China
| | - Shicheng Gao
- Department of infectious disease, Zhongnan Hospital of Wuhan
University, Wuhan, China
| | - Yunyun Wang
- Department of Forensic Medicine,
Tongji Medical College, Huazhong University of Science and
Technology, Wuhan, China
| | - Xinghuan Wang
- Department of Urology, Zhongnan Hospital of Wuhan
University, Wuhan, China
| | - Liang Liu
- Department of Forensic Medicine,
Tongji Medical College, Huazhong University of Science and
Technology, Wuhan, China
| | - Hengning Ke
- Training Center of AIDS prevention
and Cure of Hubei Province, Zhongnan Hospital Wuhan
University, Wuhan, China
| |
Collapse
|
44
|
Forsyth CB, Voigt RM, Swanson GR, Bishehsari F, Shaikh M, Zhang L, Engen P, Keshavarzian A. Alcohol use disorder as a potential risk factor for COVID-19 severity: A narrative review. Alcohol Clin Exp Res 2022; 46:1930-1943. [PMID: 36394508 PMCID: PMC9722573 DOI: 10.1111/acer.14936] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 07/25/2022] [Accepted: 08/31/2022] [Indexed: 11/19/2022]
Abstract
In Dec. 2019-January 2020, a pneumonia illness originating in Wuhan, China, designated as coronavirus disease 2019 (COVID-19) was shown to be caused by a novel RNA coronavirus designated as severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). People with advanced age, male sex, and/or underlying health conditions (obesity, type 2 diabetes, cardiovascular disease, hypertension, chronic kidney disease, and chronic lung disease) are especially vulnerable to severe COVID-19 symptoms and death. These risk factors impact the immune system and are also associated with poor health, chronic illness, and shortened longevity. However, a large percent of patients without these known risk factors also develops severe COVID-19 disease that can result in death. Thus, there must exist risk factors that promote exaggerated inflammatory and immune response to the SARS-CoV-2 virus leading to death. One such risk factor may be alcohol misuse and alcohol use disorder because these can exacerbate viral lung infections like SARS, influenza, and pneumonia. Thus, it is highly plausible that alcohol misuse is a risk factor for either increased infection rate when individuals are exposed to SARS-CoV-2 virus and/or more severe COVID-19 in infected patients. Alcohol use is a well-known risk factor for lung diseases and ARDS in SARS patients. We propose that alcohol has three key pathogenic elements in common with other COVID-19 severity risk factors: namely, inflammatory microbiota dysbiosis, leaky gut, and systemic activation of the NLRP3 inflammasome. We also propose that these three elements represent targets for therapy for severe COVID-19.
Collapse
Affiliation(s)
- Christopher B. Forsyth
- Department of Internal Medicine, Section of Gastroenterology, Rush University Medical Center, Chicago, IL 60612
- Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, IL 60612
- Department of Anatomy and Cell Biology, Rush University Medical Center, Chicago, IL 60612
- Rush University Graduate College, Rush University Medical Center, Chicago, IL 60612
| | - Robin M. Voigt
- Department of Internal Medicine, Section of Gastroenterology, Rush University Medical Center, Chicago, IL 60612
- Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, IL 60612
- Department of Anatomy and Cell Biology, Rush University Medical Center, Chicago, IL 60612
- Rush University Graduate College, Rush University Medical Center, Chicago, IL 60612
| | - Garth R. Swanson
- Department of Internal Medicine, Section of Gastroenterology, Rush University Medical Center, Chicago, IL 60612
- Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, IL 60612
- Department of Anatomy and Cell Biology, Rush University Medical Center, Chicago, IL 60612
- Rush University Graduate College, Rush University Medical Center, Chicago, IL 60612
| | - Faraz Bishehsari
- Department of Internal Medicine, Section of Gastroenterology, Rush University Medical Center, Chicago, IL 60612
- Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, IL 60612
- Department of Anatomy and Cell Biology, Rush University Medical Center, Chicago, IL 60612
- Rush University Graduate College, Rush University Medical Center, Chicago, IL 60612
| | - Maliha Shaikh
- Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, IL 60612
| | - Lijuan Zhang
- Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, IL 60612
| | - Phillip Engen
- Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, IL 60612
| | - Ali Keshavarzian
- Department of Internal Medicine, Section of Gastroenterology, Rush University Medical Center, Chicago, IL 60612
- Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, IL 60612
- Department of Anatomy and Cell Biology, Rush University Medical Center, Chicago, IL 60612
- Rush University Graduate College, Rush University Medical Center, Chicago, IL 60612
| |
Collapse
|
45
|
Baena Carstens L, Campos D’amico R, Fernandes de Moura K, Morais de Castro E, Centenaro F, Silva Barbosa G, Vieira Cavalcante da Silva G, Brenny I, Honório D’Agostini JC, Hlatchuk EC, Pissette de Lima S, Camargo Martins AP, De Castro Deus M, Konzen Klein C, Kubaski Benevides AP, Nagashima S, Machado-Souza C, Pinho RA, Pellegrino Baena C, de Noronha L. Lung Inflammasome Activation in SARS-CoV-2 Post-Mortem Biopsies. Int J Mol Sci 2022; 23:ijms232113033. [PMID: 36361818 PMCID: PMC9659061 DOI: 10.3390/ijms232113033] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 10/07/2022] [Accepted: 10/10/2022] [Indexed: 11/16/2022] Open
Abstract
The inflammasome complex is a key part of chronic diseases and acute infections, being responsible for cytokine release and cell death mechanism regulation. The SARS-CoV-2 infection is characterized by a dysregulated cytokine release. In this context, the inflammasome complex analysis within SARS-CoV-2 infection may prove beneficial to understand the disease’s mechanisms. Post-mortem minimally invasive autopsies were performed in patients who died from COVID-19 (n = 24), and lung samples were compared to a patient control group (n = 11) and an Influenza A virus H1N1 subtype group from the 2009 pandemics (n = 10). Histological analysis was performed using hematoxylin-eosin staining. Immunohistochemical (IHC) staining was performed using monoclonal antibodies against targets: ACE2, TLR4, NF-κB, NLRP-3 (or NALP), IL-1β, IL-18, ASC, CASP1, CASP9, GSDMD, NOX4, TNF-α. Data obtained from digital analysis underwent appropriate statistical tests. IHC analysis showed biomarkers that indicate inflammasome activation (ACE2; NF-κB; NOX4; ASC) were significantly increased in the COVID-19 group (p < 0.05 for all) and biomarkers that indicate cell pyroptosis and inflammasome derived cytokines such as IL-18 (p < 0.005) and CASP1 were greatly increased (p < 0.0001) even when compared to the H1N1 group. We propose that the SARS-CoV-2 pathogenesis is connected to the inflammasome complex activation. Further studies are still warranted to elucidate the pathophysiology of the disease.
Collapse
Affiliation(s)
- Lucas Baena Carstens
- Laboratory of Experimental Pathology, School of Medicine and Life Sciences, Pontifícia Universidade Católica do Paraná (PUCPR), R. Imaculada Conceição, 1155-Prado Velho, Curitiba 80215-901, PR, Brazil
| | - Raissa Campos D’amico
- Postgraduate Program of Health Sciences, School of Medicine and Life Sciences, Pontifícia Universidade Católica do Paraná (PUCPR), R. Imaculada Conceição, 1155-Prado Velho, Curitiba 80215-901, PR, Brazil
- Hospital Marcelino Champagnat, Av. Presidente Affonso Camargo, 1399-Cristo Rei, Curitiba 80050-370, PR, Brazil
| | - Karen Fernandes de Moura
- Postgraduate Program of Health Sciences, School of Medicine and Life Sciences, Pontifícia Universidade Católica do Paraná (PUCPR), R. Imaculada Conceição, 1155-Prado Velho, Curitiba 80215-901, PR, Brazil
- Hospital Marcelino Champagnat, Av. Presidente Affonso Camargo, 1399-Cristo Rei, Curitiba 80050-370, PR, Brazil
| | - Eduardo Morais de Castro
- Postgraduate in Biotechnology Applied in Health of Children and Adolescent, Faculdades Pequeno Príncipe (FPP), Instituto de Pesquisa Pelé Pequeno Príncipe (IPPPP), R. Silva Jardim, 1632-Água Verde, Curitiba 80230-020, PR, Brazil
| | - Flávia Centenaro
- Laboratory of Experimental Pathology, School of Medicine and Life Sciences, Pontifícia Universidade Católica do Paraná (PUCPR), R. Imaculada Conceição, 1155-Prado Velho, Curitiba 80215-901, PR, Brazil
| | - Giovanna Silva Barbosa
- Laboratory of Experimental Pathology, School of Medicine and Life Sciences, Pontifícia Universidade Católica do Paraná (PUCPR), R. Imaculada Conceição, 1155-Prado Velho, Curitiba 80215-901, PR, Brazil
| | - Guilherme Vieira Cavalcante da Silva
- Laboratory of Experimental Pathology, School of Medicine and Life Sciences, Pontifícia Universidade Católica do Paraná (PUCPR), R. Imaculada Conceição, 1155-Prado Velho, Curitiba 80215-901, PR, Brazil
| | - Isadora Brenny
- Laboratory of Experimental Pathology, School of Medicine and Life Sciences, Pontifícia Universidade Católica do Paraná (PUCPR), R. Imaculada Conceição, 1155-Prado Velho, Curitiba 80215-901, PR, Brazil
| | - Júlio César Honório D’Agostini
- Departmnet of Medical Pathology, Universidade Federal do Paraná (UFPR), Rua General Carneiro, 181-Alto da Glória, Curitiba 80215-901, PR, Brazil
| | - Elisa Carolina Hlatchuk
- Departmnet of Medical Pathology, Universidade Federal do Paraná (UFPR), Rua General Carneiro, 181-Alto da Glória, Curitiba 80215-901, PR, Brazil
| | - Sabrina Pissette de Lima
- Laboratory of Experimental Pathology, School of Medicine and Life Sciences, Pontifícia Universidade Católica do Paraná (PUCPR), R. Imaculada Conceição, 1155-Prado Velho, Curitiba 80215-901, PR, Brazil
| | - Ana Paula Camargo Martins
- Laboratory of Experimental Pathology, School of Medicine and Life Sciences, Pontifícia Universidade Católica do Paraná (PUCPR), R. Imaculada Conceição, 1155-Prado Velho, Curitiba 80215-901, PR, Brazil
| | - Marina De Castro Deus
- Postgraduate Program of Health Sciences, School of Medicine and Life Sciences, Pontifícia Universidade Católica do Paraná (PUCPR), R. Imaculada Conceição, 1155-Prado Velho, Curitiba 80215-901, PR, Brazil
| | - Carolline Konzen Klein
- Laboratory of Experimental Pathology, School of Medicine and Life Sciences, Pontifícia Universidade Católica do Paraná (PUCPR), R. Imaculada Conceição, 1155-Prado Velho, Curitiba 80215-901, PR, Brazil
| | - Ana Paula Kubaski Benevides
- Laboratory of Experimental Pathology, School of Medicine and Life Sciences, Pontifícia Universidade Católica do Paraná (PUCPR), R. Imaculada Conceição, 1155-Prado Velho, Curitiba 80215-901, PR, Brazil
| | - Seigo Nagashima
- Laboratory of Experimental Pathology, School of Medicine and Life Sciences, Pontifícia Universidade Católica do Paraná (PUCPR), R. Imaculada Conceição, 1155-Prado Velho, Curitiba 80215-901, PR, Brazil
- Postgraduate Program of Health Sciences, School of Medicine and Life Sciences, Pontifícia Universidade Católica do Paraná (PUCPR), R. Imaculada Conceição, 1155-Prado Velho, Curitiba 80215-901, PR, Brazil
- Correspondence: (S.N.); (L.d.N.); Tel.: +55-(41)-99942-7191 (S.N.); Tel.: +55-(41)-999994769 (L.d.N.)
| | - Cleber Machado-Souza
- Postgraduate in Biotechnology Applied in Health of Children and Adolescent, Faculdades Pequeno Príncipe (FPP), Instituto de Pesquisa Pelé Pequeno Príncipe (IPPPP), R. Silva Jardim, 1632-Água Verde, Curitiba 80230-020, PR, Brazil
| | - Ricardo A Pinho
- Postgraduate Program of Health Sciences, School of Medicine and Life Sciences, Pontifícia Universidade Católica do Paraná (PUCPR), R. Imaculada Conceição, 1155-Prado Velho, Curitiba 80215-901, PR, Brazil
| | - Cristina Pellegrino Baena
- Postgraduate Program of Health Sciences, School of Medicine and Life Sciences, Pontifícia Universidade Católica do Paraná (PUCPR), R. Imaculada Conceição, 1155-Prado Velho, Curitiba 80215-901, PR, Brazil
- Hospital Marcelino Champagnat, Av. Presidente Affonso Camargo, 1399-Cristo Rei, Curitiba 80050-370, PR, Brazil
| | - Lúcia de Noronha
- Laboratory of Experimental Pathology, School of Medicine and Life Sciences, Pontifícia Universidade Católica do Paraná (PUCPR), R. Imaculada Conceição, 1155-Prado Velho, Curitiba 80215-901, PR, Brazil
- Postgraduate Program of Health Sciences, School of Medicine and Life Sciences, Pontifícia Universidade Católica do Paraná (PUCPR), R. Imaculada Conceição, 1155-Prado Velho, Curitiba 80215-901, PR, Brazil
- Correspondence: (S.N.); (L.d.N.); Tel.: +55-(41)-99942-7191 (S.N.); Tel.: +55-(41)-999994769 (L.d.N.)
| |
Collapse
|
46
|
Wei T, Zhang C, Song Y. Molecular mechanisms and roles of pyroptosis in acute lung injury. Chin Med J (Engl) 2022; 135:2417-2426. [PMID: 36583860 PMCID: PMC9945565 DOI: 10.1097/cm9.0000000000002425] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Indexed: 12/31/2022] Open
Abstract
ABSTRACT Acute lung injury (ALI) and acute respiratory distress syndrome (ARDS), which are characterized by excessive inflammation and accompanied by diffuse injury of alveoli, can result in severe respiratory failures. The morbidity and mortality of patients remain high because the major treatments for ALI/ARDS are mainly supportive due to the lack of effective therapies. Numerous studies have demonstrated that the aggravation of coronavirus disease 2019 (COVID-19) leads to severe pneumonia and even ARDS. Pyroptosis, a biological process identified as a type of programed cell death, is mainly triggered by inflammatory caspase activation and is directly meditated by the gasdermin protein family, as well as being associated with the secretion and release of pro-inflammatory cytokines. Clinical and experimental evidence corroborates that pyroptosis of various cells in the lung, such as immune cells and structural cells, may play an important role in the pathogenesis of "cytokine storms" in ALI/ARDS, including those induced by COVID-19. Here, with a focus on ALI/ARDS and COVID-19, we summarized the recent advances in this field and proposed the theory of an inflammatory cascade in pyroptosis to identify new targets and pave the way for new approaches to treat these diseases.
Collapse
Affiliation(s)
- Tianchang Wei
- Department of Pulmonary Medicine, Shanghai Key Laboratory of Lung Inflammation and Injury, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Cuiping Zhang
- Department of Pulmonary Medicine, Shanghai Key Laboratory of Lung Inflammation and Injury, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Yuanlin Song
- Department of Pulmonary Medicine, Shanghai Key Laboratory of Lung Inflammation and Injury, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Shanghai Institute of Infectious Disease and Biosecurity, Shanghai 200032, China
- Shanghai Respiratory Research Institute, Shanghai 200032, China
- Jinshan Hospital of Fudan University, Shanghai 201508, China
| |
Collapse
|
47
|
Demoliou C, Papaneophytou C, Nicolaidou V. SARS-CoV-2 and HIV-1: So Different yet so Alike . Immune Response at the Cellular and Molecular Level. Int J Med Sci 2022; 19:1787-1795. [PMID: 36313221 PMCID: PMC9608044 DOI: 10.7150/ijms.73134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 09/07/2022] [Indexed: 01/13/2023] Open
Abstract
In the past half century, humanity has experienced two devastating pandemics; the HIV-1 pandemic and the recent pandemic caused by SARS-CoV-2. Both emerged as zoonotic pathogens. Interestingly, SARS-CoV-2 has rapidly migrated all over the world in less than two years, much as HIV-1 did almost 40 years ago. Despite these two RNA viruses being different in their mode of transmission as well as the symptoms they generate, recent evidence suggests that they cause similar immune responses. In this mini review, we compare the molecular basis for CD4+ T cell lymphopenia and other effects on the immune system induced by SARS-CoV-2 and HIV-1 infections. We considered features of the host immune response that are shared with HIV-1 and could account for the lymphopenia and other immune effects observed in COVID-19. The information provided herein, may cast the virus-induced lymphopenia and cytokine storm associated with the acute SARS-CoV-2 infection and pathogenesis in a different light for further research on host immune responses. It can also provide opportunities for the identification of novel therapeutic targets for COVID-19. Furthermore, we provide some basic information to enable a comparative framework for considering the overlapping sets of immune responses caused by HIV-1 and SARS-CoV-2.
Collapse
Affiliation(s)
| | | | - Vicky Nicolaidou
- Department of Life and Health Sciences, School of Sciences and Engineering, University of Nicosia, 46 Makedonitissas Avenue, 2417, Nicosia, Cyprus
| |
Collapse
|
48
|
Muacevic A, Adler JR. The Impact of Angiotensin Converting Enzyme-2 (ACE-2) on Bone Remodeling Marker Osteoprotegerin (OPG) in Post-COVID-19 Iraqi Patients. Cureus 2022; 14:e29926. [PMID: 36348825 PMCID: PMC9633432 DOI: 10.7759/cureus.29926] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/04/2022] [Indexed: 01/25/2023] Open
Abstract
Introduction As COVID-19 affects human genes in several types of peripheral tissue, numerous disorders occur after recovery. The virus enters host cells via angiotensin-converting enzyme-2 (ACE-2) receptors that affect bone remodeling, leading to osteopenia or osteoporosis, which is characterized by low bone mineral density (BMD). The adult skeleton undergoes about 10% remodeling annually, which is crucial for preventing fatigue damage and preserving calcium homeostasis. An imbalance in the rates of bone production and resorption causes bone loss. Osteoprotegerin (OPG) is one of the regulators involved in the bone remodeling mechanism, it decreases the activity of NF-B receptors that activates the receptor activators of NF-B ligand (RANKL) pathway, which maintains the bone homeostasis balance. This study aims to find out the disruption of bone homeostasis balance in Iraqi post-COVID-19 patients. Materials and methods It is a case-control study that includes 130 Iraqi subjects enrolled. They were divided into two groups - the first group consisted of 80 post-COVID-19 infection patients, while the second group consisted of 50 who were not infected with COVID-19. Also, the levels of ACE-2 and OPG were measured in the serum by the ELISA technique. The BMD was measured by the DEXA scan technique. Results This study found that there is an effect of coronavirus infection on the bone strength measured by the Mean of the OPG level, which was found to be highly significant in the serum of post-COVID-19 patients when compared with non-COVID-19 subjects (P-value = 0.001), but the Mean of ACE-2 level was statistically non-significant between the two groups (P-value = 0.13). Also, the BMD of post-COVID-19 patients that was measured by DEXA scan had a statistically highly significant T-score% between the two groups. Conclusion The current study found that there was an effect of COVID-19 on the bone remodeling mechanism, which may be causing osteopenia or osteoporosis for Iraqi subjects enrolled in the current study. Also, analyzing the OPG level in the serum could be helpful in predicting low BMD.
Collapse
|
49
|
Anwar MM, Sah R, Shrestha S, Ozaki A, Roy N, Fathah Z, Rodriguez-Morales AJ. Disengaging the COVID-19 Clutch as a Discerning Eye Over the Inflammatory Circuit During SARS-CoV-2 Infection. Inflammation 2022; 45:1875-1894. [PMID: 35639261 PMCID: PMC9153229 DOI: 10.1007/s10753-022-01674-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 03/29/2022] [Accepted: 04/18/2022] [Indexed: 01/08/2023]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) causes the cytokine release syndrome (CRS) and leads to multiorgan dysfunction. Mitochondrial dynamics are fundamental to protect against environmental insults, but they are highly susceptible to viral infections. Defective mitochondria are potential sources of reactive oxygen species (ROS). Infection with SARS-CoV-2 damages mitochondria, alters autophagy, reduces nitric oxide (NO), and increases both nicotinamide adenine dinucleotide phosphate oxidases (NOX) and ROS. Patients with coronavirus disease 2019 (COVID-19) exhibited activated toll-like receptors (TLRs) and the Nucleotide-binding and oligomerization domain (NOD-), leucine-rich repeat (LRR-), pyrin domain-containing protein 3 (NLRP3) inflammasome. The activation of TLRs and NLRP3 by SARS-CoV-2 induces interleukin 6 (IL-6), IL-1β, IL-18, and lactate dehydrogenase (LDH). Herein, we outline the inflammatory circuit of COVID-19 and what occurs behind the scene, the interplay of NOX/ROS and their role in hypoxia and thrombosis, and the important role of ROS scavengers to reduce COVID-19-related inflammation.
Collapse
Affiliation(s)
- Mohammed Moustapha Anwar
- Department of Biotechnology, Institute of Graduate Studies and Research (IGSR), Alexandria University, Alexandria, Egypt.
| | - Ranjit Sah
- Tribhuvan University Institute of Medicine, Kathmandu, Nepal
| | - Sunil Shrestha
- Department of Pharmaceutical and Health Service Research, Nepal Health Research and Innovation Foundation, Lalitpur, Nepal
| | - Akihiko Ozaki
- Department of Breast Surgery, Jyoban Hospital of Tokiwa Foundation, Iwaki, Japan
- Medical Governance Research Institute, Tokyo, Japan
| | - Namrata Roy
- SRM University, SRM Nagar, Kattankulathur, Chengalpattu, Tamil Nadu, 603203, India
| | - Zareena Fathah
- Kings College London, London, UK
- College of Medicine and Health Sciences, United Arab University, Abu Dhabi, United Arab Emirates
| | - Alfonso J Rodriguez-Morales
- Grupo de Investigación Biomedicina, Faculty of Medicine, Fundacion Universitaria Autonoma de Las Americas, Pereira, Risaralda, Colombia.
- Institución Universitaria Visión de Las Americas, Pereira, Risaralda, Colombia.
- Faculty of Health Sciences, Universidad Cientifica del Sur, Lima, Peru.
- School of Medicine, Universidad Privada Franz Tamayo (UNIFRANZ), Cochabamba, Bolivia.
| |
Collapse
|
50
|
Islamuddin M, Mustfa SA, Ullah SNMN, Omer U, Kato K, Parveen S. Innate Immune Response and Inflammasome Activation During SARS-CoV-2 Infection. Inflammation 2022; 45:1849-1863. [PMID: 35953688 PMCID: PMC9371632 DOI: 10.1007/s10753-022-01651-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 02/12/2022] [Accepted: 02/15/2022] [Indexed: 11/05/2022]
Abstract
The novel coronavirus SARS-CoV-2, responsible for the COVID-19 outbreak, has become a pandemic threatening millions of lives worldwide. Recently, several vaccine candidates and drugs have shown promising effects in preventing or treating COVID-19, but due to the development of mutant strains through rapid viral evolution, urgent investigations are warranted in order to develop preventive measures and further improve current vaccine candidates. Positive-sense-single-stranded RNA viruses comprise many (re)emerging human pathogens that pose a public health problem. Our innate immune system and, in particular, the interferon response form an important first line of defense against these viruses. Flexibility in the genome aids the virus to develop multiple strategies to evade the innate immune response and efficiently promotes their replication and infective capacity. This review will focus on the innate immune response to SARS-CoV-2 infection and the virus' evasion of the innate immune system by escaping recognition or inhibiting the production of an antiviral state. Since interferons have been implicated in inflammatory diseases and immunopathology along with their protective role in infection, antagonizing the immune response may have an ambiguous effect on the clinical outcome of the viral disease. This pathology is characterized by intense, rapid stimulation of the innate immune response that triggers activation of the Nod-like receptor family, pyrin-domain-containing 3 (NLRP3) inflammasome pathway, and release of its products including the pro-inflammatory cytokines IL-6, IL-18, and IL-1β. This predictive view may aid in designing an immune intervention or preventive vaccine for COVID-19 in the near future.
Collapse
Affiliation(s)
- Mohammad Islamuddin
- Molecular Virology Laboratory, Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India.
- Laboratory of Sustainable Animal Environment, Graduate School of Agricultural Science, Tohoku University, Miyagi, Japan.
| | - Salman Ahmad Mustfa
- Centre for Craniofacial and Regenerative Biology, King's College London, Strand, London, UK
| | | | - Usmaan Omer
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Kentaro Kato
- Laboratory of Sustainable Animal Environment, Graduate School of Agricultural Science, Tohoku University, Miyagi, Japan
| | - Shama Parveen
- Molecular Virology Laboratory, Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India
| |
Collapse
|